1
|
Zhou Z, Qin P, Cheng X, Shao M, Ren Z, Zhao Y, Li Q, Liu L. ChatGPT in Oncology Diagnosis and Treatment: Applications, Legal and Ethical Challenges. Curr Oncol Rep 2025; 27:336-354. [PMID: 39998782 DOI: 10.1007/s11912-025-01649-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE OF REVIEW This study aims to systematically review the trajectory of artificial intelligence (AI) development in the medical field, with a particular emphasis on ChatGPT, a cutting-edge tool that is transforming oncology's diagnosis and treatment practices. RECENT FINDINGS Recent advancements have demonstrated that ChatGPT can be effectively utilized in various areas, including collecting medical histories, conducting radiological & pathological diagnoses, generating electronic medical record (EMR), providing nutritional support, participating in Multidisciplinary Team (MDT) and formulating personalized, multidisciplinary treatment plans. However, some significant challenges related to data privacy and legal issues that need to be addressed for the safe and effective integration of ChatGPT into clinical practice. ChatGPT, an emerging AI technology, opens up new avenues and viewpoints for oncology diagnosis and treatment. If current technological and legal challenges can be overcome, ChatGPT is expected to play a more significant role in oncology diagnosis and treatment in the future, providing better treatment options and improving the quality of medical services.
Collapse
Affiliation(s)
- Zihan Zhou
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Peng Qin
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Xi Cheng
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Maoxuan Shao
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Zhaozheng Ren
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Yiting Zhao
- Stomatological College of Nanjing Medical University, Nanjing, 211166, China
| | - Qiunuo Li
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, China
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
2
|
Sharma R, Kashyap M, Zayed H, Krishnia L, Kashyap MK. Artificial blood-hope and the challenges to combat tumor hypoxia for anti-cancer therapy. Med Biol Eng Comput 2025; 63:933-957. [PMID: 39614063 DOI: 10.1007/s11517-024-03233-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/22/2024] [Indexed: 12/01/2024]
Abstract
The blood plays a vital role in the human body and serves as an intermediary between various physiological systems and organs. White blood cells, which are a part of the immune system, defend against infections and regulate the body temperature and pH balance. Blood platelets play a crucial role in clotting, the prevention of excessive bleeding, and the promotion of healing. Blood also serves as a courier system that transports hormones to facilitate communication and synchronization between different organs and systems in the body. The circulatory system, comprised of arteries, veins, and capillaries, plays a crucial role in the efficient transportation and connection of vital nutrients and oxygen. Despite the importance of natural blood, there are often supply shortages, compatibility issues, and medical conditions, which make alternatives such as artificial blood necessary. This is particularly relevant in cancer treatment, which was the focus of our study. In this study, we investigated the potential of artificial blood in cancer therapy, specifically to address tumor hypoxia. We also examined the potential of red blood cell substitutes such as hemoglobin-based oxygen carriers and perfluorocarbons. Additionally, we examined the production of hemoglobin using E. coli and the role of hemoglobin in oncogenesis. Furthermore, we explored the potential use of artificial platelets for cancer treatment. Our study emphasizes the significance of artificial blood in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Rishabh Sharma
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar (Gurugram), Haryana, 122413, India
| | - Manju Kashyap
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar (Gurugram), Haryana, 122413, India
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Lucky Krishnia
- Amity Institute of Nanotechnology, Amity School of Applied Sciences, Amity University Haryana, Panchgaon, Manesar (Gurugram), Haryana, 122413, India.
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar (Gurugram), Haryana, 122413, India.
| |
Collapse
|
3
|
Yoo SK, Fitzgerald CW, Cho BA, Fitzgerald BG, Han C, Koh ES, Pandey A, Sfreddo H, Crowley F, Korostin MR, Debnath N, Leyfman Y, Valero C, Lee M, Vos JL, Lee AS, Zhao K, Lam S, Olumuyide E, Kuo F, Wilson EA, Hamon P, Hennequin C, Saffern M, Vuong L, Hakimi AA, Brown B, Merad M, Gnjatic S, Bhardwaj N, Galsky MD, Schadt EE, Samstein RM, Marron TU, Gönen M, Morris LGT, Chowell D. Prediction of checkpoint inhibitor immunotherapy efficacy for cancer using routine blood tests and clinical data. Nat Med 2025; 31:869-880. [PMID: 39762425 PMCID: PMC11922749 DOI: 10.1038/s41591-024-03398-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 11/01/2024] [Indexed: 01/25/2025]
Abstract
Predicting whether a patient with cancer will benefit from immune checkpoint inhibitors (ICIs) without resorting to advanced genomic or immunologic assays is an important clinical need. To address this, we developed and evaluated SCORPIO, a machine learning system that utilizes routine blood tests (complete blood count and comprehensive metabolic profile) alongside clinical characteristics from 9,745 ICI-treated patients across 21 cancer types. SCORPIO was trained on data from 1,628 patients across 17 cancer types from Memorial Sloan Kettering Cancer Center. In two internal test sets comprising 2,511 patients across 19 cancer types, SCORPIO achieved median time-dependent area under the receiver operating characteristic curve (AUC(t)) values of 0.763 and 0.759 for predicting overall survival at 6, 12, 18, 24 and 30 months, outperforming tumor mutational burden (TMB), which showed median AUC(t) values of 0.503 and 0.543. Additionally, SCORPIO demonstrated superior predictive performance for predicting clinical benefit (tumor response or prolonged stability), with AUC values of 0.714 and 0.641, compared to TMB (AUC = 0.546 and 0.573). External validation was performed using 10 global phase 3 trials (4,447 patients across 6 cancer types) and a real-world cohort from the Mount Sinai Health System (1,159 patients across 18 cancer types). In these external cohorts, SCORPIO maintained robust performance in predicting ICI outcomes, surpassing programmed death-ligand 1 immunostaining. These findings underscore SCORPIO's reliability and adaptability, highlighting its potential to predict patient outcomes with ICI therapy across diverse cancer types and healthcare settings.
Collapse
Affiliation(s)
- Seong-Keun Yoo
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Conall W Fitzgerald
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Byuri Angela Cho
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bailey G Fitzgerald
- Department of Medicine, Thoracic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Catherine Han
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth S Koh
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abhinav Pandey
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hannah Sfreddo
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fionnuala Crowley
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Neha Debnath
- Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Yan Leyfman
- Internal Medicine, Icahn School of Medicine at Mount Sinai South Nassau, Rockville Centre, NY, USA
| | - Cristina Valero
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Lee
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joris L Vos
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Sangho Lee
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karena Zhao
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stanley Lam
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ezekiel Olumuyide
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fengshen Kuo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric A Wilson
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pauline Hamon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clotilde Hennequin
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Saffern
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lynda Vuong
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian Brown
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Robert M Samstein
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas U Marron
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Early Phase Trials Unit, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc G T Morris
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Laboratory of Experimental Cancer Immunogenomics, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Diego Chowell
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Diao H, Fan Y, Kang D, Chen Z, Lu Y, Huang X, Xia X, Chen W. Clinicopathological Characteristics and Outcomes of Lupus Nephritis Patients With Thrombocytopenia: A Single-Center Retrospective Study. Immun Inflamm Dis 2025; 13:e70179. [PMID: 40105598 PMCID: PMC11921463 DOI: 10.1002/iid3.70179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVES The objective of this study is to analyze and summarize the clinical characteristics and prognosis of lupus nephritis (LN) patients with thrombocytopenia and to improve the cognition of the disease. METHODS 896 LN patients were enrolled in this study and their clinical and pathological data were collected and analyzed. The primary end point was mortality. The secondary end point was adverse renal outcomes, defined as doubling of the baseline serum creatinine or end-stage renal diseases. Cox regression model was used to analyze the risk factors of mortality or renal events in LN with and without thrombocytopenia. RESULTS Among 896 LN patients, 70 (7.8%) were diagnosed with thrombocytopenia. LN patients with thrombocytopenia had lower estimated glomerular filtration rate (eGFR) and higher systemic lupus erythematosus disease activity index (SLE-DAI), proportion of anemia, leukopenia, hypocomplementemia, and positive anti-cardiolipin antibodies, compared to those without thrombocytopenia. LN patients with thrombocytopenia had higher scores of activity index and more activity features (endocapillary hypercellularity, medullary loop necrosis) on kidney biopsy. There was no significant difference in patient survival and renal survival between LN patients with and without thrombocytopenia. Anemia was a risk factor for death in LN patients with thrombocytopenia and lower eGFR was a risk factor for adverse renal outcomes. CONCLUSIONS LN patients with thrombocytopenia showed higher disease activity, more anti-cardiolipin antibody positivity and a higher activity index in kidney biopsy, but the prognosis was similar compared with those without thrombocytopenia. Anemia was a risk factor for death in LN patients with thrombocytopenia.
Collapse
Affiliation(s)
- Hui Diao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yuting Fan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Di Kang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Zhiqing Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yuewen Lu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xiamin Huang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xi Xia
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| |
Collapse
|
5
|
Sohn SH, Sul H, Kim B, Zang D. Effects of Darbepoetin Alfa and Ferric Derisomaltose Plus Darbepoetin Alfa in Functional Iron-Deficiency Anemia. Int J Mol Sci 2025; 26:2203. [PMID: 40076818 PMCID: PMC11899774 DOI: 10.3390/ijms26052203] [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: 02/07/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Functional iron-deficiency anemia (FIDA) is a side effect of many cancer treatments, occurring when chemotherapy drugs damage bone marrow cells, which are responsible for producing red blood cells, due to the myelosuppressive effects of chemotherapy, or to the cancer itself. This study was performed to compare the effects of darbepoetin alfa alone, or in combination with ferric derisomaltose in cancer patients with FIDA, and to elucidate the mechanism underlying the effects in F36E cells. F36E cells treated with darbepoetin alfa showed increased cell viability. AML and GC cells treated with darbepoetin alfa, ferric derisomaltose, or ferric derisomaltose plus darbepoetin alfa showed no induction of apoptosis. The effects of these drugs on the anticancer efficacy of PTX chemotherapy were examined by analyzing cell viability and induction of apoptosis. Darbepoetin alfa, ferric derisomaltose, and ferric derisomaltose plus darbepoetin alfa showed no significant inhibitory effects on the apoptosis-inducing activity of PTX in GC cell lines. Patients with chemotherapy-induced FIDA in Group I receiving ferric derisomaltose plus darbepoetin alfa showed higher hemoglobin levels, transferrin saturation, and ferritin levels compared to those in Group II, treated with darbepoetin alfa alone. In cancer patients with FIDA, the prognosis of anemia treatment was better in the ferric derisomaltose plus darbepoetin alfa combination group than in the group receiving darbepoetin alfa monotherapy.
Collapse
Affiliation(s)
- Sung-Hwa Sohn
- Hallym Translational Research Institute, College of Medicine, Hallym University, Anyang-si 14068, Gyeonggi-do, Republic of Korea; (S.-H.S.); (H.S.)
| | - Heejung Sul
- Hallym Translational Research Institute, College of Medicine, Hallym University, Anyang-si 14068, Gyeonggi-do, Republic of Korea; (S.-H.S.); (H.S.)
| | - Bumjun Kim
- Department of Internal Medicine, Hallym University Medical Center, College of Medicine, Hallym University, Anyang-si 14068, Gyeonggi-do, Republic of Korea;
| | - Daeyoung Zang
- Department of Internal Medicine, Hallym University Medical Center, College of Medicine, Hallym University, Anyang-si 14068, Gyeonggi-do, Republic of Korea;
| |
Collapse
|
6
|
Lu S, Wu J, Jiang J, Guo Q, Yu Y, Liu Y, Zhang H, Qian L, Dai X, Xie Y, Fu T, Lee T, Lu Y, Ma R, Eisner MD. Efficacy and Safety of Roxadustat for Anemia in Patients Receiving Chemotherapy for Nonmyeloid Malignancies: A Randomized, Open-Label, Active-Controlled Phase III Study. J Clin Oncol 2025; 43:143-153. [PMID: 39353163 PMCID: PMC11708981 DOI: 10.1200/jco.23.02742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/27/2024] [Accepted: 08/20/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE We evaluated the efficacy and safety of roxadustat, a first-in-class hypoxia-inducible factor prolyl hydroxylase inhibitor, for chemotherapy-induced anemia (CIA) in patients with nonmyeloid malignancies receiving multicycle treatments of chemotherapy. PATIENTS AND METHODS In this open-label, noninferiority phase III study conducted at 44 sites in China, 159 participants age ≥18 years with CIA nonmyeloid malignancy and CIA were randomly assigned (1:1) to oral roxadustat or subcutaneous recombinant human erythropoietin-α (rHuEPO-α) three times a week for 12 weeks. Roxadustat starting dosages were 100, 120, and 150 mg three times a week for participants weighing 40-<50, 50-60, and >60 kg, respectively. rHuEPO-α starting dosage for all participants was 150 IU/kg three times a week. Both roxadustat and rHuEPO-α dosages could be modified. The primary end point was least-squares mean (LSM) change in hemoglobin (Hb) concentration from baseline to the concentration averaged over weeks 9-13. RESULTS Of the 159 participants randomly assigned, 140 were included in the per-protocol set (roxadustat, n = 78; rHuEPO-α, n = 62). The LSM (95% two-sided CI) change from baseline to weeks 9-13 in Hb concentration was 17.1 (13.58 to 20.71) g/L with roxadustat and 15.4 (11.34 to 19.50) g/L with rHuEPO-α (mean difference [95% CI], 1.7 [-3.39 to 6.84]). The lower bound of the one-sided 97.5% CI for the treatment difference (‒3.4 g/L) was greater than the predefined noninferiority margin of ‒6.6 g/L, establishing noninferiority. Noninferiority was supported by five of six key secondary end points. Rates of adverse events were generally comparable between treatments and consistent with previous findings. CONCLUSION Roxadustat was noninferior to rHuEPO-α in treating CIA in participants with nonmyeloid malignancies receiving multicycle treatments of myelosuppressive chemotherapy. The oral formulation of roxadustat may potentially increase compliance.
Collapse
Affiliation(s)
- Shun Lu
- Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiong Wu
- Cancer Hospital, Fudan University, Shanghai, China
| | - Jin Jiang
- The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qisen Guo
- Shandong First Medical University Cancer Hospital, Jinan, China
| | - Yan Yu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Liu
- Neijiang Second People's Hospital, Neijiang, China
| | - Hua Zhang
- The First People's Hospital of Foshan, Foshan, China
| | - Ling Qian
- Shanghai Fifth People's Hospital, Shanghai, China
| | | | - Yanyan Xie
- The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ting Fu
- Zhuzhou Central Hospital, Zhuzhou, China
| | | | - Yan Lu
- FibroGen (China) Medical Technology Development Co, Ltd, Shanghai, China
| | - Rui Ma
- FibroGen (China) Medical Technology Development Co, Ltd, Shanghai, China
| | | |
Collapse
|
7
|
Schmitz KH, Brown JC, Irwin ML, Robien K, Scott JM, Berger NA, Caan B, Cercek A, Crane TE, Evans SR, Ligibel JA, Meyerhardt JA, Agurs-Collins T, Basen-Engquist K, Bea JW, Cai SF, Cartmel B, Chinchilli VM, Demark-Wahnefried W, Dieli-Conwright CM, DiPietro L, Doerksen SE, Edelstein SL, Elena J, Evans W, Ferrucci LM, Foldi J, Freylersythe S, Furberg H, Jones LW, Levine R, Moskowitz CS, Owusu C, Penedo F, Rabin BA, Ratner E, Rosenzweig M, Salz T, Sanft T, Schlumbrecht M, Spielmann G, Thomson CA, Tjaden AH, Weiser MR, Yang S, Yu AF, Perna FM. Exercise and Nutrition to Improve Cancer Treatment-Related Outcomes (ENICTO). J Natl Cancer Inst 2025; 117:9-19. [PMID: 39118255 PMCID: PMC11717426 DOI: 10.1093/jnci/djae177] [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: 03/29/2024] [Revised: 06/26/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Chemotherapy treatment-related side effects are common and increase the risk of suboptimal outcomes. Exercise interventions during cancer treatment improve self-reported physical functioning, fatigue, anxiety, and depression, but it is unclear whether these interventions improve important clinical outcomes, such as chemotherapy relative dose intensity. The National Cancer Institute funded the Exercise and Nutrition to Improve Cancer Treatment-Related Outcomes (ENICTO) Consortium to address this knowledge gap. This article describes the mechanisms hypothesized to underpin intervention effects on clinically relevant treatment outcomes, briefly outlines each project's distinct research aims, summarizes the scope and organizational structure of ENICTO, and provides an overview of the integrated common data elements used to pursue research questions collectively. In addition, the article includes a description of consortium-wide activities and broader research community opportunities for collaborative research. Findings from the ENICTO Consortium have the potential to accelerate a paradigm shift in oncology care such that patients with cancer could receive exercise and nutrition programming as the standard of care in tandem with chemotherapy to improve relative dose intensity for a curative outcome.
Collapse
Affiliation(s)
- Kathryn H Schmitz
- Hematology and Oncology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Justin C Brown
- Department of Cancer Energetics, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Melinda L Irwin
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Kim Robien
- Department of Exercise and Nutrition Sciences and Department of Epidemiology, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Jessica M Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Nathan A Berger
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Bette Caan
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA, USA
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tracy E Crane
- Department of Medicine, Division of Medical Oncology, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Scott R Evans
- Biostatistics Center and Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Jennifer A Ligibel
- Division of Breast Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Karen Basen-Engquist
- Department of Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer W Bea
- Health Promotion Sciences, University of Arizona and University of Arizona Cancer Center, Tucson, AZ, USA
| | - Sheng F Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brenda Cartmel
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Vernon M Chinchilli
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | | | | | - Loretta DiPietro
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Shawna E Doerksen
- Hematology and Oncology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sharon L Edelstein
- Biostatistics Center, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Joanne Elena
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - William Evans
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Leah M Ferrucci
- Department of Chronic Disease Epidemiology, Yale School of Public Health and Yale Cancer Center, New Haven, CT, USA
| | - Julia Foldi
- Hematology and Oncology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sarah Freylersythe
- Department of Medicine, Division of Medical Oncology, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Helena Furberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lee W Jones
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Ross Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chaya S Moskowitz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cynthia Owusu
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Frank Penedo
- Departments of Psychology and Medicine and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Borsika A Rabin
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, USA
| | - Elena Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University, New Haven, CT, USA
| | | | - Talya Salz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tara Sanft
- Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Matthew Schlumbrecht
- Division of Gynecologic Oncology, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | | | - Cynthia A Thomson
- Department of Health Promotion Sciences, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Ashley H Tjaden
- Biostatistics Center, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Martin R Weiser
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Anthony F Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Frank M Perna
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
8
|
Akagi T, Hamano H, Miyamoto H, Takeda T, Zamami Y, Ohyama K. Evaluating the impact of loperamide on irinotecan-induced adverse events: a disproportionality analysis of data from the World Health Organization pharmacovigilance database (VigiBase). Eur J Clin Pharmacol 2025; 81:129-137. [PMID: 39443366 DOI: 10.1007/s00228-024-03767-6] [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: 08/01/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE SN-38, the active metabolite of irinotecan, may cause adverse events necessitating treatment discontinuation and management. Diarrhea, which is treated with loperamide, is one such event. However, loperamide may delay SN-38 elimination, causing more adverse events. Therefore, understanding the adverse events caused by symptomatic drugs is crucial for safe drug therapy. This study aimed to assess the association between loperamide and irinotecan-induced adverse events. METHODS We analyzed data up to December 2022 from VigiBase, the World Health Organization's adverse event database. The study used reporting odds ratios (RORs) to evaluate the associations between concomitant medications and irinotecan-induced adverse events. Fisher's exact probability test was used to analyze the adverse events. Logistic regression analysis was performed to identify associated adverse event signals. RESULTS Of the 32,520,983 cases analyzed, 57,454 involved the use of irinotecan. Among these, 1589 (2.8%) patients were co-treated with loperamide. Signals for neutropenia (ROR 1.37, 95% confidence interval (CI) 1.20-1.57, p < 0.001), anemia (ROR 1.81, 95% CI 1.43-2.30, p < 0.001), and alopecia (ROR 1.89, 95% CI 1.30-2.74, p < 0.01) were detected with concomitant loperamide. Multivariate logistic regression analysis confirmed that concomitant loperamide use was associated with signals for neutropenia, anemia, and alopecia. CONCLUSION Our results suggest that loperamide increases the risk of irinotecan-induced adverse events and enhances irinotecan toxicity. The study methodology may be useful for predicting adverse event risk when choosing symptomatic therapy drugs during irinotecan use.
Collapse
Affiliation(s)
- Tomoaki Akagi
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan
| | - Hirofumi Hamano
- Department of Hospital Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Hirotaka Miyamoto
- Department of Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tatsuaki Takeda
- Department of Education and Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoshito Zamami
- Department of Hospital Pharmacy, Okayama University Hospital, Okayama, Japan.
| | - Kaname Ohyama
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Japan.
| |
Collapse
|
9
|
Corner TP, Salah E, Tumber A, Kaur S, Nakashima Y, Allen MD, Schnaubelt LI, Fiorini G, Brewitz L, Schofield CJ. Crystallographic and Selectivity Studies on the Approved HIF Prolyl Hydroxylase Inhibitors Desidustat and Enarodustat. ChemMedChem 2024; 19:e202400504. [PMID: 39291299 DOI: 10.1002/cmdc.202400504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/19/2024]
Abstract
Prolyl hydroxylase domain-containing proteins 1-3 (PHD1-3) are 2-oxoglutarate (2OG)-dependent oxygenases catalysing C-4 hydroxylation of prolyl residues in α-subunits of the heterodimeric transcription factor hypoxia-inducible factor (HIF), modifications that promote HIF-α degradation via the ubiquitin-proteasome pathway. Pharmacological inhibition of the PHDs induces HIF-α stabilisation, so promoting HIF target gene transcription. PHD inhibitors are used to treat anaemia caused by chronic kidney disease (CKD) due to their ability to stimulate erythropoietin (EPO) production. We report studies on the effects of the approved PHD inhibitors Desidustat and Enarodustat, and the clinical candidate TP0463518, on activities of a representative set of isolated recombinant human 2OG oxygenases. The three molecules manifest selectivity for PHD inhibition over that of the other 2OG oxygenases evaluated. We obtained crystal structures of Desidustat and Enarodustat in complex with the human 2OG oxygenase factor inhibiting hypoxia-inducible factor-α (FIH), which, together with modelling studies, inform on the binding modes of Desidustat and Enarodustat to active site Fe(II) in 2OG oxygenases, including PHD1-3. The results will help in the design of selective inhibitors of both the PHDs and other 2OG oxygenases, which are of medicinal interest due to their involvement inter alia in metabolic regulation, epigenetic signalling, DNA-damage repair, and agrochemical resistance.
Collapse
Affiliation(s)
- Thomas P Corner
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
- Present Address: Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, 06511, United States of America
| | - Eidarus Salah
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Anthony Tumber
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Samanpreet Kaur
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Yu Nakashima
- Institute of Natural Medicine, University of Toyama, 2630-Sugitani, Toyama, 930-0194, Japan
| | - Mark D Allen
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Lara I Schnaubelt
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Giorgia Fiorini
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Lennart Brewitz
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| |
Collapse
|
10
|
Tęcza K, Kalinowska-Herok M, Rusinek D, Zajkowicz A, Pfeifer A, Oczko-Wojciechowska M, Pamuła-Piłat J. Are the Common Genetic 3'UTR Variants in ADME Genes Playing a Role in Tolerance of Breast Cancer Chemotherapy? Int J Mol Sci 2024; 25:12283. [PMID: 39596349 PMCID: PMC11594993 DOI: 10.3390/ijms252212283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
We studied the associations between 3'UTR genetic variants in ADME genes, clinical factors, and the risk of breast cancer chemotherapy toxicity. Those variants and factors were tested in relation to seven symptoms belonging to myelotoxicity (anemia, leukopenia, neutropenia), gastrointestinal side effects (vomiting, nausea), nephrotoxicity, and hepatotoxicity, occurring in overall, early, or recurrent settings. The cumulative risk of overall symptoms of anemia was connected with AKR1C3 rs3209896 AG, ERCC1 rs3212986 GT, and >6 cycles of chemotherapy; leukopenia was determined by ABCC1 rs129081 allele G and DPYD rs291593 allele T; neutropenia risk was correlated with accumulation of genetic variants of DPYD rs291583 allele G, ABCB1 rs17064 AT, and positive HER2 status. Risk of nephrotoxicity was determined by homozygote DPYD rs291593, homozygote AKR1C3 rs3209896, postmenopausal age, and negative ER status. Increased risk of hepatotoxicity was connected with NR1/2 rs3732359 allele G, postmenopausal age, and with present metastases. The risk of nausea and vomiting was linked to several genetic factors and premenopausal age. We concluded that chemotherapy tolerance emerges from the simultaneous interaction of many genetic and clinical factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jolanta Pamuła-Piłat
- Department of Clinical and Molecular Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (K.T.); (M.K.-H.); (D.R.); (A.Z.); (A.P.); (M.O.-W.)
| |
Collapse
|
11
|
Isingizwe ZR, Meelheim BA, Benbrook DM. Elevated Platelet Aggregation in Patients with Ovarian Cancer: More than Just Increased Platelet Count. Cancers (Basel) 2024; 16:3583. [PMID: 39518024 PMCID: PMC11545395 DOI: 10.3390/cancers16213583] [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: 09/30/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Patients with ovarian cancer have high platelet counts, which correlate with disease burden, incidence, and lethality of blood clots (thrombosis). We hypothesized that elevated aggregation is associated with both increased platelet number and altered behavior of platelets in patients with ovarian cancer. Methods: Healthy controls and patients with suspected or diagnosed ovarian cancer were evaluated for complete blood counts. To evaluate the effects of platelet count versus platelet behavior, equal platelet-rich plasma (PRP) volumes versus equal platelet numbers were used in platelet aggregation assays. Arachidonic acid, adenosine diphosphate, and collagen platelet agonists were used to induce aggregation. Volunteers were grouped into healthy controls (23), benign/borderline cases (7), and cancer cases (25 ovarian, 1 colorectal, and 2 endometrial). Results: The rate and amount of platelet aggregation were higher in patients compared to healthy controls regardless of whether the same platelet number or PRP volume was used. Compared to healthy controls, patients with untreated ovarian cancer exhibited high levels of platelet activation markers, P-selectin (27.06 vs. 31.06 ng/mL, p = 0.03), and beta-thromboglobulin (3.073 vs. 4.091 µg/mL, p = 0.02) in their plasma. The significance of the elevation and its correlations with platelet number or PRP volume varied depending on the agonist. Platelet (305.88 vs. 134.12, p < 0.0001) and white blood cell (8.459 vs. 5.395, p < 0.01) counts (×109/L) were elevated pre-chemotherapy and decreased post-chemotherapy, respectively. Conclusions: Elevated platelet aggregation is caused by both altered platelet number and behavior in patients with ovarian cancer. These results support the study of antiplatelet agents for thrombosis prevention in these patients.
Collapse
Affiliation(s)
- Zitha Redempta Isingizwe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA;
| | - Brooke A. Meelheim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Doris Mangiaracina Benbrook
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA;
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| |
Collapse
|
12
|
Phulari RGS, Solanki B. Immune checkpoint inhibitors: Utilizing patient's own immunity to treat oral cancer. J Oral Maxillofac Pathol 2024; 28:641-650. [PMID: 39949682 PMCID: PMC11819625 DOI: 10.4103/jomfp.jomfp_327_22] [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: 08/02/2022] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 02/16/2025] Open
Abstract
Head and Neck squamous cell carcinoma is an immunosuppressive state. HNSCC evades immune responses through multiple resistance mechanisms. Because of better understanding of interaction between tumour microenvironment and immune regulators, there is increasing interest in role of immunotherapy as a treatment modality of HNSCC. Many clinical trials have been performed using checkpoint inhibitors, as monotherapies and combination therapies. Immune checkpoint molecule, programmed cell death 1 (PD-1) has shown promising results as a treatment of Recurrent and Metastatic HNSCC. This review discusses immune checkpoint molecules, their functional mechanisms, role of immunotherapy as a monotherapies and combination therapy for better treatment and prognosis of HNSCC patients.
Collapse
Affiliation(s)
- Rashmi GS Phulari
- Department of Oral and Maxillofacial Pathology and Microbiology, Manubhai Patel Dental College and Hospital and Oral Research Institute, Vishwajyoti Ashram, Near Vidyakunj School, Maunjmauda, Vadodara, Gujarat, India
| | - Bharvi Solanki
- Department of Oral and Maxillofacial Pathology and Microbiology, Manubhai Patel Dental College and Hospital and Oral Research Institute, Vishwajyoti Ashram, Near Vidyakunj School, Maunjmauda, Vadodara, Gujarat, India
| |
Collapse
|
13
|
Zhao X, Zhao Z, Li B, Huan S, Li Z, Xie J, Liu G. ACSL4-mediated lipid rafts prevent membrane rupture and inhibit immunogenic cell death in melanoma. Cell Death Dis 2024; 15:695. [PMID: 39343834 PMCID: PMC11439949 DOI: 10.1038/s41419-024-07098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024]
Abstract
Chemotherapy including platinum-based drugs are a possible strategy to enhance the immune response in advanced melanoma patients who are resistant to immune checkpoint blockade (ICB) therapy. However, the immune-boosting effects of these drugs are a subject of controversy, and their impact on the tumor microenvironment are poorly understood. In this study, we discovered that lipid peroxidation (LPO) promotes the formation of lipid rafts in the membrane, which mediated by Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4) impairs the sensitivity of melanoma cells to platinum-based drugs. This reduction primarily occurs through the inhibition of immunogenic ferroptosis and pyroptosis by reducing cell membrane pore formation. By disrupting ACSL4-mediaged lipid rafts via the removal of membrane cholesterol, we promoted immunogenic cell death, transformed the immunosuppressive environment, and improved the antitumor effectiveness of platinum-based drugs and immune response. This disruption also helped reverse the decrease in CD8+ T cells while maintaining their ability to secrete cytokines. Our results reveal that ACSL4-dependent LPO is a key regulator of lipid rafts formation and antitumor immunity, and that disrupting lipid rafts has the potential to enhance platinum-based drug-induced immunogenic ferroptosis and pyroptosis in melanoma. This novel strategy may augment the antitumor immunity of platinum-based therapy and further complement ICB therapy.
Collapse
Affiliation(s)
- Xi Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zenglu Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Bingru Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Shuyu Huan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zixi Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianlan Xie
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guoquan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Department of Biomedical Engineering, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| |
Collapse
|
14
|
Smit ER, Muñoz Sandoval D, Kreft IC, van der Meer PF, van der Zwaan C, Voorberg J, Ypma PF, Hoogendijk AJ, Kerkhoffs JL, van den Biggelaar M. Plasma proteomes of acute myeloid leukemia patients treated with transfusions reveal signatures of inflammation and hemostatic dysregulation. TRANSLATIONAL MEDICINE COMMUNICATIONS 2024; 9:27. [PMID: 40078206 PMCID: PMC11893646 DOI: 10.1186/s41231-024-00189-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/30/2024] [Indexed: 03/14/2025]
Abstract
Background Bone marrow aplasia is a common feature in acute myeloid leukemia (AML) patients during their remission induction treatment, and is associated with potential complications such as bleeding, infection and anemia. Frequent platelet and red cell transfusions are administered to prevent and treat these complications. However, platelet counts are poorly associated with bleeding events in this population. Therefore, plasma protein levels could add valuable insights to improve our understanding of the patient's health state. In this study, we aimed to delineate the plasma proteome, including inflammatory pathways, hemostatic and immune components, of AML patients during treatment with intensive transfusion support. Methods We employed unbiased mass spectrometry (MS)-based proteomics on longitudinal plasma samples from 10 AML patients during intensive-transfusion treatment phase with healthy individuals as baseline control. Results A total of 450 proteins were quantified in plasma samples from AML patients and healthy controls. Alteration in proteins levels were mainly observed for proteins involved in inflammation (e.g. SAA1 and CRP), and complement (e.g. C9 and MASP2) when comparing AML versus healthy individuals. Correlation analysis revealed additional affected protein dynamics, including proteins associated with coagulation cascade, endopeptidase inhibitors activity and lipoprotein remodeling. Conclusion The plasma proteome from AML patients during intensive treatment shows a disbalance in inflammation, endopeptidase inhibitors activity, lipoprotein remodeling, coagulation and complement. These effects and potential associations with bleeding risk will be further studied in a bigger cohort. Supplementary Information The online version contains supplementary material available at 10.1186/s41231-024-00189-5.
Collapse
Affiliation(s)
- Eva R. Smit
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Diana Muñoz Sandoval
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Iris C. Kreft
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Pieter F. van der Meer
- Department of Hematology, Haga Teaching Hospital, the Hague, the Netherlands
- Department of Product and Process Development, Sanquin Blood Bank, Amsterdam, the Netherlands
| | - Carmen van der Zwaan
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Jan Voorberg
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Paula F. Ypma
- Department of Hematology, Haga Teaching Hospital, the Hague, the Netherlands
| | - Arie J. Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Jean-Louis Kerkhoffs
- Department of Hematology, Haga Teaching Hospital, the Hague, the Netherlands
- Unit Transfusion Medicine, Sanquin Blood Bank, Amsterdam, the Netherlands
| | | |
Collapse
|
15
|
Yap AJY, Teo DCH, Ang PS, Yap ES, Tan SH, Loke CWP, Dorajoo SR. Validation of a Major and Clinically Relevant Nonmajor Bleeding Phenotyping Algorithm on Electronic Health Records. Pharmacoepidemiol Drug Saf 2024; 33:e5875. [PMID: 39090800 DOI: 10.1002/pds.5875] [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: 11/27/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE Bleeding is an important health outcome of interest in epidemiological studies. We aimed to develop and validate rule-based algorithms to identify (1) major bleeding and (2) all clinically relevant bleeding (CRB) (composite of major and all clinically relevant nonmajor bleeding) within real-world electronic healthcare data. METHODS We took a random sample (n = 1630) of inpatient admissions to Singapore public healthcare institutions in 2019 and 2020, stratifying by hospital and year. We included patients of all age groups, sex, and ethnicities. Presence of major bleeding and CRB were ascertained by two annotators through chart review. A total of 630 and 1000 records were used for algorithm development and validation, respectively. We formulated two algorithms: sensitivity- and positive predictive value (PPV)-optimized algorithms. A combination of hemoglobin test patterns and diagnosis codes were used in the final algorithms. RESULTS During validation, diagnosis codes alone yielded low sensitivities for major bleeding (0.16) and CRB (0.24), although specificities and PPV were high (>0.97). For major bleeding, the sensitivity-optimized algorithm had much higher sensitivity and negative predictive values (NPVs) (sensitivity = 0.94, NPV = 1.00), however false positive rates were also relatively high (specificity = 0.90, PPV = 0.34). PPV-optimized algorithm had improved specificity and PPV (specificity = 0.96, PPV = 0.52), with little reduction in sensitivity and NPV (sensitivity = 0.88, NPV = 0.99). For CRB events, our algorithms had lower sensitivities (0.50-0.56). CONCLUSIONS The use of diagnosis codes alone misses many genuine major bleeding events. We have developed major bleeding algorithms with high sensitivities, which can ascertain events within populations of interest.
Collapse
Affiliation(s)
- Aaron Jun Yi Yap
- Vigilance & Compliance Branch, Health Products Regulation Group, Health Sciences Authority, Singapore
| | - Desmond Chun Hwee Teo
- Vigilance & Compliance Branch, Health Products Regulation Group, Health Sciences Authority, Singapore
| | - Pei San Ang
- Vigilance & Compliance Branch, Health Products Regulation Group, Health Sciences Authority, Singapore
| | - Eng Soo Yap
- Department of Laboratory Medicine, National University Hospital, Singapore
| | - Siew Har Tan
- Vigilance & Compliance Branch, Health Products Regulation Group, Health Sciences Authority, Singapore
| | - Celine Wei Ping Loke
- Vigilance & Compliance Branch, Health Products Regulation Group, Health Sciences Authority, Singapore
| | - Sreemanee Raaj Dorajoo
- Vigilance & Compliance Branch, Health Products Regulation Group, Health Sciences Authority, Singapore
| |
Collapse
|
16
|
Wondm SA, Dagnew SB, Gubae K, Tesfaye TC, Tamene FB. Determinants of anemia among patients receiving cancer chemotherapy in Northwest Ethiopia. Front Med (Lausanne) 2024; 11:1415877. [PMID: 39055698 PMCID: PMC11269183 DOI: 10.3389/fmed.2024.1415877] [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: 04/11/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024] Open
Abstract
Background Chemotherapy-induced anemia (CIA) is a hematologic complication that frequently affects patients with cancer undergoing chemotherapy. It is associated with worse treatment outcomes, higher rates of morbidity and mortality, worse quality of life, and higher healthcare costs. The incidence and predictors of CIA in Ethiopia, particularly in Northwest Ethiopian oncology centers, are poorly understood. This study was conducted at Northwest Ethiopian oncology centers to evaluate the incidence and determinants of chemotherapy-induced anemia in adult patients with cancer undergoing chemotherapy. Methods This 3-year hospital-based retrospective follow-up study included adult patients with cancer receiving chemotherapy between 2019 and 2021 at two oncology centers in Northwest Ethiopia. Data were collected from October to December 2021. A binary logistic regression model was used to select variables and determine the Crude Odds Ratio (COR). Variables with P-value < 0.2 were entered into the multivariable logistic regression and Adjusted odds ratio (AOR) with 95% Confidence intervals (CI) for variables with P-value < 0.05 were estimated to show determinants of chemotherapy-induced anemia among cancer patients who received chemotherapy. Results A total of 402 patients were included in the final analysis. The overall incidence of CIA was 75.4% (95% CI 70.7, 79.8). Older age [AOR = 1.8, 95% CI (1.4-3.5); P = 0.043], hematologic cancer [AOR = 3.7, 95% CI (3.2-5.7), P = 0.021], obesity [AOR = 3.4, 95% CI (2.3-6.9); P = 0.028], ≥6 chemotherapy cycles [AOR = 3.8, 95% CI (3.2-5.1), P = 0.019], cancer metastasis to bone [AOR = 2.9, 95% CI (1.2-4.7), P = 0.025] were statistically significant predictors of chemotherapy-induced anemia. Conclusion Chemotherapy-induced anemia persisted in a significant percentage of cancer patients. Chemotherapy-induced anemia developed in three-quarters of patients undergoing chemotherapy. Chemotherapy-induced anemia was significantly associated with older age, hematologic malignancy, obesity, a greater number of chemotherapy cycles, and cancer metastasis to bone. To lower the risk of morbidity related to anemia, patients with chemotherapy-induced anemia should be regularly evaluated and treated with appropriate treatment.
Collapse
Affiliation(s)
- Samuel Agegnew Wondm
- Clinical Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Samuel Berihun Dagnew
- Clinical Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Kale Gubae
- Pharmacology Unit, Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tegenu Chanie Tesfaye
- Clinical Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Fasil Bayafers Tamene
- Clinical Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
17
|
Yoon KW, Park S, Park CM. Prevalence and factors influencing anemia recovery after intensive care. Transfus Apher Sci 2024; 63:103922. [PMID: 38664087 DOI: 10.1016/j.transci.2024.103922] [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: 11/02/2023] [Revised: 03/30/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Anemia is associated with adverse outcomes and prolonged hospitalizations in critically ill patients. Regarding the recent adoption of restrictive transfusion protocols in intensive care unit (ICU) management, anemia remains highly prevalent even after ICU discharge. This study aimed to investigate the prevalence of anemia following ICU discharge and factors affecting recovery from anemia. METHODS In this retrospective cohort study involving 3969 adult ICU survivors, we assessed anemia severity using the National Cancer Institute criteria at six time points: ICU admission, ICU discharge, hospital discharge, and at 3-, 6-, and 12-month post-hospital discharge. In addition, baseline characteristics, including age, sex, comorbidities, and recent iron supplementation or erythropoietin administration, were evaluated. RESULTS Our findings revealed an in-hospital mortality rate of 28.6%. The median hospital and ICU stays were 20 and 5 days, respectively, with common comorbidities including hypertension, and diabetes mellitus (DM). Among the patients, the hemoglobin levels of 3967 patients were confirmed at the time of discharge from the ICU, representing 99.95% of the total. The prevalence of anemia persisted post- ICU discharge; less than 30% of patients recovered, whereas 13.6% of them experienced worsening of anemia post-ICU discharge. Factors contributing to anemia severity were female sex, DM, chronic renal failure, malignant solid tumors, and administration of iron supplements. CONCLUSIONS This study highlighted the need for targeted interventions to manage anemia post-ICU discharge and suggested potential factors that influence recovery from anemia.
Collapse
Affiliation(s)
- Kyoung Won Yoon
- Division of Critical Care, Department of Surgery, Chung‑Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong-si, Gyeonggi-do, South Korea
| | - Sungjoo Park
- Division of Pediatric Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Chi-Min Park
- Department of Surgery and Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
18
|
Bai H, Huang W, Li J, Ji Y, He S, Hu H. Enhancing anticancer treatment: Development of cRGD-Conjugated F-OH-Evo prodrugs for targeted delivery. Bioorg Med Chem 2024; 107:117759. [PMID: 38795572 DOI: 10.1016/j.bmc.2024.117759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/28/2024]
Abstract
Small molecule drugs sourced from natural products are pivotal for novel therapeutic discoveries. However, their clinical deployment is often impeded by non-specific activity and severe adverse effects. This study focused on 3-fluoro-10-hydroxy-Evodiamine (F-OH-Evo), a potent derivative of Evodiamine, whose development is curtailed due to suboptimal tumor selectivity and heightened cytotoxicity. By harnessing the remarkable stability, specificity, and αvβ3 integrin affinity of c(RGDFK), a novel prodrug by conjugating F-OH-Evo with cRGD was synthesized. This innovative prodrug substantially enhanced the tumor-specific targeting of F-OH-Evo and improved the anti-tumor activities. Among them, compound 3c demonstrated the best selective inhibitory activity toward U87 cancer cells in vitro. It selectively enterd U87 cells by binding to αvβ3 integrin, releasing the parent molecule under the dual response of ROS and GSH to exert inhibitory activity on topo I. The results highlight the potential of cRGD-conjugated prodrugs in targeted cancer therapy. This approach signifies a significant advancement in developing safer and more effective chemotherapy drugs, emphasizing the role of prodrug strategies in overcoming the limitations of traditional cancer treatments.
Collapse
Affiliation(s)
- Haohao Bai
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Wenjing Huang
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Jinqiu Li
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Yajing Ji
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Shipeng He
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China.
| | - Honggang Hu
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China.
| |
Collapse
|
19
|
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
|
20
|
Bozzini C, Busti F, Marchi G, Vianello A, Cerchione C, Martinelli G, Girelli D. Anemia in patients receiving anticancer treatments: focus on novel therapeutic approaches. Front Oncol 2024; 14:1380358. [PMID: 38628673 PMCID: PMC11018927 DOI: 10.3389/fonc.2024.1380358] [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: 02/01/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Anemia is common in cancer patients and impacts on quality of life and prognosis. It is typically multifactorial, often involving different pathophysiological mechanisms, making treatment a difficult task. In patients undergoing active anticancer treatments like chemotherapy, decreased red blood cell (RBC) production due to myelosuppression generally predominates, but absolute or functional iron deficiency frequently coexists. Current treatments for chemotherapy-related anemia include blood transfusions, erythropoiesis-stimulating agents, and iron supplementation. Each option has limitations, and there is an urgent need for novel approaches. After decades of relative immobilism, several promising anti-anemic drugs are now entering the clinical scenario. Emerging novel classes of anti-anemic drugs recently introduced or in development for other types of anemia include activin receptor ligand traps, hypoxia-inducible factor-prolyl hydroxylase inhibitors, and hepcidin antagonists. Here, we discuss their possible role in the treatment of anemia observed in patients receiving anticancer therapies.
Collapse
Affiliation(s)
- Claudia Bozzini
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Giacomo Marchi
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Claudio Cerchione
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giovanni Martinelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
- EuroBloodNet Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| |
Collapse
|
21
|
Jin K, Liao YC, Cheng TC, Li X, Lee WJ, Pi F, Jasinski D, Chen LC, Phelps MA, Ho YS, Guo P. In Vitro and In Vivo Evaluation of the Pathology and Safety Aspects of Three- and Four-Way Junction RNA Nanoparticles. Mol Pharm 2024; 21:718-728. [PMID: 38214504 PMCID: PMC10976369 DOI: 10.1021/acs.molpharmaceut.3c00845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
RNA therapeutics has advanced into the third milestone in pharmaceutical drug development, following chemical and protein therapeutics. RNA itself can serve as therapeutics, carriers, regulators, or substrates in drug development. Due to RNA's motile, dynamic, and deformable properties, RNA nanoparticles have demonstrated spontaneous targeting and accumulation in cancer vasculature and fast excretion through the kidney glomerulus to urine to prevent possible interactions with healthy organs. Furthermore, the negatively charged phosphate backbone of RNA results in general repulsion from negatively charged lipid cell membranes for further avoidance of vital organs. Thus, RNA nanoparticles can spontaneously enrich tumor vasculature and efficiently enter tumor cells via specific targeting, while those not entering the tumor tissue will clear from the body quickly. These favorable parameters have led to the expectation that RNA has low or little toxicity. RNA nanoparticles have been well characterized for their anticancer efficacy; however, little detail on RNA nanoparticle pathology and safety is known. Here, we report the in vitro and in vivo assessment of the pathology and safety aspects of different RNA nanoparticles including RNA three-way junction (3WJ) harboring 2'-F modified pyrimidine, folic acid, and Survivin siRNA, as well as the RNA four-way junction (4WJ) harboring 2'-F modified pyrimidine and 24 copies of SN38. Both animal models and patient serum were investigated. In vitro studies include hemolysis, platelet aggregation, complement activation, plasma coagulation, and interferon induction. In vivo studies include hematoxylin and eosin (H&E) staining, hematological and biochemical analysis as the serum profiling, and animal organ weight study. No significant toxicity, side effect, or immune responses were detected during the extensive safety evaluations of RNA nanoparticles. These results further complement previous cancer inhibition studies and demonstrate RNA nanoparticles as an effective and safe drug delivery vehicle for future clinical translations.
Collapse
Affiliation(s)
- Kai Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - You-Cheng Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110031, Taiwan
| | - Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 406040, Taiwan
| | - Xin Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Wen-Jui Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Fengmei Pi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Daniel Jasinski
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Li-Ching Chen
- Department of Biological Science and Technology, China Medical University, Taichung 406040, Taiwan
| | - Mitch A Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yuan-Soon Ho
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 406040, Taiwan
| | - Peixuan Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, Ohio 43210, United States
- James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
22
|
Singh K, Agrawal L, Gupta R, Singh D, Kathpalia M, Kaur N. Lectins as a promising therapeutic agent for breast cancer: A review. Breast Dis 2024; 43:193-211. [PMID: 38905027 PMCID: PMC11307042 DOI: 10.3233/bd-230047] [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: 06/23/2024]
Abstract
Efficient treatment of cancer has been a subject of research by scientists for many years. Current treatments for cancer, such as radiotherapy, chemotherapy and surgery have been used in traditional combination therapy, but they have major setbacks like non-specificity, non-responsiveness in certain cancer types towards treatment, tumor recurrence, etc. Epidemiological data has shown that breast cancer accounts for 14% of cancer cases occurring in Indian women. In recent years, scientists have started to focus on the use of natural compounds like lectins obtained from various sources to counter the side effects of traditional therapy. Lectins like Sambucus nigra Agglutinin, Maackia amurensis lectin, Okra lectins, Haliclona caerulea lectin, Sclerotium rolfsii lectin, etc., have been discovered to have both diagnostic and therapeutic potential for breast cancer patients. Lectins have been found to have inhibitory effects on various cancer cell activities such as neo-angiogenesis, causing cell cycle arrest at the G1 phase, and inducing apoptosis. The major idea behind the use of lectins in cancer diagnostics and therapeutics is their capability to bind to glycosylated proteins that are expressed on the cell surface. This review focuses on an exploration of the roles of post-translational modification in cancer cells, especially glycosylation, and the potential of lectins in cancer diagnosis and therapeutics.
Collapse
Affiliation(s)
- Keerti Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Lokita Agrawal
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Rhea Gupta
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Divyam Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Meghavi Kathpalia
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Navkiran Kaur
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
23
|
Richard ES, Hrycyshyn A, Salman N, Remtulla Tharani A, Abbruzzino A, Smith J, Kachura JJ, Sholzberg M, Mosko JD, Chadi SA, Burkes RL, Pankiw M, Brezden-Masley C. Iron Surveillance and Management in Gastro-Intestinal Oncology Patients: A National Physician Survey. Curr Oncol 2023; 30:9836-9848. [PMID: 37999134 PMCID: PMC10670404 DOI: 10.3390/curroncol30110714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/10/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
PURPOSE Iron deficiency (ID) is a complication of gastrointestinal (GI) cancers that may manifest as iron deficiency anemia (IDA). Serum ferritin monitoring and oral iron supplementation have the limitations of being falsely elevated and poorly absorbed, respectively. This study aims to assess the discordance in surveillance, treatment practices, and awareness of ID/IDA in GI cancer patients by Canadian physicians treating these patients. METHODS From February 2020 to September 2021, a 22-question electronic survey was sent to medical oncologists (MOs), surgical oncologists (SOs), and gastroenterologists (GEs). The survey collected information about four domains: physician demographics, surveillance practices, treatment practices, and awareness of ID/IDA in GI cancer patients and ASCO/ASH guidelines. RESULTS A total of 108 (34 MOs, 19 SOs, and 55 GEs) of the 872 (12.4%) invited physicians completed the survey. Of these, 26.5% of MOs, 36.8% of SOs, and 70.9% of GEs measured baseline iron parameters, with few continuing surveillance throughout treatment. Ferritin was widely measured by MOs (88.9%), SOs (100%), and GEs (91.4%). Iron was supplemented if ID/IDA was identified pre-treatment by 66.7% of MOs, 85.7% of SOs, and 94.2% of GEs. Parenteral iron was prescribed by SOs (100%), while oral iron was prescribed by MOs (83.3%) and GEs (87.9%). Only 18.6% of physicians were aware of the ASCO/ASH guidelines regarding erythropoiesis-stimulating agents with parenteral iron for treating chemotherapy-induced anemia. CONCLUSION Results illustrate variations in practice patterns for IDA management across the different physician specialties. Moreover, there appeared to be gaps in the knowledge and care surrounding evidence-based IDA management principles which may contribute to poor clinical outcomes.
Collapse
Affiliation(s)
- Emilie S. Richard
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Adriyan Hrycyshyn
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Noor Salman
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | | | - Alexandria Abbruzzino
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Janet Smith
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Jacob J. Kachura
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Michelle Sholzberg
- Department of Medicine, Division of Oncology/Hematology, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Jeffrey D. Mosko
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
- Department of Medicine, Division of Gastroenterology, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Sami A. Chadi
- Sprott Department of Surgery, Division of General Surgery, Toronto General Hospital, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ronald L. Burkes
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Maya Pankiw
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Christine Brezden-Masley
- Department of Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
24
|
Ruiz de Porras V, Figols M, Font A, Pardina E. Curcumin as a hepatoprotective agent against chemotherapy-induced liver injury. Life Sci 2023; 332:122119. [PMID: 37741319 DOI: 10.1016/j.lfs.2023.122119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Despite significant advances in cancer therapeutics, chemotherapy remains the cornerstone of treatment for many tumors. Importantly, however, chemotherapy-induced toxicity, including hepatotoxicity, can lead to the interruption or discontinuation of potentially effective therapy. In recent years, special attention has been paid to the search for complementary therapies to mitigate chemotherapy-induced toxicity. Although there is currently a lack of specific interventions to mitigate or prevent hepatotoxicity in chemotherapy-treated patients, the polyphenol compound curcumin has emerged as a potential strategy to overcome this adverse effect. Here we review, firstly, the molecular and physiological mechanisms and major risk factors of chemotherapy-induced hepatotoxicity. We then present an overview of how curcumin has the potential to mitigate hepatotoxicity by targeting specific molecular mechanisms. Hepatotoxicity is a well-described side effect of cytotoxic drugs that can limit their clinical application. Inflammation and oxidative stress are the most common mechanisms involved in hepatotoxicity. Several studies have shown that curcumin could prevent and/or palliate chemotherapy-induced liver injury, mainly due to its anti-inflammatory, antioxidant, antifibrotic and hypolipidemic properties. Further clinical investigation using bioavailable curcumin formulations is warranted to demonstrate its efficacy as an hepatoprotective agent in cancer patients.
Collapse
Affiliation(s)
- Vicenç Ruiz de Porras
- Grup de Recerca en Toxicologia (GRET), Unitat de Toxicologia, Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Avda Joan XXIII s/n, 08028 Barcelona, Spain; CARE program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain; Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain.
| | - Mariona Figols
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243, Manresa, Barcelona, Spain
| | - Albert Font
- CARE program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain; Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain; Medical Oncology Department, Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain
| | - Eva Pardina
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Diagonal 643, 08028 Barcelona, Spain.
| |
Collapse
|
25
|
Živalj M, Van Ginderachter JA, Stijlemans B. Lipocalin-2: A Nurturer of Tumor Progression and a Novel Candidate for Targeted Cancer Therapy. Cancers (Basel) 2023; 15:5159. [PMID: 37958332 PMCID: PMC10648573 DOI: 10.3390/cancers15215159] [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: 09/15/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Within the tumor microenvironment (TME) exists a complex signaling network between cancer cells and stromal cells, which determines the fate of tumor progression. Hence, interfering with this signaling network forms the basis for cancer therapy. Yet, many types of cancer, in particular, solid tumors, are refractory to the currently used treatments, so there is an urgent need for novel molecular targets that could improve current anti-cancer therapeutic strategies. Lipocalin-2 (Lcn-2), a secreted siderophore-binding glycoprotein that regulates iron homeostasis, is highly upregulated in various cancer types. Due to its pleiotropic role in the crosstalk between cancer cells and stromal cells, favoring tumor progression, it could be considered as a novel biomarker for prognostic and therapeutic purposes. However, the exact signaling route by which Lcn-2 promotes tumorigenesis remains unknown, and Lcn-2-targeting moieties are largely uninvestigated. This review will (i) provide an overview on the role of Lcn-2 in orchestrating the TME at the level of iron homeostasis, macrophage polarization, extracellular matrix remodeling, and cell migration and survival, and (ii) discuss the potential of Lcn-2 as a promising novel drug target that should be pursued in future translational research.
Collapse
Affiliation(s)
- Maida Živalj
- Brussels Center for Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Jo A. Van Ginderachter
- Brussels Center for Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Benoit Stijlemans
- Brussels Center for Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| |
Collapse
|
26
|
Myers G, Wang Y, Wang Q, Friedman A, Sanchez-Martinez A, Liu X, Sharon SA, Lim KC, Khoriaty R, Engel JD, Yu L. Murine erythroid differentiation kinetics in vivo under normal and anemic stress conditions. Blood Adv 2023; 7:5727-5732. [PMID: 37552129 PMCID: PMC10539864 DOI: 10.1182/bloodadvances.2023010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/10/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023] Open
Abstract
Our current understanding of the kinetics and dynamics of erythroid differentiation is based almost entirely on the ex vivo expansion of cultured hematopoietic progenitor cells. In this study, we used an erythroid-specific, inducible transgenic mouse line to investigate for the first time, the in vivo erythroid differentiation kinetics under steady-state conditions. We demonstrated that bipotent premegakaroycyte/erythroid (PreMegE) progenitor cells differentiate into erythroid-committed proerythroblast/basophilic erythroblasts (ProBasoE) after 6.6 days under steady-state conditions. During this process, each differentiation phase (from PreMegE to precolony forming unit-erythroid [PreCFU-E], PreCFU-E to CFU-E, and CFU-E to ProBasoE) took ∼2 days in vivo. Upon challenge with 5-flurouracil (5-FU), which leads to the induction of stress erythropoiesis, erythroid maturation time was reduced from 6.6 to 4.7 days. Furthermore, anemia induced in 5-FU-treated mice was shown to be due not only to depleted bone marrow erythroid progenitor stores but also to a block in reticulocyte exit from the bone marrow into the circulation, which differed from the mechanism induced by acute blood loss.
Collapse
Affiliation(s)
- Greggory Myers
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Yu Wang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Qing Wang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Ann Friedman
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | | | - Xiaofang Liu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Singh A. Sharon
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI
| | - Kim-Chew Lim
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Rami Khoriaty
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | - James Douglas Engel
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | - Lei Yu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
27
|
Alghareeb SA, Alsughayyir J, Alfhili MA. Stimulation of Hemolysis and Eryptosis by α-Mangostin through Rac1 GTPase and Oxidative Injury in Human Red Blood Cells. Molecules 2023; 28:6495. [PMID: 37764276 PMCID: PMC10535552 DOI: 10.3390/molecules28186495] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Chemotherapy-related anemia is prevalent in up to 75% of patients, which may arise due to hemolysis and eryptosis. Alpha-mangostin (α-MG) is a polyphenolic xanthonoid found in the mangosteen tree (Garcinia mangostana) whose antitumor medicinal properties are well-established. Nevertheless, the potential toxic effects of α-MG on red blood cells (RBCs) have, as of yet, not been as well studied. METHODS RBCs were exposed to 1-40 μM of α-MG for 24 h at 37 °C. Hemolysis and related markers were measured using colorimetric assays, eryptotic cells were identified through Annexin-V-FITC, Ca2+ was detected with Fluo4/AM, and oxidative stress was assessed through H2DCFDA using flow cytometry. The toxicity of α-MG was also examined in the presence of specific signal transduction inhibitors and in whole blood. RESULTS α-MG at 10-40 μM caused dose-dependent hemolysis with concurrent significant elevation in K+, Mg2+, and LDH leakage, but at 2.5 μM it significantly increased the osmotic resistance of cells. A significant increase was also noted in Annexin-V-binding cells, along with intracellular Ca2+, oxidative stress, and cell shrinkage. Moreover, acetylcholinesterase activity was significantly inhibited by α-MG, whose hemolytic potential was significantly ameliorated by the presence of BAPTA-AM, vitamin C, NSC23766, and isosmotic sucrose but not urea. In whole blood, α-MG significantly depleted intracellular hemoglobin stores and was selectively toxic to platelets and monocytes. CONCLUSIONS α-MG possesses hemolytic and eryptotic activities mediated through Ca2+ signaling, Rac1 GTPase activity, and oxidative injury. Also, α-MG leads to accelerated cellular aging and specifically targets platelet and monocyte populations in a whole blood milieu.
Collapse
Affiliation(s)
| | | | - Mohammad A. Alfhili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia (J.A.)
| |
Collapse
|
28
|
Berrada S, Martínez-Balsalobre E, Larcher L, Azzoni V, Vasquez N, Da Costa M, Abel S, Audoly G, Lee L, Montersino C, Castellano R, Combes S, Gelot C, Ceccaldi R, Guervilly JH, Soulier J, Lachaud C. A clickable melphalan for monitoring DNA interstrand crosslink accumulation and detecting ICL repair defects in Fanconi anemia patient cells. Nucleic Acids Res 2023; 51:7988-8004. [PMID: 37395445 PMCID: PMC10450163 DOI: 10.1093/nar/gkad559] [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: 11/25/2022] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023] Open
Abstract
Fanconi anemia (FA) is a genetic disorder associated with developmental defects, bone marrow failure and cancer. The FA pathway is crucial for the repair of DNA interstrand crosslinks (ICLs). In this study, we have developed and characterized a new tool to investigate ICL repair: a clickable version of the crosslinking agent melphalan which we name click-melphalan. Our results demonstrate that click-melphalan is as effective as its unmodified counterpart in generating ICLs and associated toxicity. The lesions induced by click-melphalan can be detected in cells by post-labelling with a fluorescent reporter and quantified using flow cytometry. Since click-melphalan induces both ICLs and monoadducts, we generated click-mono-melphalan, which only induces monoadducts, in order to distinguish between the two types of DNA repair. By using both molecules, we show that FANCD2 knock-out cells are deficient in removing click-melphalan-induced lesions. We also found that these cells display a delay in repairing click-mono-melphalan-induced monoadducts. Our data further revealed that the presence of unrepaired ICLs inhibits monoadduct repair. Finally, our study demonstrates that these clickable molecules can differentiate intrinsic DNA repair deficiencies in primary FA patient cells from those in primary xeroderma pigmentosum patient cells. As such, these molecules may have potential for developing diagnostic tests.
Collapse
Affiliation(s)
- Sara Berrada
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | - Lise Larcher
- University Paris Cité, Institut de Recherche Saint-Louis, INSERM U944, and CNRS UMR7212, Paris, France
- Laboratoire de biologie médicale de référence (LBMR) “Aplastic anemia”, Service d’Hématologie biologique, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Violette Azzoni
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Nadia Vasquez
- University Paris Cité, Institut de Recherche Saint-Louis, INSERM U944, and CNRS UMR7212, Paris, France
- Laboratoire de biologie médicale de référence (LBMR) “Aplastic anemia”, Service d’Hématologie biologique, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mélanie Da Costa
- University Paris Cité, Institut de Recherche Saint-Louis, INSERM U944, and CNRS UMR7212, Paris, France
- Laboratoire de biologie médicale de référence (LBMR) “Aplastic anemia”, Service d’Hématologie biologique, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Sébastien Abel
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Gilles Audoly
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Lara Lee
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Camille Montersino
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Rémy Castellano
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Sébastien Combes
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Camille Gelot
- Inserm U830, PSL Research University, Institut Curie, Paris, France
| | - Raphaël Ceccaldi
- Inserm U830, PSL Research University, Institut Curie, Paris, France
| | | | - Jean Soulier
- University Paris Cité, Institut de Recherche Saint-Louis, INSERM U944, and CNRS UMR7212, Paris, France
- Laboratoire de biologie médicale de référence (LBMR) “Aplastic anemia”, Service d’Hématologie biologique, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Christophe Lachaud
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| |
Collapse
|
29
|
Shiraishi C, Hirai T, Kaneda M, Okamoto A, Kato H, Tanaka K, Kondo E, Ikeda T, Iwamoto T. Factors for the development of anemia in patients with newly introduced olaparib: A retrospective case-control study. Medicine (Baltimore) 2023; 102:e34123. [PMID: 37505180 PMCID: PMC10378826 DOI: 10.1097/md.0000000000034123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 07/29/2023] Open
Abstract
Anemia is the most common dose-limiting toxicity of olaparib. However, few studies have analyzed the clinical features of olaparib-induced anemia. This study investigated the clinical features of olaparib-induced anemia. Additionally, the role of folate or vitamin B12 in olaparib-induced anemia was examined. This retrospective case-control study included patients who received olaparib at Mie University Hospital between January 2018 and December 2020. Data were collected between initiation of olaparib and discontinuation of olaparib or till December 2021. We investigated the development of grade ≥ 3 anemia during olaparib administration for at least 1 year. We examined patients with grade ≥ 3 anemia considering the mean corpuscular volume (MCV), its association with gastrointestinal events and cumulative dose of carboplatin. For the sub-study analysis, data on patients treated with olaparib for ovarian or endometrial cancer were collected to evaluate the Common Terminology Criteria for Adverse Events (CTCAE) or monthly changes in folate or vitamin B12 levels from baseline to 3 months after olaparib initiation. These data were collected between initiation of olaparib and discontinuation of olaparib or till November 2022. Patients with no data on folic acid or vitamin B12 levels were excluded from the sub-study. In the main study, 40 patients were included. Eighteen patients (45%) developed grade ≥ 3 anemia, and all patients discontinued treatment (94%) or reduced olaparib dose (67%) after developing anemia. Among the patients with grade ≥ 3 anemia, 9 (50%) exhibited macrocytic anemia and 15 (83%) had previously received carboplatin. The incidence of grade ≥ 2 dysgeusia was significantly higher in patients with grade ≥ 3 anemia (P = .034). Moreover, the cumulative dose of previously administered carboplatin was higher in patients who had 3 episodes of anemia (P = .102). In sub-study, 12 had data on folic acid and vitamin B12 levels. Sub-study analysis showed that none fulfilled the criteria for deficiency of folate or vitamin B12, while 3 developed grade 3 anemia. This study revealed that olaparib-induced anemia frequently occurs as macrocytic and normocytic erythroblastic anemia without folate or vitamin B12 deficiencies. A high cumulative dose of previously administered carboplatin and dysgeusia may be associated with olaparib-induced anemia.
Collapse
Affiliation(s)
| | | | - Michiko Kaneda
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Mie, Japan
| | | | - Hideo Kato
- Department of Pharmacy, Mie University Hospital, Mie, Japan
| | - Kayo Tanaka
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Mie, Japan
| | - Eiji Kondo
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Mie, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Mie, Japan
| | - Takuya Iwamoto
- Department of Pharmacy, Mie University Hospital, Mie, Japan
| |
Collapse
|
30
|
Alhebshi SA, Alsanosi SM, AlQashqri HS, Alhindi YZ, Bamagous GA, Ayoub NA, Falemban AH. Toxicity of Nab-Paclitaxel Compared to Paclitaxel in a Tertiary Hospital in Jeddah, Saudi Arabia: A Retrospective Cohort Study. Cureus 2023; 15:e39872. [PMID: 37404399 PMCID: PMC10315099 DOI: 10.7759/cureus.39872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 07/06/2023] Open
Abstract
Background Nanoparticle albumin-bound paclitaxel has been developed to avoid the toxicities associated with Cremophor-solved paclitaxel. Although many studies confirm this hypothesis, there is recent evidence showing no difference between paclitaxel and nab-paclitaxel in their efficacy and safety profile. This study further assesses the toxicity of both paclitaxel and nab-paclitaxel in adult patients with breast and pancreatic cancer in a tertiary hospital in Jeddah, Saudi Arabia. These toxicities include neutropenia, anaemia, and effects on kidney and liver functions. Methods The study is a retrospective cohort study done at King Abdulaziz University Hospital, Jeddah, Saudi Arabia, from January 2018 to December 2021, conducted on patients diagnosed with breast or pancreatic cancer treated with paclitaxel or nab-paclitaxel. Results There is a statistically significant difference between the two groups in developing anaemia, renal, and liver toxicity (P<0.05). On the other hand, there are no statistically significant differences in developing neutropenia between the two groups (P=0.084). Conclusions Nab-paclitaxel might not be better than paclitaxel in reducing the risk of neutropenia, anaemia, and liver toxicity, as predicted. Nevertheless, both medications require that the patient's renal functions be monitored during the treatment. Further studies conducted in multiple oncology centres with a larger sample are needed to evaluate the toxicity of paclitaxel and nab-paclitaxel in adult patients with breast and pancreatic cancer.
Collapse
Affiliation(s)
- Suha A Alhebshi
- Pharmaceutical Services, King Abdulaziz University Hospital, Jeddah, SAU
| | - Safaa M Alsanosi
- Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Hamsa S AlQashqri
- Community and Family Medicine, Faculty of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Yosra Z Alhindi
- Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Ghazi A Bamagous
- Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Nahla A Ayoub
- Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, SAU
| | - Alaa H Falemban
- Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, SAU
| |
Collapse
|
31
|
Motwani SS, Kaur SS, Kitchlu A. Cisplatin Nephrotoxicity: Novel Insights Into Mechanisms and Preventative Strategies. Semin Nephrol 2023; 42:151341. [PMID: 37182407 DOI: 10.1016/j.semnephrol.2023.151341] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cisplatin is a highly effective chemotherapeutic agent that has been used for more than 50 years for a variety of cancers; however, its use is limited by toxicity, including nephrotoxicity. In this in-depth review, we discuss the incidence of cisplatin-associated acute kidney injury, as well as common risk factors for its development. Cisplatin accumulates in the kidney tubules and causes AKI through various mechanisms, including DNA damage, oxidative stress, and apoptosis. We also discuss the spectrum of nephrotoxicity, including acute and chronic impairment of kidney function, electrolyte disturbances, and thrombotic microangiopathy. We discuss the limited options for the diagnosis, prevention, and management of these complications, along with factors that may impact future therapy with or without cisplatin. We conclude with directions for future research in this expanding and important area.
Collapse
Affiliation(s)
- Shveta S Motwani
- Division of Nephrology, Lahey Hospital and Medical Center, Burlington, MA.
| | - Sharneet Sandhu Kaur
- Division of Nephrology, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Abhijat Kitchlu
- Division of Nephrology, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Chambers C, Cermakova K, Chan YS, Kurtz K, Wohlan K, Lewis AH, Wang C, Pham A, Dejmek M, Sala M, Loeza Cabrera M, Aguilar R, Nencka R, Lacorazza HD, Rau RE, Hodges HC. SWI/SNF Blockade Disrupts PU.1-Directed Enhancer Programs in Normal Hematopoietic Cells and Acute Myeloid Leukemia. Cancer Res 2023; 83:983-996. [PMID: 36662812 PMCID: PMC10071820 DOI: 10.1158/0008-5472.can-22-2129] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/09/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
In acute myeloid leukemia (AML), SWI/SNF chromatin remodeling complexes sustain leukemic identity by driving high levels of MYC. Previous studies have implicated the hematopoietic transcription factor PU.1 (SPI1) as an important target of SWI/SNF inhibition, but PU.1 is widely regarded to have pioneer-like activity. As a result, many questions have remained regarding the interplay between PU.1 and SWI/SNF in AML as well as normal hematopoiesis. Here we found that PU.1 binds to most of its targets in a SWI/SNF-independent manner and recruits SWI/SNF to promote accessibility for other AML core regulatory factors, including RUNX1, LMO2, and MEIS1. SWI/SNF inhibition in AML cells reduced DNA accessibility and binding of these factors at PU.1 sites and redistributed PU.1 to promoters. Analysis of nontumor hematopoietic cells revealed that similar effects also impair PU.1-dependent B-cell and monocyte populations. Nevertheless, SWI/SNF inhibition induced profound therapeutic response in an immunocompetent AML mouse model as well as in primary human AML samples. In vivo, SWI/SNF inhibition promoted leukemic differentiation and reduced the leukemic stem cell burden in bone marrow but also induced leukopenia. These results reveal a variable therapeutic window for SWI/SNF blockade in AML and highlight important off-tumor effects of such therapies in immunocompetent settings. SIGNIFICANCE Disruption of PU.1-directed enhancer programs upon SWI/SNF inhibition causes differentiation of AML cells and induces leukopenia of PU.1-dependent B cells and monocytes, revealing the on- and off-tumor effects of SWI/SNF blockade.
Collapse
Affiliation(s)
- Courtney Chambers
- Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas
| | - Katerina Cermakova
- Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Yuen San Chan
- Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Kristen Kurtz
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Katharina Wohlan
- Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
| | - Andrew Henry Lewis
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Christiana Wang
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas
| | - Anh Pham
- Department of Bioengineering, Rice University, Houston, Texas
| | - Milan Dejmek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michal Sala
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Mario Loeza Cabrera
- Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Rogelio Aguilar
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas
| | - Radim Nencka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - H. Daniel Lacorazza
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Rachel E. Rau
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas
| | - H. Courtney Hodges
- Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
- Department of Bioengineering, Rice University, Houston, Texas
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
33
|
Chemotherapy-induced tumor immunogenicity is mediated in part by megakaryocyte-erythroid progenitors. Oncogene 2023; 42:771-781. [PMID: 36646904 PMCID: PMC9984299 DOI: 10.1038/s41388-023-02590-0] [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: 05/18/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
Chemotherapy remains one of the main treatment modalities for cancer. While chemotherapy is mainly known for its ability to kill tumor cells directly, accumulating evidence indicates that it also acts indirectly by enhancing T cell-mediated anti-tumor immunity sometimes through immunogenic cell death. However, the role of immature immune cells in chemotherapy-induced immunomodulation has not been studied. Here, we utilized a mouse pancreatic cancer model to characterize the effects of gemcitabine chemotherapy on immature bone marrow cells in the context of tumor immunogenicity. Single cell RNA sequencing of hematopoietic stem and progenitor cells revealed a 3-fold increase in megakaryocyte-erythroid progenitors (MEPs) in the bone marrow of gemcitabine-treated mice in comparison to untreated control mice. Notably, adoptive transfer of MEPs to pancreatic tumor-bearing mice significantly reduced tumor growth and increased the levels of anti-tumor immune cells in tumors and peripheral blood. Furthermore, MEPs increased the tumor cell killing activity of CD8 + T cells and NK cells, an effect that was dependent on MEP-secreted CCL5 and CXCL16. Collectively, our findings demonstrate that chemotherapy-induced enrichment of MEPs in the bone marrow compartment contributes to anti-tumor immunity.
Collapse
|
34
|
Kavya D, Nadumane VK. A combination of semi-purified L-methioninase with tamoxifen citrate to ameliorate breast cancer in athymic nude mice. Mol Biol Rep 2023; 50:2925-2932. [PMID: 36566301 DOI: 10.1007/s11033-022-08144-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/22/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Chemotherapy nonspecifically targets both tumor and healthy proliferating cells. Methionine deprivation using L-methioninase along with chemotherapy appears promising towards cancer management. The present study is an attempt to use a new combination of L-methioninase with Tamoxifen (TAM) to treat breast cancer in mice. METHODS AND RESULTS L-Methioninase from Methylobacterium sp. was partially purified (SPMet's) by cold acetone precipitation and lyophilized. Its cytotoxicity effect, alone and in combination with Tamoxifen, was evaluated in vitro (MCF-7) cells and in vivo (athymic nude mice) conditions. SPMet's was found to inhibit the growth of MCF-7 cells with an IC50 value of 47.05 µg/ml, while the combination of SPMet's and TAM had an IC50 of 6.4 µg/ml. Athymic nude mice were grouped into: Group-I - Tumor control; Group-II - TAM; Group-III - SPMet's; Group-IV - SPMet's + TAM. Tumor growth inhibition (TGI) was maximum in Group-IV with 84.65% followed by Group-II with 65.12%. Hematological and Biochemical parameters in Group-II, III, and IV were restored to normal levels. Tumor histopathology showed increased apoptosis and necrosis in Group-IV. Caspases 3 & 8 gene upregulation was significantly higher in Group-IV than other treated groups, indicating higher efficacy of the combination approach. CONCLUSION This is the first study report about a combination of SPMet's and TAM on in vivo breast cancer model, with significantly higher anticancer activity and without noticeable side effects. The findings of this study have several important implications for future clinical studies.
Collapse
Affiliation(s)
- D Kavya
- Department of Biotechnology, School of Sciences, Jain Deemed-to-be-University, Bangalore, Karnataka, India
| | | |
Collapse
|
35
|
Sakaeva DD. Anemia and iron deficiency in cancer patients: the role of intravenous iron supplements (a literature review). JOURNAL OF MODERN ONCOLOGY 2023. [DOI: 10.26442/18151434.2022.4.202018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Anemia in patients with malignancies is a common disorder that has a markedly negative impact on quality of life and overall prognosis. The pathogenesis of anemia is complex and multifactorial, depending on the type and stage of malignancy, nutritional status, renal function, age and gender, cytostatic drug, dose, and chemotherapeutic regimen, with iron deficiency often being the main and potentially treatable factor for anemia. In cancer patients, it can be caused by various concomitant mechanisms, including bleeding (e.g., in malignant gastrointestinal tumors or after surgery), malnutrition, medication, and hepcidin-induced iron sequestration in macrophages, with subsequent iron-deficient erythropoiesis. The variety of clinical manifestations of anemia makes it challenging to establish universal criteria to develop optimal treatments. Current therapy for anemia in malignant tumors includes replacement therapy with an iron supplement, erythropoiesis-stimulating agents (erythropoietins), and blood transfusions. However, blood transfusions should be minimized due to the high risks and costs. Therapy with an iron supplement is an effective approach to correcting the iron deficiency. It can increase the efficacy of erythropoiesis-stimulating drugs and reduce the need for blood transfusions. Published guidelines suggest the wide use of intravenous iron supplements. This article discusses possible approaches to treating iron deficiency in cancer patients in various clinical settings. We build on current guidelines and emphasize the need for further research in this area.
Collapse
|
36
|
Parveen N, Abourehab MA, Shukla R, Thanikachalam PV, Jain GK, Kesharwani P. Immunoliposomes as an emerging nanocarrier for breast cancer therapy. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2022.111781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
37
|
Uner OC, Kuru HI, Cinbis RG, Tastan O, Cicek AE. DeepSide: A Deep Learning Approach for Drug Side Effect Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:330-339. [PMID: 34995191 DOI: 10.1109/tcbb.2022.3141103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Drug failures due to unforeseen adverse effects at clinical trials pose health risks for the participants and lead to substantial financial losses. Side effect prediction algorithms have the potential to guide the drug design process. LINCS L1000 dataset provides a vast resource of cell line gene expression data perturbed by different drugs and creates a knowledge base for context specific features. The state-of-the-art approach that aims at using context specific information relies on only the high-quality experiments in LINCS L1000 and discards a large portion of the experiments. In this study, our goal is to boost the prediction performance by utilizing this data to its full extent. We experiment with 5 deep learning architectures. We find that a multi-modal architecture produces the best predictive performance among multi-layer perceptron-based architectures when drug chemical structure (CS), and the full set of drug perturbed gene expression profiles (GEX) are used as modalities. Overall, we observe that the CS is more informative than the GEX. A convolutional neural network-based model that uses only SMILES string representation of the drugs achieves the best results and provides 13.0% macro-AUC and 3.1% micro-AUC improvements over the state-of-the-art. We also show that the model is able to predict side effect-drug pairs that are reported in the literature but was missing in the ground truth side effect dataset. DeepSide is available at http://github.com/OnurUner/DeepSide.
Collapse
|
38
|
VanderVeen BN, Cardaci TD, McDonald SJ, Madero SS, Unger CA, Bullard BM, Enos RT, Velázquez KT, Kubinak JL, Fan D, Murphy EA. Obesity reduced survival with 5-fluorouracil and did not protect against chemotherapy-induced cachexia or immune cell cytotoxicity in mice. Cancer Biol Ther 2022; 23:1-15. [PMID: 35968771 PMCID: PMC9377261 DOI: 10.1080/15384047.2022.2108306] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/28/2022] [Accepted: 07/24/2022] [Indexed: 01/12/2023] Open
Abstract
Fluorouracil/5-flourouracil (5FU) is a first-line chemotherapy drug for many cancer types; however, its associated toxicities contribute to poor quality of life and reduced dose intensities negatively impacting patient prognosis. While obesity remains a critical risk factor for most cancers, our understanding regarding how obesity may impact chemotherapy's toxicities is extremely limited. C56BL/6 mice were given high fat (Obese) or standard diets (Lean) for 4 months and then subjected to three cycles of 5FU (5d-40 mg/kg Lean Mass, 9d rest) or PBS vehicle control. Shockingly, only 60% of Obese survived 3 cycles compared to 100% of Lean, and Obese lost significantly more body weight. Dihydropyrimidine dehydrogenase (DPD), the enzyme responsible for 5FU catabolism, was reduced in obese livers. Total white blood cells, neutrophils, and lymphocytes were reduced in Obese 5FU compared to Lean 5FU and PBS controls. While adipocyte size was not affected by 5FU in Obese, skeletal muscle mass and myofibrillar cross section area were decreased following 5FU in Lean and Obese. Although adipose tissue inflammatory gene expression was not impacted by 5FU, distinct perturbations to skeletal muscle inflammatory gene expression and immune cell populations (CD45+ Immune cells, CD45+CD11b+CD68+ macrophages and CD45+CD11b+Ly6clo/int macrophage/monocytes) were observed in Obese only. Our evidence suggests that obesity induced liver pathologies and reduced DPD exacerbated 5FU toxicities. While obesity has been suggested to protect against cancer/chemotherapy-induced cachexia and other toxicities, our results demonstrate that obese mice are not protected, but rather show evidence of increased susceptibility to 5FU-induced cytotoxicity even when dosed for relative lean mass.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Thomas D. Cardaci
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Sierra J. McDonald
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Sarah S. Madero
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Christian A. Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Brooke M. Bullard
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Reilly T. Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Kandy T. Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Jason L. Kubinak
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| |
Collapse
|
39
|
Wadowska K, Błasiak P, Rzechonek A, Bil-Lula I, Śliwińska-Mossoń M. Hepcidin as a Diagnostic Biomarker in Anaemic Lung Cancer Patients. Cancers (Basel) 2022; 15:cancers15010224. [PMID: 36612220 PMCID: PMC9818260 DOI: 10.3390/cancers15010224] [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: 11/17/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
We aim to describe the characteristics of hepcidin, IL-6, and TNF-α levels in anaemia of lung cancer patients with operative tumour as well as to investigate the potential diagnostic capabilities of hepcidin in combination with IL-6, TNF-α, and acute phase proteins. We present a retrospective study of 112 lung cancer patients (41 women and 71 men) who were surgically treated at the Lower Silesian Centre for Lung Diseases in Wroclaw, Poland. Serum blood samples were collected from all these patients prior to any surgical treatment and used to determine hepcidin, IL-6, TNF-α, SAA1, and CRP concentrations. Patients were also examined with a complete blood count several times during their hospitalization. The female and male groups were divided based on the occurrence of anaemia during their hospitalization. Patients who developed anaemia post-operatively had significantly lower hepcidin concentrations than non-anaemic patients (p = 0.000694 in females with ≥3 complete blood count examinations and p = 0.007905 in males with 2 complete blood count examinations), whereas patients with anaemia since hospital admission had higher hepcidin concentrations. We observed two hepcidin roles related to two cancer anaemia pathogeneses: (1) higher hepcidin concentrations in patients with anaemia since hospital admission (anaemia of inflammation) and (2) lower hepcidin concentrations in patients who developed anaemia after surgery (anaemia of iron deficiency). Our data support the role of hepcidin, IL-6, and TNF-α in cancer-related anaemia and provide diagnostic values for predicting post-operative anaemia in lung cancer patients.
Collapse
Affiliation(s)
- Katarzyna Wadowska
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
- Correspondence: ; Tel.: +48-71-784-0626
| | - Piotr Błasiak
- Department and Clinic of Thoracic Surgery, Faculty of Medicine, Wroclaw Medical University, Grabiszyńska 105, 53-439 Wroclaw, Poland
- Lower Silesian Centre of Oncology, Pulmonology and Haematology, Lower Silesian Thoracic Surgery Centre, Grabiszyńska 105, 53-439 Wroclaw, Poland
| | - Adam Rzechonek
- Department and Clinic of Thoracic Surgery, Faculty of Medicine, Wroclaw Medical University, Grabiszyńska 105, 53-439 Wroclaw, Poland
- Lower Silesian Centre of Oncology, Pulmonology and Haematology, Lower Silesian Thoracic Surgery Centre, Grabiszyńska 105, 53-439 Wroclaw, Poland
| | - Iwona Bil-Lula
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Mariola Śliwińska-Mossoń
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
40
|
Kim NH, Park JH, Koo DH, Jung YS, Yang JY, Lee HY. A Pilot Study of Peritumor Administration of 5-FU for Preventing Bleeding in Advanced Gastric Cancer. THE KOREAN JOURNAL OF GASTROENTEROLOGY = TAEHAN SOHWAGI HAKHOE CHI 2022; 80:273-280. [PMID: 36567441 DOI: 10.4166/kjg.2022.099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 12/27/2022]
Abstract
Five-FU is a potent chemotherapeutic agent for suppressing endothelial cell growth. The purpose of this study was to investigate the usefulness of local peritumor injection of 5-FU for patients with advanced gastric cancer (AGC) for the prevention of anemia. Between January 2020 and January 2022, patients aged 18 years or older with AGC and moderate anemia were included. A total of 200 mg of 5-FU was injected per session at ten points of the lesion (20 mg at each point) every 7 days for 4 to 12 weeks. Patients received a blood test for toxicity at every cycle. From one of these patients, endoscopic biopsy specimens were taken from gastric cancer before and after injecting 5-FU for immunostaining. A total of five AGC patients participated in this study. For most patients, hemoglobin levels were maintained without transfusions during 5-FU injection, and expression levels of thrombospondin-1 was increased after injection compared to those before injection. Blood test results during 5-FU injection showed no significant change in serum glutamic oxalacetic transaminase/glutamic pyruvic transaminase, total bilirubin, or creatinine level. The results of this study showed the possibility of local peritumor 5-FU injection as a treatment for relieving anemia of patients with gastric cancer.
Collapse
Affiliation(s)
- Nam-Hee Kim
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - Jung Ho Park
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - Dong-Hoe Koo
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - Yoon Suk Jung
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - Jeong-Yoon Yang
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - Hee-Young Lee
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
Root Bark Extract of Oroxylum indicum Vent. Inhibits Solid and Ascites Tumors and Prevents the Development of DMBA-Induced Skin Papilloma Formation. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238459. [PMID: 36500567 PMCID: PMC9738881 DOI: 10.3390/molecules27238459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/11/2022]
Abstract
Oroxylum indicum is a traditionally used plant in Ayurvedic and folk medicines. The plant is useful for the management of gastrointestinal diseases as well as skin diseases. In the present study, we analyzed the antitumor potential of O. indicum in Dalton's lymphoma ascites tumor cells (DLA) and Ehrlich ascites carcinoma (EAC)-induced solid and ascites tumors. Further, the potential of O. indicum extract (OIM) on skin papilloma induction by dimethyl benz(a) anthracene (DMBA) and croton oil was evaluated. The chemical composition of the extract was analyzed using UPLC-Q-TOF-MS. The predominant compounds present in the extract were demethoxycentaureidin 7-O-rutinoside, isorhamnetin-3-O-rutinoside, baicalein-7-O-glucuronide, 5,6,7-trihydroxyflavone, 3-Hydroxy-3',4',5'-trimethoxyflavone, 5,7-dihydroxy-3-(4-methoxyphenyl) chromen-4-one, and 4'-Hydroxy-5,7-dimethoxyflavanone. Treatment with high-dose OIM enhanced the percentage of survival in ascites tumor-bearing mice by 34.97%. Likewise, high and low doses of OIM reduced the tumor volume in mice by 61.84% and 54.21%, respectively. Further, the skin papilloma formation was brought down by the administration of low- and high-dose groups of OIM (by 67.51% and 75.63%). Overall, the study concludes that the Oroxylum indicum root bark extract is a potentially active antitumor and anticancer agent.
Collapse
|
42
|
Bhattacharjee R, Das D, Bhadhuri R, Chakraborty S, Dey T, Buragohain R, Nath A, Muduli K, Barman P, Gundamaraju R. Cellular Landscaping of COVID-19 and Gynaecological Cancers: An Infrequent Correlation. JOURNAL OF ONCOLOGY 2022; 2022:5231022. [PMID: 36299504 PMCID: PMC9592241 DOI: 10.1155/2022/5231022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/16/2022] [Indexed: 01/08/2023]
Abstract
COVID-19 resulted in a mortality rate of 3-6% caused by SARS-CoV-2 and its variant leading to unprecedented consequences of acute respiratory distress septic shock and multiorgan failure. In such a situation, evaluation, diagnosis, treatment, and care for cancer patients are difficult tasks faced by medical staff. Moreover, patients with gynaecological cancer appear to be more prone to severe infection and mortality from COVID-19 due to immunosuppression by chemotherapy and coexisting medical disorders. To deal with such a circumtances oncologists have been obliged to reconsider the entire diagnostic, treatment, and management approach. This review will provide and discuss the molecular link with gynaecological cancer under COVID-19 infection, providing a novel bilateral relationship between the two infections. Moreover, the authors have provided insights to discuss the pathobiology of COVID-19 in gynaecological cancer and their risks associated with such comorbidity. Furthermore, we have depicted the overall impact of host immunity along with guidelines for the treatment of patients with gynaecological cancer under COVID-19 infection. We have also discussed the feasible scope for the management of COVID-19 and gynaecological cancer.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Debanjan Das
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | | | | | - Tanima Dey
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Rupam Buragohain
- Department of Biotechnology, Gauhati UNiversity, Gopinath Bordoloi Nagar, Guwahati 781014, Assam, India
| | - Asim Nath
- Department of Biotechnology, Gauhati UNiversity, Gopinath Bordoloi Nagar, Guwahati 781014, Assam, India
| | - Kartik Muduli
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Pranjan Barman
- Department of Biotechnology, Gauhati UNiversity, Gopinath Bordoloi Nagar, Guwahati 781014, Assam, India
| | - Rohit Gundamaraju
- ER Stress and Mucosal Immunology Lab, School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
- Division of Gastroenterology, School of Medicine, Washington University at St Louis, St Louis, MO, USA
| |
Collapse
|
43
|
Chou DB, Furlong BA, Posey RR, Kyprianou C, O'Sullivan LR, David R, Randle SJ, Polanska UM, Travers J, Urosevic J, Hutchinson JN, Che J, Howley AM, Hasserjian RP, Prantil-Baun R, Ingber DE. Differential ABC transporter expression during hematopoiesis contributes to neutrophil-biased toxicity of Aurora kinase inhibitors. Nat Commun 2022; 13:6021. [PMID: 36224199 PMCID: PMC9556712 DOI: 10.1038/s41467-022-33672-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 09/21/2022] [Indexed: 11/09/2022] Open
Abstract
Drug-induced cytopenias are a prevalent and significant issue that worsens clinical outcomes and hinders the effective treatment of cancer. While reductions in blood cell numbers are classically associated with traditional cytotoxic chemotherapies, they also occur with newer targeted small molecules and the factors that determine the hematotoxicity profiles of oncologic drugs are not fully understood. Here, we explore why some Aurora kinase inhibitors cause preferential neutropenia. By studying drug responses of healthy human hematopoietic cells in vitro and analyzing existing gene expression datasets, we provide evidence that the enhanced vulnerability of neutrophil-lineage cells to Aurora kinase inhibition is caused by early developmental changes in ATP-binding cassette (ABC) transporter expression. These data show that hematopoietic cell-intrinsic expression of ABC transporters may be an important factor that determines how some Aurora kinase inhibitors affect the bone marrow.
Collapse
Affiliation(s)
- David B Chou
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Brooke A Furlong
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Ryan R Posey
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Christos Kyprianou
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Lucy R O'Sullivan
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Rhiannon David
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Suzanne J Randle
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | - Jon Travers
- Early Oncology, R&D, AstraZeneca, Cambridge, UK
| | | | | | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Anna M Howley
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | | | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA, USA.
| |
Collapse
|
44
|
Adams A, Scheckel B, Habsaoui A, Haque M, Kuhr K, Monsef I, Bohlius J, Skoetz N. Intravenous iron versus oral iron versus no iron with or without erythropoiesis- stimulating agents (ESA) for cancer patients with anaemia: a systematic review and network meta-analysis. Cochrane Database Syst Rev 2022; 6:CD012633. [PMID: 35724934 PMCID: PMC9208863 DOI: 10.1002/14651858.cd012633.pub2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Anaemia is common among cancer patients and they may require red blood cell transfusions. Erythropoiesis-stimulating agents (ESAs) and iron might help in reducing the need for red blood cell transfusions. However, it remains unclear whether the combination of both drugs is preferable compared to using one drug. OBJECTIVES To systematically review the effect of intravenous iron, oral iron or no iron in combination with or without ESAs to prevent or alleviate anaemia in cancer patients and to generate treatment rankings using network meta-analyses (NMAs). SEARCH METHODS We identified studies by searching bibliographic databases (CENTRAL, MEDLINE, Embase; until June 2021). We also searched various registries, conference proceedings and reference lists of identified trials. SELECTION CRITERIA We included randomised controlled trials comparing intravenous, oral or no iron, with or without ESAs for the prevention or alleviation of anaemia resulting from chemotherapy, radiotherapy, combination therapy or the underlying malignancy in cancer patients. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed risk of bias. Outcomes were on-study mortality, number of patients receiving red blood cell transfusions, number of red blood cell units, haematological response, overall mortality and adverse events. We conducted NMAs and generated treatment rankings. We assessed the certainty of the evidence using GRADE. MAIN RESULTS Ninety-six trials (25,157 participants) fulfilled our inclusion criteria; 62 trials (24,603 participants) could be considered in the NMA (12 different treatment options). Here we present the comparisons of ESA with or without iron and iron alone versus no treatment. Further results and subgroup analyses are described in the full text. On-study mortality We estimated that 92 of 1000 participants without treatment for anaemia died up to 30 days after the active study period. Evidence from NMA (55 trials; 15,074 participants) suggests that treatment with ESA and intravenous iron (12 of 1000; risk ratio (RR) 0.13, 95% confidence interval (CI) 0.01 to 2.29; low certainty) or oral iron (34 of 1000; RR 0.37, 95% CI 0.01 to 27.38; low certainty) may decrease or increase and ESA alone (103 of 1000; RR 1.12, 95% CI 0.92 to 1.35; moderate certainty) probably slightly increases on-study mortality. Additionally, treatment with intravenous iron alone (271 of 1000; RR 2.95, 95% CI 0.71 to 12.34; low certainty) may increase and oral iron alone (24 of 1000; RR 0.26, 95% CI 0.00 to 19.73; low certainty) may increase or decrease on-study mortality. Haematological response We estimated that 90 of 1000 participants without treatment for anaemia had a haematological response. Evidence from NMA (31 trials; 6985 participants) suggests that treatment with ESA and intravenous iron (604 of 1000; RR 6.71, 95% CI 4.93 to 9.14; moderate certainty), ESA and oral iron (527 of 1000; RR 5.85, 95% CI 4.06 to 8.42; moderate certainty), and ESA alone (467 of 1000; RR 5.19, 95% CI 4.02 to 6.71; moderate certainty) probably increases haematological response. Additionally, treatment with oral iron alone may increase haematological response (153 of 1000; RR 1.70, 95% CI 0.69 to 4.20; low certainty). Red blood cell transfusions We estimated that 360 of 1000 participants without treatment for anaemia needed at least one transfusion. Evidence from NMA (69 trials; 18,684 participants) suggests that treatment with ESA and intravenous iron (158 of 1000; RR 0.44, 95% CI 0.31 to 0.63; moderate certainty), ESA and oral iron (144 of 1000; RR 0.40, 95% CI 0.24 to 0.66; moderate certainty) and ESA alone (212 of 1000; RR 0.59, 95% CI 0.51 to 0.69; moderate certainty) probably decreases the need for transfusions. Additionally, treatment with intravenous iron alone (268 of 1000; RR 0.74, 95% CI 0.43 to 1.28; low certainty) and with oral iron alone (333 of 1000; RR 0.92, 95% CI 0.54 to 1.57; low certainty) may decrease or increase the need for transfusions. Overall mortality We estimated that 347 of 1000 participants without treatment for anaemia died overall. Low-certainty evidence from NMA (71 trials; 21,576 participants) suggests that treatment with ESA and intravenous iron (507 of 1000; RR 1.46, 95% CI 0.87 to 2.43) or oral iron (482 of 1000; RR 1.39, 95% CI 0.60 to 3.22) and intravenous iron alone (521 of 1000; RR 1.50, 95% CI 0.63 to 3.56) or oral iron alone (534 of 1000; RR 1.54, 95% CI 0.66 to 3.56) may decrease or increase overall mortality. Treatment with ESA alone may lead to little or no difference in overall mortality (357 of 1000; RR 1.03, 95% CI 0.97 to 1.10; low certainty). Thromboembolic events We estimated that 36 of 1000 participants without treatment for anaemia developed thromboembolic events. Evidence from NMA (50 trials; 15,408 participants) suggests that treatment with ESA and intravenous iron (66 of 1000; RR 1.82, 95% CI 0.98 to 3.41; moderate certainty) probably slightly increases and with ESA alone (66 of 1000; RR 1.82, 95% CI 1.34 to 2.47; high certainty) slightly increases the number of thromboembolic events. None of the trials reported results on the other comparisons. Thrombocytopenia or haemorrhage We estimated that 76 of 1000 participants without treatment for anaemia developed thrombocytopenia/haemorrhage. Evidence from NMA (13 trials, 2744 participants) suggests that treatment with ESA alone probably leads to little or no difference in thrombocytopenia/haemorrhage (76 of 1000; RR 1.00, 95% CI 0.67 to 1.48; moderate certainty). None of the trials reported results on other comparisons. Hypertension We estimated that 10 of 1000 participants without treatment for anaemia developed hypertension. Evidence from NMA (24 trials; 8383 participants) suggests that treatment with ESA alone probably increases the number of hypertensions (29 of 1000; RR 2.93, 95% CI 1.19 to 7.25; moderate certainty). None of the trials reported results on the other comparisons. AUTHORS' CONCLUSIONS When considering ESAs with iron as prevention for anaemia, one has to balance between efficacy and safety. Results suggest that treatment with ESA and iron probably decreases number of blood transfusions, but may increase mortality and the number of thromboembolic events. For most outcomes the different comparisons within the network were not fully connected, so ranking of all treatments together was not possible. More head-to-head comparisons including all evaluated treatment combinations are needed to fill the gaps and prove results of this review.
Collapse
Affiliation(s)
- Anne Adams
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Benjamin Scheckel
- Institute of Health Economics and Clinical Epidemiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Anissa Habsaoui
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Madhuri Haque
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kathrin Kuhr
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Julia Bohlius
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Nicole Skoetz
- Cochrane Cancer, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
45
|
Chemotherapy-Induced Myelosuppression in Esophageal Cancer Patients: Risks and Suggestions for Its Management. Curr Med Sci 2022; 42:530-537. [DOI: 10.1007/s11596-022-2587-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/22/2021] [Indexed: 11/03/2022]
|
46
|
Tan J, Du S, Zang X, Ding K, Ginzburg Y, Chen H. The addition of oral iron improves chemotherapy-induced anemia in patients receiving erythropoiesis-stimulating agents. Int J Cancer 2022; 151:1555-1564. [PMID: 35639027 DOI: 10.1002/ijc.34142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/18/2022] [Indexed: 11/07/2022]
Abstract
Although many studies have shown that supplementation with iron and erythropoiesis-stimulating agents (ESA) is frequently used for managing chemotherapy-induced anemia (CIA), optimal combination therapy using these agents together to ameliorate anemia is not well characterized. To assess the effects of ESA combined with oral or intravenous (IV) iron on relieving CIA, PubMed, Cochrane Library, Embase, China National Knowledge Infrastructure (CNKI) were searched for articles. Data collected in the articles were meta-analyzed using RevMan 5.3 software with a random-effects model. Our comprehensive search yielded 1666 potentially relevant trials. A total of 41 trials randomizing 4200 patients with CIA fulfilled inclusion criteria, including 34 Chinese articles and 7 English articles. Meta-analysis showed that treatment with both ESA and iron more effectively improved CIA relative to iron supplementation alone, with increased hemoglobin, hematocrit, red blood cell count and haematopoietic response rate. Subgroup analyses revealed iron administration, both oral and IV iron, improved anemia in ESA-treated cancer patients with CIA. Our analysis demonstrates that iron supplementation combined with ESA more effectively ameliorates CIA relative to iron supplementation alone, without regard to whether IV or oral iron was used. Together, our findings may contribute to the clinical treatment of CIA using iron therapy with or without ESA. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jingyong Tan
- Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Sitong Du
- Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Xueyan Zang
- Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Kaiyue Ding
- Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yelena Ginzburg
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Huiyong Chen
- Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
47
|
Xu XY, Jiang XM, Xu Q, Xu H, Luo JH, Yao C, Ding LY, Zhu SQ. Skeletal Muscle Change During Neoadjuvant Therapy and Its Impact on Prognosis in Patients With Gastrointestinal Cancers: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:892935. [PMID: 35692760 PMCID: PMC9186070 DOI: 10.3389/fonc.2022.892935] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundGastrointestinal cancers are the most common malignant tumors worldwide. As the improvement of survival by surgical resection alone for cancers is close to the bottleneck, recent neoadjuvant therapy has been emphasized and applied in the treatment. Despite the advantage on improving the prognosis, some studies have reported neoadjuvant therapy could reduce skeletal muscle and therefore affect postoperative outcomes. However, the conclusions are still controversial.MethodsPubMed, CINAHL, Embase, and Cochrane Library were searched from inception to September 2, 2021. The inclusion criteria were observational studies, published in English, of individuals aged ≥18 years who underwent neoadjuvant therapy with gastrointestinal cancers and were assessed skeletal muscle mass before and after neoadjuvant therapy, with sufficient data on skeletal muscle change or the association with clinical outcomes. Meta-analysis was conducted by using the STATA 12.0 package when more than two studies reported the same outcome.ResultsA total of 268 articles were identified, and 19 studies (1,954 patients) were included in the review. The fixed effects model showed that the risk of sarcopenia increased 22% after receiving neoadjuvant therapy (HR=1.22, 95% CI 1.14, 1.31, Z=4.286, P<0.001). In the random effects model, neoadjuvant therapy was associated with skeletal muscle loss, with a standardized mean difference of -0.20 (95% CI -0.31, -0.09, Z=3.49, P<0.001) and a significant heterogeneity (I2 =62.2%, P<0.001). Multiple meta regression indicated that population, neoadjuvant therapy type, and measuring tool were the potential sources of heterogeneity. The funnel plot revealed that there was no high publication bias in these studies (Begg’s test, P=0.544) and the sensitivity analysis showed stable results when separately excluding studies. For the postoperative outcomes, the results revealed that muscle loss during neoadjuvant therapy was significantly related to overall survival (HR=2,08, 95% CI =1.47, 2.95, Z=4.12, P<0.001, I2 = 0.0%), but not related to disease-free survival and other short-term outcomes.ConclusionsThis systematic review and meta-analysis revealed that skeletal muscle decreased significantly during neoadjuvant therapy in patients with gastrointestinal cancers and skeletal muscle loss was strongly associated with worse overall survival. More high-quality studies are needed to update and valid these conclusions in a more specific or stratified way.Systematic Review Registration[https://www.crd.york.ac.uk/PROSPERO/], identifier PROSPERO (CRD42021292118)
Collapse
Affiliation(s)
- Xin-Yi Xu
- Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Xiao-Man Jiang
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Qin Xu
- School of Nursing, Nanjing Medical University, Nanjing, China
- *Correspondence: Qin Xu,
| | - Hao Xu
- Department of Gastric Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin-Hua Luo
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui Yao
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling-Yu Ding
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Shu-Qin Zhu
- School of Nursing, Nanjing Medical University, Nanjing, China
| |
Collapse
|
48
|
Basak T, Kanwar RK. Iron imbalance in cancer: Intersection of deficiency and overload. Cancer Med 2022; 11:3837-3853. [PMID: 35460205 PMCID: PMC9582687 DOI: 10.1002/cam4.4761] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/09/2022] [Accepted: 02/25/2022] [Indexed: 12/19/2022] Open
Abstract
Iron, an essential trace element, plays a complex role in tumour biology. While iron causes cancer clearance through toxic free radical generation, iron‐induced free radical flux also acts as a cancer promoter. These fates majorly guided through cellular response towards pro‐oxidant and antioxidant settings in a tumour microenvironment, designate iron‐induced oxidative stress as a common yet paradoxical factor in pro‐tumorigenesis as well as anti‐tumorigenesis, posing a challenge to laying down iron thresholds favouring tumour clearance. Additionally, complexity of iron's association with carcinogenesis has been extended to iron‐induced ROS's involvement in states of both iron deficiency and overload, conditions identified as comparable, inevitable and significant coexisting contributors as well as outcomes in chronic infections and tumorigenesis. Besides, iron overload may also develop as an unwanted outcome in certain cancer patients, as a result of symptomatic anaemia treatment owed to irrational iron‐restoration therapies without a prior knowledge of body's iron status with both conditions synergistically acting towards tumour aggravation. The co‐play of iron deficiency and overload along with iron's pro‐tumour and antitumour roles with intersecting mechanisms, thus presents an unpredictable regulatory response loop in a state of malignancy. The relevance of iron's thresholds beyond which it proves to be beneficial against tumorigenesis hence becomes questionable. These factors pose a challenge, over establishing if iron chelation or iron flooding acts as a better approach towards antitumour therapies. This review presents a critical picture of multiple contrasting features of iron's behaviour in cancer, leading towards two conditions lying at opposite ends of a spectrum: iron deficiency and overload in chronic disease conditions including cancer, hence, validating the critical significance of diagnosis of patients' iron status prior to opting for subsequent therapies.
Collapse
Affiliation(s)
- Tulika Basak
- Institute for Innovation in Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Faculty of Health, Deakin University, Geelong, Victoria, Australia
| | - Rupinder Kaur Kanwar
- Institute for Innovation in Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Faculty of Health, Deakin University, Geelong, Victoria, Australia.,Department of Translational Medicine Centre, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, India
| |
Collapse
|
49
|
Mestrum SGC, Vanblarcum RBY, Drent RJM, Boonen BT, van Hemert WLW, Ramaekers FCS, Hopman AHN, Leers MPG. Proliferative and anti‐apoptotic fractions in maturing hematopoietic cell lineages and their role in homeostasis of normal bone marrow. Cytometry A 2022; 101:552-563. [PMID: 35429122 PMCID: PMC9540078 DOI: 10.1002/cyto.a.24558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/04/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022]
Abstract
Recent developments in clinical flow cytometry allow the simultaneous assessment of proliferative and anti‐apoptotic activity in the different hematopoietic cell lineages and during their maturation process. This can further advance the flow cytometric diagnosis of myeloid malignancies. In this study we established indicative reference values for the Ki‐67 proliferation index and Bcl‐2 anti‐apoptotic index in blast cells, as well as maturing erythroid, myeloid, and monocytic cells from normal bone marrow (BM). Furthermore, the cell fractions co‐expressing both proliferation and anti‐apoptotic markers were quantified. Fifty BM aspirates from femoral heads of patients undergoing hip replacement were included in this study. Ten‐color/twelve‐parameter flow cytometry in combination with a software‐based maturation tool was used for immunophenotypic analysis of Ki‐67 and Bcl‐2 positive fractions during the erythro‐, myelo‐, and monopoiesis. Indicative reference values for the Ki‐67 and Bcl‐2 positive fractions were established for different relevant hematopoietic cell populations in healthy BM. Ki‐67 and Bcl‐2 were equally expressed in the total CD34 positive blast cell compartment and 30% of Ki‐67 positive blast cells also showed Bcl‐2 positivity. The Ki‐67 and Bcl‐2 positive fractions were highest in the more immature erythroid, myeloid and monocytic cells. Both fractions then gradually declined during the subsequent maturation phases of these cell lineages. We present a novel application of an earlier developed assay that allows the simultaneous determination of the Ki‐67 proliferative and Bcl‐2 anti‐apoptotic indices in maturing hematopoietic cell populations of the BM. Their differential expression levels during the maturation process were in accordance with the demand and lifespan of these cell populations. The indicative reference values established in this study can act as a baseline for further cell biological and biomedical studies involving hematological malignancies.
Collapse
Affiliation(s)
- Stefan G. C. Mestrum
- Department of Molecular Cell Biology, GROW‐School for Oncology and Developmental Biology Maastricht University Medical Center Maastricht The Netherlands
- Department of Clinical Chemistry & Hematology Zuyderland Medical Center Sittard‐Geleen The Netherlands
| | - Roanalis B. Y. Vanblarcum
- Department of Clinical Chemistry & Hematology Zuyderland Medical Center Sittard‐Geleen The Netherlands
| | - Roosmarie J. M. Drent
- Department of Clinical Chemistry & Hematology Zuyderland Medical Center Sittard‐Geleen The Netherlands
| | - Bert T. Boonen
- Department of Orthopedic Surgery Zuyderland Medical Center Heerlen The Netherlands
| | | | - Frans C. S. Ramaekers
- Department of Molecular Cell Biology, GROW‐School for Oncology and Developmental Biology Maastricht University Medical Center Maastricht The Netherlands
- Nordic‐MUbio, Susteren The Netherlands
| | - Anton H. N. Hopman
- Department of Molecular Cell Biology, GROW‐School for Oncology and Developmental Biology Maastricht University Medical Center Maastricht The Netherlands
| | - Math P. G. Leers
- Department of Clinical Chemistry & Hematology Zuyderland Medical Center Sittard‐Geleen The Netherlands
| |
Collapse
|
50
|
Pagnotti GM, Trivedi T, Mohammad KS. Translational Strategies to Target Metastatic Bone Disease. Cells 2022; 11:1309. [PMID: 35455987 PMCID: PMC9030480 DOI: 10.3390/cells11081309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Metastatic bone disease is a common and devastating complication to cancer, confounding treatments and recovery efforts and presenting a significant barrier to de-escalating the adverse outcomes associated with disease progression. Despite significant advances in the field, bone metastases remain presently incurable and contribute heavily to cancer-associated morbidity and mortality. Mechanisms associated with metastatic bone disease perpetuation and paralleled disruption of bone remodeling are highlighted to convey how they provide the foundation for therapeutic targets to stem disease escalation. The focus of this review aims to describe the preclinical modeling and diagnostic evaluation of metastatic bone disease as well as discuss the range of therapeutic modalities used clinically and how they may impact skeletal tissue.
Collapse
Affiliation(s)
- Gabriel M. Pagnotti
- Department of Endocrine, Neoplasia and Hormonal Disorders, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (G.M.P.); (T.T.)
| | - Trupti Trivedi
- Department of Endocrine, Neoplasia and Hormonal Disorders, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (G.M.P.); (T.T.)
| | - Khalid S. Mohammad
- Department of Anatomy and Genetics, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|