1
|
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
|
2
|
Alyamany R, Alnughmush A, Alzahrani H, Alfayez M. Let It Grow: The Role of Growth Factors in Managing Chemotherapy-Induced Cytopenia. Curr Oncol 2024; 31:8094-8109. [PMID: 39727719 DOI: 10.3390/curroncol31120596] [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: 11/05/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
Chemotherapy-induced cytopenia (CIC) is characterized by neutropenia, anemia, and thrombocytopenia, which are common and serious complications in cancer treatment. These conditions affect approximately 60% of patients undergoing chemotherapy and can significantly impact quality of life, treatment continuity, and overall survival. The use of growth factors, including granulocyte colony-stimulating factors (GCSFs), erythropoietin-stimulating agents (ESAs), and thrombopoietin receptor agonists (TPO-RAs), has emerged as a promising strategy for managing CIC. However, the use of these growth factors must be approached with caution. This review provides an overview of the mechanisms, efficacy, and safety of growth factors in the management of CIC. Additionally, we discuss predictive markers for treatment response, potential risks, and highlight areas for future research.
Collapse
Affiliation(s)
- Ruah Alyamany
- Department of Hematology, Stem Cell Transplant and Cellular Therapy, Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Ahmed Alnughmush
- Department of Hematology, Stem Cell Transplant and Cellular Therapy, Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Hazzaa Alzahrani
- Department of Hematology, Stem Cell Transplant and Cellular Therapy, Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Mansour Alfayez
- Department of Hematology, Stem Cell Transplant and Cellular Therapy, Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
3
|
Wu W, Miao L, Zhao L, Zhu Y, Mao J, Cai Z, Ji Y, Wang L, Wang Y, Jia T. Prognostic value of lactate dehydrogenase, serum albumin and the lactate dehydrogenase/albumin ratio in patients with diffuse large B-cell lymphoma. Hematology 2024; 29:2293514. [PMID: 38108323 DOI: 10.1080/16078454.2023.2293514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVE To investigate the prognostic value of lactate dehydrogenase (LDH), serum albumin (ALB) and the lactate dehydrogenase/albumin ratio (LAR) in diffuse large B-cell lymphoma (DLBCL) before primary treatment. METHODS The clinical data of 212 primary adult DLBCL patients admitted to the First People's Hospital of Lianyungang from January 2017 to December 2022 were analyzed retrospectively. The optimal cutoff values of LDH, ALB, and LAR were determined using ROC curves. Survival curves of LDH, ALB, and LAR were plotted and analyzed using the Cox regression model and Kaplan-Meier method with the log-rank test. RESULTS Among the 212 patients admitted, the study derived the optimal cutoff values for ALB, LDH, and LAR as 38, 301, and 6, respectively. The Kaplan-Meier method and log-rank test analysis indicated a significant association between lower ALB levels, elevated LDH levels, elevated LAR levels, and shorter overall survival (OS) and progression-free survival (PFS) (P < 0.05). Additionally, the critical values of ALB and LDH were grouped into three categories. The differences in OS and PFS among these three groups were statistically significant (P < 0.05). Cox multifactorial analysis revealed that the LAR was an independent factor influencing the prognosis of OS and PFS, with a higher prognostic value than LDH and ALB alone. CONCLUSION Decreased ALB levels and elevated LDH and LAR levels at the time of initial diagnosis are indicative of a poor prognosis in DLBCL patients. Furthermore, the study highlighted that the LAR has a higher prognostic value than LDH and ALB alone.
Collapse
Affiliation(s)
- Wenke Wu
- Jinzhou Medical University, Jinzhou, People's Republic of China
- Department of Hematology, Postgraduate Training Base of the Lian Yungang First People's Hospital of Jinzhou Medical University, Lianyungang, People's Republic of China
| | - Lei Miao
- Department of Hematology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Lidong Zhao
- Department of Hematology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Yuanxin Zhu
- Department of Hematology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Jianping Mao
- Department of Hematology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Zhimei Cai
- Department of Hematology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Yajun Ji
- Department of Oncology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Lei Wang
- Department of Oncology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Ying Wang
- Department of Hematology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | - Tao Jia
- Department of Hematology, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| |
Collapse
|
4
|
Rioja P, Rey-Cardenas M, De Velasco G. Targeting HIF-2α and anemia: A therapeutic breakthrough for clear-cell renal cell carcinoma. Cancer Treat Rev 2024; 129:102801. [PMID: 39032449 DOI: 10.1016/j.ctrv.2024.102801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Renal cell carcinoma (RCC) is a heterogenous disease which the incidence is increasing worldwide. The identification and understanding of the role of the Von Hipple Lindau (VHP) in regulating the hypoxia-inducible factor signaling pathway has revolutionized the treatment of this disease. Belzutifan is an oral hypoxia-inducible factor (HIF)-2α inhibitor, which has demonstrated efficacy in treating von Hippel-Lindau (VHL) disease and for the treatment of adults with RCC who experienced disease progression after PD-1/PD-L1- and VEGFR-targeted therapies. One of the most common adverse effect of this drug is anemia; however, it is treatment is not well known. This review summarizes role of the VHL-HIF pathway in ccRCC aroused the interest of targeting HIF activity, the history of belzutifan development and their relationship to anemia as well as propose a management algorithm.
Collapse
Affiliation(s)
- Patricia Rioja
- Department of Medical Oncology, National Institute of Neoplastic Diseases, Lima, Peru.
| | - M Rey-Cardenas
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Guillermo De Velasco
- Department of Medical Oncology, University Hospital 12 de Octubre, Instituto de investigación (imas12), Madrid, Spain
| |
Collapse
|
5
|
Martins-Branco D, Kassapian M, Debien V, Caparica R, Eiger D, Dafni U, Andriakopoulou C, El-Abed S, Ellard SL, Izquierdo M, Vicente M, Chumsri S, Piccart-Gebhart M, Moreno-Aspitia A, Knop AS, Lombard J, de Azambuja E. The impact of erythropoiesis-stimulating agents administration concomitantly with adjuvant anti-HER2 treatments on the outcomes of patients with early breast cancer: a sub-analysis of the ALTTO study. Breast Cancer Res Treat 2024; 203:497-509. [PMID: 37938495 PMCID: PMC11052564 DOI: 10.1007/s10549-023-07159-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/05/2023] [Indexed: 11/09/2023]
Abstract
PURPOSE To assess whether erythropoiesis-stimulating agents (ESA) administration impacts the outcomes of patients with HER2-positive early breast cancer (EBC). METHODS ALTTO (NCT00490139) patients were categorized by ESA use during adjuvant anti-HER2 treatment. Disease-free-survival (DFS), overall survival (OS), and time-to-distant recurrence (TTDR) were analyzed by ESA administration, with subgroup analyses according to prognostic factors. Log-rank tests and Cox modeling were performed. Adverse events (AEs) of ESA-interest were compared. RESULTS Among 8381 patients recruited in ALTTO, 123 (1.5%) received ESA concomitantly with study treatment. The median age of patients receiving ESA was 54 years, 39.0% premenopausal, most had tumor size > 2 cm (56.9%), node-positive (58.5%), and positive estrogen receptor expression (61.8%). Median follow-up was shorter in the ESA group [6.1 years (IQR 5.3-7.0) vs. 6.9 years (6.0-7.1); p < 0.001]. There was no DFS difference by ESA administration (log-rank p = 0.70), with 3- and 7-year DFS of 89.2% (95% CI 81.8-93.8%) and 81.6% (71.4-88.5%) in ESA group vs. 88.3% (87.6-89.0%) and 80.0% (79.1-80.9%) in No-ESA group. In subgroup analyses, the interaction of ESA administration with menopausal status was statistically significant (unadjusted p = 0.024; stratified p = 0.033), favoring premenopausal women receiving ESA. We observed no significant association of ESA administration with OS (log-rank p = 0.57; 7-year OS in ESA 88.6% vs. 90.0% in non-ESA) or TTDR. ESA-interest AEs were experienced by eight (6.5%) patients receiving ESA and 417 (5.1%) in the No-ESA group (p = 0.41). CONCLUSION ESA administration to patients receiving adjuvant anti-HER2 treatment for HER2-positive EBC was safe and not associated with a negative impact on survival outcomes.
Collapse
Affiliation(s)
- Diogo Martins-Branco
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | | | - Véronique Debien
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Rafael Caparica
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Daniel Eiger
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Urania Dafni
- National and Kapodistrian University of Athens & Frontier Science Foundation-Hellas, Athens, Greece
| | | | | | | | | | - Malou Vicente
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Saranya Chumsri
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Martine Piccart-Gebhart
- Medical Oncology Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Ann Søegaard Knop
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Janine Lombard
- Calvary Mater Hospital & Australia and New Zealand Breast Cancer Trials Group (BCT-ANZ), Newcastle, Australia
| | - Evandro de Azambuja
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium.
| |
Collapse
|
6
|
Rastin F, Javid H, Oryani MA, Rezagholinejad N, Afshari AR, Karimi-Shahri M. Immunotherapy for colorectal cancer: Rational strategies and novel therapeutic progress. Int Immunopharmacol 2024; 126:111055. [PMID: 37992445 DOI: 10.1016/j.intimp.2023.111055] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 11/24/2023]
Abstract
There are increasing incidences and mortality rates for colorectal cancer in the world. It is common for chemotherapy and radiation given to patients with colorectal cancer to cause toxicities that limit their effectiveness and cause cancer cells to become resistant to these treatments. Additional targeted treatments are needed to improve patient's quality of life and outcomes. Immunotherapy has rapidly emerged as an incredibly exciting and promising avenue for cancer treatment in recent years. This innovative approach provides novel options for tackling solid tumors, effectively establishing itself as a new cornerstone in cancer treatment. Specifically, in the realm of colorectal cancer (CRC), there is great promise in developing new drugs that target immune checkpoints, offering a hopeful and potentially transformative solution. While immunotherapy of CRC has made significant advances, there are still obstacles and limitations. CRC patients have a poor response to treatment because of the immune-suppressing function of their tumor microenvironment (TME). In addition to blocking inhibitory immune checkpoints, checkpoint-blocking antibodies may also boost immune responses against tumors. The review summarizes recent advances in immune checkpoint inhibitors (ICIs) for CRC, including CTLA-4, PD-1, PD-L1, LAG-3, and TIM-3.
Collapse
Affiliation(s)
- Farangis Rastin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir-R Afshari
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
7
|
Fan X, Krzyzanski W, Wong RSM, Liu D, Yan X. Novel Combination of Erythropoietin and Romiplostim to Treat Chemotherapy-Induced Anemia and Thrombocytopenia via Pharmacodynamic Interaction on Hematopoietic Stem and Progenitor Cells. ACS Pharmacol Transl Sci 2023; 6:1884-1897. [PMID: 38093847 PMCID: PMC10714423 DOI: 10.1021/acsptsci.3c00194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/17/2024]
Abstract
Chemotherapy-induced anemia and thrombocytopenia (CIAT) in cancer patients are often caused by the damage of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. We have previously shown that romiplostim, a thrombopoietin receptor agonist that could stimulate the expansion of HSPCs, could synergize with recombinant human erythropoietin (rHuEPO) to promote erythropoiesis in addition to stimulating platelet production, whereas rHuEPO could influence the platelet count through stem cell competition. Therefore, we hypothesize that a combination of romiplostim with rHuEPO can alleviate CIAT simultaneously, while minimizing the risk of thrombosis. In this study, we demonstrated that rHuEPO and romiplostim exhibit no stimulatory effects on the growth and invasion of LA-7 cancer cells both in vitro and in vivo. Using a rat model with carboplatin-induced anemia and thrombocytopenia, we showed that the red blood cells and hemoglobin concentration recovered faster, and the secondary thrombocytopenia was alleviated in the rHuEPO and romiplostim combination therapy groups compared with the corresponding rHuEPO monotherapy groups. The rebound phenomenon of platelets was inhibited compared with the romiplostim monotherapy group. In vitro study further demonstrated that romiplostim expands HSPCs and synergizes with rHuEPO to promote erythropoiesis, while rHuEPO inhibited megakaryopoiesis. Furthermore, we developed a mechanism-based pharmacokinetic-pharmacodynamic model to quantify the effects of the two drugs. This study suggests that rHuEPO and romiplostim combination therapy can treat CIAT simultaneously in rats while minimizing the risk of thrombosis, indicating that combination therapy might be superior to monotherapy in the supportive therapy of cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Xiaoqing Fan
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, Hong Kong 999077, China SAR
| | - Wojciech Krzyzanski
- Department
of Pharmaceutical Sciences, The State University
of New York at Buffalo, Buffalo, New York 14068, United States
| | - Raymond S. M. Wong
- Division
of Hematology, Department of Medicine and Therapeutics, Faculty of
Medicine, The Chinese University of Hong
Kong, Shatin, Hong Kong 999077, China SAR
| | - Dongyang Liu
- Drug
Clinical Trial Center, Peking University
Third Hospital, Beijing 100191, China
| | - Xiaoyu Yan
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, Hong Kong 999077, China SAR
| |
Collapse
|
8
|
Zhang T, Liu W, Xu C. Correlation analysis of hemoglobin, albumin, lymphocyte, platelet score and platelet to albumin ratio and prognosis in patients with lung adenosquamous carcinoma. Front Oncol 2023; 13:1166802. [PMID: 37746281 PMCID: PMC10514357 DOI: 10.3389/fonc.2023.1166802] [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: 05/24/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Objective To investigate the effect of hemoglobin, albumin, lymphocytes, platelet (HALP) score and platelet to albumin ratio (PAR) on prognosis of patients with lung adenosquamous carcinoma (ASC) after surgery. Patients and methods A total of 52 patients diagnosed with ASC after surgical resection were collected from Nanjing Chest Hospital from 2012 to 2021, and their general clinical data, pathological data and laboratory indexes were collected. The changes of Alb and Plt levels before and after surgery, HALP scores (hemoglobin albumin lymphocytes/platelets), and postoperative PAR, PLR, NLR were retrospectively analyzed, and their influence on the prognosis of patients with ASC was investigated. The cut-off value of △Alb, △Plt, postoperative PAR, PLR and NLR were determined by the receiver operating characteristic (ROC) curve, the optimal cut-off value of HALP score before and after surgery was calculated by using X-tile software, and the clinicopathological characteristics were compared between the high PAR and low PAR groups and between high HALP score and low HALP score group to analyze the factors influencing the prognosis of patients with ASC. Univariate and multivariate Cox proportional regression analyses were used to assess independent risk factors affecting overall survival (OS) and disease-free survival (DFS) in patients with ASC. Kaplan-Meier method was used to evaluate the correlation between OS, DFS and PAR and HALP score. Results A critical value of PAR was 7.40×10^9 and an area under the curve (AUC) of 0.737 (95%CI: 0.597-0.876, P = 0.004). The best cut-off value of the preoperative HALP score was 24.3. Univariate Cox analysis showed that the cut margin (P = 0.013), the degree of differentiation (P = 0.021), N stage (P = 0.049), △Plt (P = 0.010), △Alb (P = 0.016), PAR (P = 0.003), NLR (P = 0.025), PLR (P = 0.029), preoperative HALP score (P = 0.000) and post-operative HALP score (P = 0.010) were all associated with postoperative OS in ASC patients. Cut margin (P = 0.029), the degree of differentiation (P = 0.045), maximum tumor diameter (P = 0.018), N stage (P = 0.035), △Plt (P = 0.007), △Alb (P = 0.007), PAR (P = 0.004), NLR (P = 0.041), PLR (P = 0.030), preoperative HALP score (P = 0.000), and postoperative HALP score (P = 0.011) were related to postoperative DFS in ASC patients. Multivariate analysis revealed that PAR (HR: 6.877, 95%CI: 1.817-26.038, P = 0.005), differentiation degree (HR: 0.059, 95%CI: 0.006-0.591, P = 0.016) and preoperative HALP score (HR: 0.224, 95%CI: 0.068-0.733, P = 0.013) had significant effect on OS. Tumor maximum diameter (HR: 3.442, 95%CI: 1.148-10.318, P = 0.027) and preoperative HALP score (HR: 0.268, 95%CI: 0.085-0.847, P = 0.025) had significant influence on DFS. Conclusion PAR and preoperative HALP score were potentially useful biomarkers for evaluating the outcome of patients with postoperative ASC. PAR, the degree of differentiation and preoperative HALP score were independent prognostic factors for postoperative OS in ASC patients. Maximum tumor diameter and preoperative HALP score were independent prognostic factors for postoperative DFS in ASC patients.
Collapse
Affiliation(s)
- Tiantian Zhang
- Department of Respiratory Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Clinical Center of Nanjing Respiratory Diseases and Imaging, Nanjing Chest Hospital, Nanjing, Jiangsu, China
| | - Wei Liu
- Department of Respiratory Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Clinical Center of Nanjing Respiratory Diseases and Imaging, Nanjing Chest Hospital, Nanjing, Jiangsu, China
| | - Chunhua Xu
- Department of Respiratory Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Clinical Center of Nanjing Respiratory Diseases and Imaging, Nanjing Chest Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Yuan T, Jia Q, Zhu B, Chen D, Long H. Synergistic immunotherapy targeting cancer-associated anemia: prospects of a combination strategy. Cell Commun Signal 2023; 21:117. [PMID: 37208766 DOI: 10.1186/s12964-023-01145-w] [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: 02/14/2023] [Accepted: 04/23/2023] [Indexed: 05/21/2023] Open
Abstract
Cancer-associated anemia promotes tumor progression, leads to poor quality of life in patients with cancer, and even obstructs the efficacy of immune checkpoint inhibitors therapy. However, the precise mechanism for cancer-associated anemia remains unknown and the feasible strategy to target cancer-associated anemia synergizing immunotherapy needs to be clarified. Here, we review the possible mechanisms of cancer-induced anemia regarding decreased erythropoiesis and increased erythrocyte destruction, and cancer treatment-induced anemia. Moreover, we summarize the current paradigm for cancer-associated anemia treatment. Finally, we propose some prospective paradigms to slow down cancer-associated anemia and synergistic the efficacy of immunotherapy. Video Abstract.
Collapse
Affiliation(s)
- Ting Yuan
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
10
|
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
|
11
|
Glaspy J, Gabrail NY, Locantore-Ford P, Lee T, Modelska K, Samal V, Henry DH. Open-label, Phase 2 study of roxadustat for the treatment of anemia in patients receiving chemotherapy for non-myeloid malignancies. Am J Hematol 2023; 98:703-711. [PMID: 36710399 DOI: 10.1002/ajh.26865] [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: 11/24/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
Anemia is a common side effect of myelosuppressive chemotherapy; however, chemotherapy-induced anemia (CIA) management options are suboptimal. We evaluated the efficacy and safety of roxadustat in this setting. This open-label Phase 2 study included patients with non-myeloid malignancies and CIA (hemoglobin [Hb] ≤10 g/dL) who had planned concurrent myelosuppressive chemotherapy for ≥8 additional weeks. Oral roxadustat was administered for ≤16 weeks (starting dose 2.0 or 2.5 mg/kg, then titrated every 4 weeks). The primary efficacy endpoint was mean maximum change in Hb within 16 weeks of baseline without red blood cell (RBC) transfusion. Patients were assigned to roxadustat 2.0 (n = 31) or 2.5 mg/kg (n = 61) starting doses, and 89 were assessed for efficacy. The mean (standard deviation) maximum Hb change from baseline without RBC transfusion was 2.4 (1.5) and 2.5 (1.5) g/dL in the roxadustat 2.0 and 2.5 mg/kg groups, respectively. Median (range) time to Hb increase of ≥2 g/dL was 71 (57-92) days. Twelve patients (14.5%) had RBC transfusions (Week 5 to the end of treatment). Roxadustat was efficacious regardless of tumor type and chemotherapy regimen. Deep vein thrombosis (DVT) and pulmonary embolism (PE) occurred in 14 (15.2%) and nine (9.8%) patients, respectively, and three had serious adverse events attributed to roxadustat in the opinion of the investigators (PE: n = 2 [2.2%]; DVT: n = 1 [1.1%]). Roxadustat increased Hb in patients with CIA regardless of tumor type and chemotherapy regimen. Adverse events were consistent with observations in patients with advanced-stage malignancies.
Collapse
Affiliation(s)
- John Glaspy
- Department of Medicine, University of California Los Angeles School of Medicine, Los Angeles, California, USA
| | - Nashat Y Gabrail
- Department of Oncology, Gabrail Cancer Center, Canton, Ohio, USA
| | | | - Tyson Lee
- Clinical Development, FibroGen, Inc., San Francisco, California, USA
| | | | - Vivek Samal
- Clinical Development, FibroGen, Inc., San Francisco, California, USA
| | - David H Henry
- Department of Medicine, Pennsylvania Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Li Z, Kwon SM, Li D, Li L, Peng X, Zhang J, Sueyoshi T, Raufman JP, Negishi M, Wang XW, Wang H. Human constitutive androstane receptor represses liver cancer development and hepatoma cell proliferation by inhibiting erythropoietin signaling. J Biol Chem 2022; 298:101885. [PMID: 35367211 PMCID: PMC9052153 DOI: 10.1016/j.jbc.2022.101885] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
The constitutive androstane receptor (CAR) is a nuclear receptor that plays a crucial role in regulating xenobiotic metabolism and detoxification, energy homeostasis, and cell proliferation by modulating the transcription of numerous target genes. CAR activation has been established as the mode of action by which phenobarbital-like nongenotoxic carcinogens promote liver tumor formation in rodents. This paradigm, however, appears to be unrelated to the function of human CAR (hCAR) in hepatocellular carcinoma (HCC), which remains poorly understood. Here, we show that hCAR expression is significantly lower in HCC than that in adjacent nontumor tissues and, importantly, reduced hCAR expression is associated with a worse HCC prognosis. We also show overexpression of hCAR in human hepatoma cells (HepG2 and Hep3B) profoundly suppressed cell proliferation, cell cycle progression, soft-agar colony formation, and the growth of xenografts in nude mice. RNA-Seq analysis revealed that the expression of erythropoietin (EPO), a pleiotropic growth factor, was markedly repressed by hCAR in hepatoma cells. Addition of recombinant EPO in HepG2 cells partially rescued hCAR-suppressed cell viability. Mechanistically, we showed that overexpressing hCAR repressed mitogenic EPO-EPO receptor signaling through dephosphorylation of signal transducer and activator of transcription 3, AKT, and extracellular signal-regulated kinase 1/2. Furthermore, we found that hCAR downregulates EPO expression by repressing the expression and activity of hepatocyte nuclear factor 4 alpha, a key transcription factor regulating EPO expression. Collectively, our results suggest that hCAR plays a tumor suppressive role in HCC development, which differs from that of rodent CAR and offers insight into the hCAR-hepatocyte nuclear factor 4 alpha-EPO axis in human liver tumorigenesis.
Collapse
Affiliation(s)
- Zhihui Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - So Mee Kwon
- Laboratory of Human Carcinogenesis, and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daochuan Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Xiwei Peng
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Junran Zhang
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Ohio, USA
| | - Tatsuya Sueyoshi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Office of Research and Development, Biomedical Laboratory Research and Development, VA Maryland Healthcare System, Baltimore, Maryland, USA
| | - Masahiko Negishi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA.
| |
Collapse
|
13
|
Rashidi A, Garimella PS, Al-Asaad A, Kharadjian T, Torres MN, Thakkar J. Anemia Management in the Cancer Patient With CKD and End-Stage Kidney Disease. Adv Chronic Kidney Dis 2022; 29:180-187.e1. [PMID: 35817525 DOI: 10.1053/j.ackd.2022.03.005] [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: 07/18/2021] [Revised: 02/18/2022] [Accepted: 03/10/2022] [Indexed: 11/11/2022]
Abstract
Anemia is a common medical problem among patients with cancer and chronic kidney disease (CKD). Although anemia in patients with CKD is often treated with iron and erythropoietin-stimulating agents, there are controversies with regard to the use of erythropoietin-stimulating agents in cancer patients. In this article, we review the treatment of anemia in patients with cancer and CKD, in addition to summarizing the current guidelines in treatment of anemia in these patients.
Collapse
Affiliation(s)
- Arash Rashidi
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, OH.
| | - Pranav S Garimella
- Division of Nephrology and Hypertension, University of California San Diego, La Jolla, CA
| | - Abdullah Al-Asaad
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Talar Kharadjian
- Division of Nephrology and Hypertension, University of California San Diego, La Jolla, CA
| | - Mariela Navarro Torres
- Department of Medicine/Division of Nephrology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| | - Jyotsana Thakkar
- Department of Medicine/Division of Nephrology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| |
Collapse
|
14
|
Burkert FR, Lanser L, Bellmann-Weiler R, Weiss G. Coronavirus Disease 2019: Clinics, Treatment, and Prevention. Front Microbiol 2021; 12:761887. [PMID: 34858373 PMCID: PMC8631905 DOI: 10.3389/fmicb.2021.761887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by a novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), emerged at the end of 2019 in China and affected the entire world population, either by infection and its health consequences, or by restrictions in daily life as a consequence of hygiene measures and containment strategies. As of September 2021, more than 231,000.000 infections and 4,740.000 deaths due to COVID-19 have been reported. The infections present with varied clinical symptoms and severity, ranging from asymptomatic course to fatal outcome. Several risk factors for a severe course of the disease have been identified, the most important being age, gender, comorbidities, lifestyle, and genetics. While most patients recover within several weeks, some report persistent symptoms restricting their daily lives and activities, termed as post-COVID. Over the past 18months, we have acquired significant knowledge as reflected by an almost uncountable number of publications on the nature of the underlying virus and its evolution, host responses to infection, modes of transmission, and different clinical presentations of the disease. Along this line, new diagnostic tests and algorithms have been developed paralleled by the search for and clinical evaluation of specific treatments for the different stages of the disease. In addition, preventive non-pharmacological measures have been implemented to control the spread of infection in the community. While an effective antiviral therapy is not yet available, numerous vaccines including novel vaccine technologies have been developed, which show high protection from infection and specifically from a severe course or death from COVID-19. In this review, we tried to provide an up-to-date schematic of COVID-19, including aspects of epidemiology, virology, clinical presentation, diagnostics, therapy, and prevention.
Collapse
Affiliation(s)
- Francesco Robert Burkert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| | - Lukas Lanser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
15
|
Avancini A, Belluomini L, Tregnago D, Trestini I, Milella M, Lanza M, Pilotto S. Exercise and anemia in cancer patients: could make the difference? Expert Rev Hematol 2021; 14:979-985. [PMID: 34784852 DOI: 10.1080/17474086.2021.2007764] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Anemia is one of the most common hematological manifestations occurring in cancer patients, related to a poorer prognosis. Among supportive care in cancer, exercise is emerging as a crucial tool demonstrating to reduce mortality risk, as well as to counteract several diseases and treatment-related adverse events. Nevertheless, the effect of exercise on anemia in cancer is still a relatively unexplored area. AREA COVERED Six studies investigated the impact of exercise on anemia trajectory in cancer patients, with intriguing results. The potential mechanisms by which exercise may modulate the blood system are heterogenous, including bone marrow stimulation, improvements in the hematopoietic microenvironment, and control of both inflammation and hormones levels. Although training and/or living at high altitudes is a recognized method to increase erythropoiesis, the effect of a hypoxic external environment on tumor progression should be clarified before considering this strategy in cancer patients. EXPERT OPINION Although the available investigations about exercise in anemic patients with cancer are few, exercise emerges as an important supportive oncological care capable of assisting patients maintain their autonomy. Our special report offers several suggestions to address future research in this field and determine the real contribution of an active lifestyle on this condition.
Collapse
Affiliation(s)
- Alice Avancini
- Medical Oncology, Department of Medicine, University of Verona Hospital Trust, 37134, Verona, Italy
| | - Lorenzo Belluomini
- Medical Oncology, Department of Medicine, University of Verona Hospital Trust, 37134, Verona, Italy
| | - Daniela Tregnago
- Medical Oncology, Department of Medicine, University of Verona Hospital Trust, 37134, Verona, Italy
| | - Ilaria Trestini
- Medical Oncology, Department of Medicine, University of Verona Hospital Trust, 37134, Verona, Italy
| | - Michele Milella
- Medical Oncology, Department of Medicine, University of Verona Hospital Trust, 37134, Verona, Italy
| | - Massimo Lanza
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona Hospital Trust, 37134, Verona, Italy
| | - Sara Pilotto
- Medical Oncology, Department of Medicine, University of Verona Hospital Trust, 37134, Verona, Italy
| |
Collapse
|
16
|
Abstract
Despite increasing use of targeted therapies to treat cancer, anemia remains a common complication of cancer therapy. Physician concerns about the safety of intravenous (IV) iron products and erythropoiesis-stimulating agents (ESAs) have resulted in many patients with cancer receiving no or suboptimal anemia therapy. In this article, we present 4 patient cases that illustrate both common and complex clinical scenarios. We first present a review of erythropoiesis and then describe our approach to cancer-associated anemia by identifying the contributing causes before selecting specific treatments. We summarize clinical trial data affirming the safety and efficacy of currently available IV iron products used to treat cancer-associated anemia and illustrate how we use commonly available laboratory tests to assess iron status during routine patient management. We compare adverse event rates associated with IV iron vs red cell transfusion and discuss using first-line IV iron monotherapy to treat anemic patients with cancer, which decreases the need for ESAs. A possible mechanism behind ESA-induced tumor progression is discussed. Finally, we review the potential of novel therapies such as ascorbic acid, prolyl hydroxylase inhibitors, activin traps, hepcidin, and bone morphogenetic protein antagonists in treating cancer-associated anemia.
Collapse
|
17
|
De Bellis M, Girelli D, Ruzzenente A, Bagante F, Ziello R, Campagnaro T, Conci S, Nifosì F, Guglielmi A, Iacono C. Pancreatic resections in patients who refuse blood transfusions. The application of a perioperative protocol for a true bloodless surgery. Pancreatology 2020; 20:1550-1557. [PMID: 32950387 DOI: 10.1016/j.pan.2020.08.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/16/2020] [Accepted: 08/26/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The refusal of blood transfusions compels surgeons to face ethical and clinical issues. A single-institution experience with a dedicated perioperative blood management protocol was reviewed to assess feasibility and short-term outcomes of true bloodless pancreatic surgery. METHODS The institutional database was reviewed to identify patients who refused transfusion and were scheduled for elective pancreatic surgery from 2010 through 2018. A protocol to optimize the hemoglobin values by administration of drugs stimulating erythropoiesis was systematically used. RESULTS Perioperative outcomes of 32 Jehovah's Witnesses patients were included. Median age was 67 years (range, 31-77). Nineteen (59.4%) patients were treated with preoperative erythropoietin. Twenty-four (75%) patients underwent pylorus-preserving pancreaticoduodenectomy, 4 (12.5%) distal pancreatectomy (DP) with splenectomy, 3 (9.4%) spleen-preserving DP, and 1 (3.1%) total pancreatectomy. Median estimated blood loss and surgical duration were 400 mL (range, 100-1000) and 470 min (range, 290-595), respectively. Median preoperative hemoglobin was 13.9 g/dL (range, 11.7-15.8) while median postoperative nadir hemoglobin was 10.5 g/dL (range, 7.1-14.1). The most common histological diagnosis (n = 15, 46.9%) was pancreatic ductal adenocarcinoma. Clavien-Dindo grade I-II complications occurred in fourteen (43.8%) patients while one (3.1%) patient had a Clavien-Dindo grade IIIa complication wich was an abdominal collection that required percutaneous drainage. Six (18.8%) patients presented biochemical leak or postoperative pancreatic fistula grade B. Median hospital stay was 16 days (range, 8-54) with no patient requiring transfusion or re-operation and no 90-day mortality. CONCLUSIONS A multidisciplinary approach and specific perioperative management allowed performing pancreatic resections in patients who refused transfusion with good short-term outcomes.
Collapse
Affiliation(s)
- Mario De Bellis
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine, University of Verona, School of Medicine, Verona, Italy
| | - Andrea Ruzzenente
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Fabio Bagante
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Raffaele Ziello
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Tommaso Campagnaro
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Simone Conci
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Filippo Nifosì
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Alfredo Guglielmi
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy
| | - Calogero Iacono
- Department of Surgery, Division of General and Hepato-Pancreato-Biliary Surgery, University of Verona, School of Medicine, Verona, Italy.
| |
Collapse
|
18
|
Abstract
Anemia is a frequent complication of kidney disease. When severe, it causes symptoms that can be debilitating. The course of anemia tends to track the decline in kidney function, with prevalence increasing in more advanced disease. Although the most common cause is relative erythropoietin deficiency, other factors such as reduced iron availability contribute to the pathobiology. In this review, we use cases to explore the surprising complexity of decision-making in management of renal anemia.
Collapse
|
19
|
Sbrana A, Antonuzzo A, Brunello A, Petrelli F, Pronzato P, Tralongo A, Turrini M, Zoratto F, Danova M. Management of anemia in patients with cancer: 2019 Italian Association of Medical Oncology (AIOM) guidelines. TUMORI JOURNAL 2020; 106:337-345. [PMID: 32482149 DOI: 10.1177/0300891620921942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This article summarizes the latest Italian Medical Oncology Association (AIOM) guidelines on the management of cancer-related and chemotherapy-related anemia with a particular attention to the use of erythropoiesis-stimulating agents and iron supplementation.
Collapse
Affiliation(s)
- Andrea Sbrana
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Andrea Antonuzzo
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | - Fausto Petrelli
- Medical Oncology Unit, ASST Bergamo Ovest, Treviglio, Bergamo, Italy
| | - Paolo Pronzato
- Medical Oncology Unit, AOU San Martino IRCCS IST, Genova, Italy
| | - Antonino Tralongo
- Medical Oncology Unit, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Marianna Turrini
- Medical Oncology Unit, Ospedale del Valdarno, Santa Maria La Gruccia, Azienda USL Toscana Sud-Est, Montevarchi, Toscana, Italy
| | - Federica Zoratto
- Medical Oncology Unit, Ospedale S. Maria Goretti, Latina, Lazio, Italy
| | - Marco Danova
- Internal Medicine and Medical Oncology, ASST Pavia, Pavia, Lombardia, Italy
| |
Collapse
|
20
|
Li WH, Zhang JY, Liu WH, Chen XX. Role of the initial degree of anaemia and treatment model in the prognosis of gastric cancer patients treated by chemotherapy: a retrospective analysis. BMC Cancer 2020; 20:414. [PMID: 32404067 PMCID: PMC7222574 DOI: 10.1186/s12885-020-06881-7] [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: 06/06/2019] [Accepted: 04/21/2020] [Indexed: 11/30/2022] Open
Abstract
Background Anaemia is highly prevalent in gastric cancer (GC) patients. The role of initial haemoglobin levels in predicting the prognosis of GC patients treated by chemotherapy has not been well determined. Our present study aims to evaluate the relationship between the degree of anaemia and the overall survival (OS) and progression-free survival (PFS) of patients with GC. Methods Our retrospective study enrolled 598 patients who were treated with chemotherapy when the recurrent or metastatic GCs were unsuitable for surgical resection. Univariate and multivariate analyses were performed to identify risk factors that had the potential to affect patient prognosis. Additionally, the relationship between clinicopathological characteristics, including treatment method, and degree of cancer-related reduction in haemoglobin was further analysed. Results Our results revealed that patients with HBini level ≤ 80 g/L had a trend toward a shortened median OS and PFS (p = 0.009 and p = 0.049, respectively). Interestingly, we also found that HBdec ≥ 30 g/L was associated with a significantly shortened median OS and PFS (p = 0.039 and p = 0.001, respectively). Multivariate analysis showed that HBini levels ≤80 g/L could be used as an independent prognostic factor for recurrent and metastatic GC. More importantly, HBdec ≥ 30 g/L and treatment response were also significantly associated with OS and PFS. Furthermore, the degree of haemoglobin decrease was associated with chemotherapy including platinum and the number of chemotherapy cycles. Conclusion Our study concludes that the initial degree of anaemia and a decrease in haemoglobin of ≥30 g/L can serve as biomarkers to predict prognosis in recurrent or metastatic GC patients, while chemotherapy treatment rather than red blood cell (RBC) transfusion can improve their prognosis. Additionally, platinum should not be recommended for treating severely anaemic GC patients.
Collapse
Affiliation(s)
- Wen-Huan Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu RD, Jinan, 250021, Shandong, People's Republic of China.
| | - Ji-Yu Zhang
- Shandong Center for Diseases Control and Prevention, 16992 Jingshi RD, Jinan, 250014, Shandong, People's Republic of China
| | - Wen-Hui Liu
- School of Public Health, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Xian-Xian Chen
- Shandong Center for Diseases Control and Prevention, 16992 Jingshi RD, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
21
|
Locatelli F, Del Vecchio L, De Nicola L, Minutolo R. Are all erythropoiesis-stimulating agents created equal? Nephrol Dial Transplant 2020; 36:1369-1377. [PMID: 32206785 DOI: 10.1093/ndt/gfaa034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/29/2019] [Indexed: 12/17/2022] Open
Abstract
Erythropoiesis-stimulating agents (ESAs) are effective drugs to correct and maintain haemoglobin (Hb) levels, however, their use at doses to reach high Hb targets has been associated with an increased risk of cardiovascular adverse events, mortality and cancer. Presently used ESAs have a common mechanism of action but different pharmacokinetic and pharmacodynamic characteristics. Accordingly, the mode of activation of the erythropoietin (EPO) receptor can exert marked differences in downstream events. It is unknown whether the various ESA molecules have different efficacy/safety profiles. The relative mortality and morbidity risks associated with the use of different types of ESAs remains poorly evaluated. Recently an observational study and a randomized clinical trial provided conflicting results regarding this matter. However, these two studies displayed several differences in patient characteristics and ESA molecules used. More importantly, by definition, randomized clinical trials avoid bias by indication and suffer less from confounding factors. Therefore they bring a higher degree of evidence. The scenario becomes even more complex when considering the new class of ESAs, called prolyl-hydroxylase domain (PHD) inhibitors. They are oral drugs that mimic exposure to hypoxia and stabilize hypoxia-inducible factor α. They profoundly differ from presently used ESAs, as they have multiple targets of action, including the stimulation of endogenous EPO synthesis, direct mobilization/absorption of iron and a higher reduction of hepcidin. Accordingly, they have the potential to be more effective in inflamed patients with functional iron deficiency, i.e. the setting of patients who are at higher risk of cardiovascular events and mortality in response to present ESA use. As for ESAs, individual PHD inhibitors differ in molecular structure and degree of selectivity for the three main PHD isoforms; their efficacy and safety profiles may therefore be different from that of presently available ESAs.
Collapse
Affiliation(s)
- Francesco Locatelli
- Past Director of the Department of Nephrology and Dialysis, Alessandro Manzoni Hospital, ASST Lecco, Lecco, Italy
| | | | - Luca De Nicola
- Department of Scienze Mediche e Chirurgiche Avanzate, Division of Nephrology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Roberto Minutolo
- Department of Scienze Mediche e Chirurgiche Avanzate, Division of Nephrology, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
22
|
Pujol JL, Roch B. Is Darbepoietin Alfa Linked to Mortality During Non-Small Cell Lung Cancer Chemotherapy? J Thorac Oncol 2020; 15:159-162. [PMID: 32127181 DOI: 10.1016/j.jtho.2019.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 11/19/2022]
Affiliation(s)
- Jean-Louis Pujol
- Thoracic Oncology Unit, Université de Montpellier, Hôpital Universitaire de Montpellier, Hôpital Arnaud de Villeneuve, Montpellier, France; Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier, France.
| | - Benoit Roch
- Thoracic Oncology Unit, Université de Montpellier, Hôpital Universitaire de Montpellier, Hôpital Arnaud de Villeneuve, Montpellier, France; Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier, France
| |
Collapse
|
23
|
Kenar G, Köksoy EB, Ürün Y, Utkan G. Prevalence, etiology and risk factors of anemia in patients with newly diagnosed cancer. Support Care Cancer 2020; 28:5235-5242. [DOI: 10.1007/s00520-020-05336-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 02/02/2020] [Indexed: 12/13/2022]
|
24
|
Wu CJ, Sundararajan V, Sheu BC, Huang RYJ, Wei LH. Activation of STAT3 and STAT5 Signaling in Epithelial Ovarian Cancer Progression: Mechanism and Therapeutic Opportunity. Cancers (Basel) 2019; 12:cancers12010024. [PMID: 31861720 PMCID: PMC7017004 DOI: 10.3390/cancers12010024] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal of all gynecologic malignancies. Despite advances in surgical and chemotherapeutic options, most patients with advanced EOC have a relapse within three years of diagnosis. Unfortunately, recurrent disease is generally not curable. Recent advances in maintenance therapy with anti-angiogenic agents or Poly ADP-ribose polymerase (PARP) inhibitors provided a substantial benefit concerning progression-free survival among certain women with advanced EOC. However, effective treatment options remain limited in most recurrent cases. Therefore, validated novel molecular therapeutic targets remain urgently needed in the management of EOC. Signal transducer and activator of transcription-3 (STAT3) and STAT5 are aberrantly activated through tyrosine phosphorylation in a wide variety of cancer types, including EOC. Extrinsic tumor microenvironmental factors in EOC, such as inflammatory cytokines, growth factors, hormones, and oxidative stress, can activate STAT3 and STAT5 through different mechanisms. Persistently activated STAT3 and, to some extent, STAT5 increase EOC tumor cell proliferation, survival, self-renewal, angiogenesis, metastasis, and chemoresistance while suppressing anti-tumor immunity. By doing so, the STAT3 and STAT5 activation in EOC controls properties of both tumor cells and their microenvironment, driving multiple distinct functions during EOC progression. Clinically, increasing evidence indicates that the activation of the STAT3/STAT5 pathway has significant correlation with reduced survival of recurrent EOC, suggesting the importance of STAT3/STAT5 as potential therapeutic targets for cancer therapy. This review summarizes the distinct role of STAT3 and STAT5 activities in the progression of EOC and discusses the emerging therapies specifically targeting STAT3 and STAT5 signaling in this disease setting.
Collapse
Affiliation(s)
- Chin-Jui Wu
- Department of Obstetrics & Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; (C.-J.W.); (B.-C.S.)
| | - Vignesh Sundararajan
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, Singapore 117599, Singapore;
| | - Bor-Ching Sheu
- Department of Obstetrics & Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; (C.-J.W.); (B.-C.S.)
| | - Ruby Yun-Ju Huang
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore 119077, Singapore;
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Lin-Hung Wei
- Department of Obstetrics & Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; (C.-J.W.); (B.-C.S.)
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 71570); Fax: +886-2-2311-4965
| |
Collapse
|
25
|
Jing W, Guo X, Wang G, Bi Y, Han L, Zhu Q, Qiu C, Tanaka M, Zhao Y. Breast cancer cells promote CD169 + macrophage-associated immunosuppression through JAK2-mediated PD-L1 upregulation on macrophages. Int Immunopharmacol 2019; 78:106012. [PMID: 31865052 DOI: 10.1016/j.intimp.2019.106012] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/27/2019] [Accepted: 10/27/2019] [Indexed: 01/10/2023]
Abstract
Macrophages are recognized as one of the major cell types in tumor microenvironment, and macrophage infiltration has been predominantly associated with poor prognosis among patients with breast cancer. Using the murine models of triple-negative breast cancer in CD169-DTR mice, we found that CD169+ macrophages support tumor growth and metastasis. CD169+ macrophage depletion resulted in increased accumulation of CD8+ T cells within tumor, and produced significant expansion of CD8+ T cells in circulation and spleen. In addition, we observed that CD169+ macrophage depletion alleviated tumor-induced splenomegaly in mice, but had no improvement in bone loss and repression of bone marrow erythropoiesis in tumor-bearing mice. Cancer cells and tumor associated macrophages exploit the upregulation of the immunosuppressive protein PD-L1 to subvert T cell-mediated immune surveillance. Within the tumor microenvironment, our understanding of the regulation of PD-L1 protein expression is limited. We showed that there was a 5-fold higher relative expression of PD-L1 on macrophages as compared with 4T1 tumor cells; coculture of macrophages with 4T1 cells augmented PD-L1 levels on macrophages, but did not upregulate the expression of PD-L1 on 4T1 cells. JAK2/STAT3 signaling pathway was activated in macrophages after coculture, and we further identified the JAK2 as a critical regulator of PD-L1 expression in macrophages during coculture with 4T1 cells. Collectively, our data reveal that breast cancer cells and CD169+ macrophages exhibit bidirectional interactions that play a critical role in tumor progression, and inhibition of JAK2 signaling pathway in CD169+ macrophages may be potential strategy to block tumor microenvironment-derived immune escape.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xing Guo
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ganyu Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yuxuan Bi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Lihui Han
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qingfen Zhu
- Shandong Institute for Food and Drug Control, Jinan, China.
| | - Chunhong Qiu
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Masato Tanaka
- Laboratory of Immune Regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Yunxue Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China; Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
26
|
Thavarajah S, Choi MJ. The Use of Erythropoiesis-Stimulating Agents in Patients With CKD and Cancer: A Clinical Approach. Am J Kidney Dis 2019; 74:667-674. [DOI: 10.1053/j.ajkd.2019.04.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 04/18/2019] [Indexed: 01/13/2023]
|
27
|
Abstract
Cancer and kidney disease are linked by causality and comorbidities. Observational data show an increased risk of malignancy as renal function declines. Erythropoietin stimulating agents (ESAs), which are the cornerstone therapy for anemia patients with chronic kidney disease and cancer, are associated with increased risks for cancer, cancer-related mortality, progression of disease, and thromboembolic events. This article examines the recently published guidelines for ESA use in cancer patients from the American Society of Clinical Oncology and American Society of Hematology and attempts to contextualize them to the care of patients with coexistent CKD, cancer, and anemia.
Collapse
Affiliation(s)
- Sheron Latcha
- Renal Division, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
28
|
Li JH, Luo JF, Jiang Y, Ma YJ, Ji YQ, Zhu GL, Zhou C, Chu HW, Zhang HD. Red Blood Cell Lifespan Shortening in Patients with Early-Stage Chronic Kidney Disease. Kidney Blood Press Res 2019; 44:1158-1165. [PMID: 31550724 DOI: 10.1159/000502525] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/02/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Although reduced red blood cell (RBC) lifespan has been reported to be a contributory factor to anemia in patients with end-stage chronic kidney disease (CKD), there are limited data regarding RBC lifespan in early-stage CKD. Serum erythropoietin (EPO) is considered a primary causative factor of renal anemia. The aims of this study were to compare the RBC lifespan, serum EPO levels, and other renal anemia indicators across CKD-stage groups of patients and to analyze the impacts of etiological factors on renal anemia. METHODS A cohort of 74 non-smoking patients with CKD were enrolled, including 15 in stage 1, 18 in stage 2, 15 in stage 3, 15 in stage 4, and 11 in stage 5. RBC lifespan was determined by CO breath tests. Potential correlations of hemoglobin (Hb) concentration with RBC lifespan, reticulocyte count (Ret), and levels of EPO, ferritin, folic acid, and vitamin B12 were analyzed. RESULTS CKD progression was associated with decreases in (Hb) and RBC lifespan. RBC lifespan durations in CKD stages 1-5 were 122 ± 50, 112 ± 26, 90 ± 32, 88 ± 28, and 60 ± 24 days, respectively. RBC lifespan means for the stage 3, 4 and 5 groups were significantly shorter than those for the stage 1 and 2 groups. Serum EPO did not differ significantly between the CKD stage groups. (Hb) correlated directly with RBC lifespan (r = 0.372, p = 0.002) and Ret (r = 0.308, p = 0.011), but did not correlate with serum EPO, ferritin, folic acid, or vitamin B12 levels. CONCLUSIONS Reduced RBC lifespan in early-stage CKD, demonstrated in this study, suggests that increased RBC destruction may play a more important etiological role in renal anemia than other indicators in patients with CKD.
Collapse
Affiliation(s)
- Jiu-Hong Li
- Department of Nephrology, Nanshan Hospital, Guangdong Medical University, Shenzhen, China
| | - Jun-Feng Luo
- Guangdong Breath Test Engineering and Technology Research Center, Shenzhen University, Shenzhen, China
| | - Ying Jiang
- Department of Nephrology, Nanshan Hospital, Guangdong Medical University, Shenzhen, China
| | - Yong-Jian Ma
- Guangdong Breath Test Engineering and Technology Research Center, Shenzhen University, Shenzhen, China
| | - Yong-Qiang Ji
- Institute of Seekya Breath Test Technology, Shenzhen, China
| | - Guo-Liang Zhu
- Institute of Seekya Breath Test Technology, Shenzhen, China
| | - Cong Zhou
- Institute of Seekya Breath Test Technology, Shenzhen, China
| | - Hong-Wei Chu
- Guangdong Breath Test Engineering and Technology Research Center, Shenzhen University, Shenzhen, China
| | - Hou-De Zhang
- Department of Gastroenterology, Nanshan Hospital, Guangdong Medical University, Shenzhen, China,
| |
Collapse
|
29
|
Elliott S. Impact of Inadequate Methods and Data Analysis on Reproducibility. J Pharm Sci 2019; 109:1211-1219. [PMID: 31351867 DOI: 10.1016/j.xphs.2019.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/13/2019] [Accepted: 07/17/2019] [Indexed: 11/29/2022]
Abstract
Failure to reproduce results of articles is recognized, but the causes, and therefore solutions, are not. One possibility is that deficits in quality of the work result in varying or inconclusive results. Erythropoiesis-stimulating agents have been used to treat anemia in patients with cancer, but there are concerns that erythropoiesis-stimulating agents might stimulate Epo receptors on tumor cells (Epo receptor-cancer hypothesis). Articles have been published on the topic, but the data and conclusions conflict, making them suitable for examination of a relationship between quality and reproducibility. Comprehensive literature searches were performed, and 280 relevant articles were identified. Numerous conflicts between and within these articles were apparent. The incidence of faults in quality parameters was high, including absence of adequate controls (90% of articles), inadequate validation of reagents and methods (87% of articles), and inadequate or improper statistical methods (84% of articles) with questionable interpretation of the data (81% of articles). This resulted in false-positive/negative data that varied with the reagents and methods used. The low quality of evidence may explain the poor reproducibility of Epo receptor-cancer articles.
Collapse
|
30
|
Choi MJ, Yee J. Erythropoiesis-Stimulating Agents and Cancer: Myth or Truth. Adv Chronic Kidney Dis 2019; 26:221-224. [PMID: 31477251 DOI: 10.1053/j.ackd.2019.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 11/11/2022]
|
31
|
Liu Y, Bai YP, Zhou ZF, Jiang CR, Xu Z, Fan XX. Preoperative anemia as a prognostic factor in patients with lung cancer: a systematic review and meta-analysis of epidemiological studies. J Cancer 2019; 10:2047-2056. [PMID: 31205565 PMCID: PMC6548169 DOI: 10.7150/jca.29410] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 04/23/2019] [Indexed: 12/21/2022] Open
Abstract
The evidence of current epidemiological studies investigating the relationship between preoperative anemia and progression of lung cancer (LC) patients remains controversial. The PubMed, EMBASE, and Web of Science databases were comprehensively searched by two independent authors to identify related epidemiological studies from inception through January 31, 2019. Similarly, two researchers separately extracted data and any differences were resolved by discussion. Summarized hazard ratios (HRs) and 95% confidence intervals (CIs) were summarized with inverse variance weighted random effects meta-analysis. Heterogeneity among studies was assessed with the I² statistic. Twenty-two studies were included in this meta-analysis. As compared with LC patients without anemia, those with pre-operative anemia were at a 1.6-fold greater risk of death (summarized HR = 1.58; 95% CI = 1.44-1.75), with moderate heterogeneity (I2 = 53.1%). Funnel plot and statistical analyses showed no evidence of publication bias. Associations between pre-operative anemia and OS were broadly consistent across numerous subgroups analyses stratified by the study design, geographic location, number of cases, tumor, node, and metastasis (TNM) stage, histology, quality, and adjustment for potential confounders (age, sex, body mass index, TNM stage, histology, performance status, surgery, blood transfusion, and systemic inflammatory response markers). Similar patterns were observed in the sensitivity analyses. The results of meta-regression analysis suggested no evidence of significant heterogeneity between subgroups. In conclusion, pre-operative anemia was associated with poorer overall survival among LC patients.
Collapse
Affiliation(s)
- Yang Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yun-Peng Bai
- Department of Thoracic Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zi-Fang Zhou
- Department of Thoracic Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chang-Rui Jiang
- Department of Thoracic Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhe Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Xi Fan
- Department of Thoracic Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Sotatercept and luspatercept are recombinant soluble activin type-II receptor-IgG-Fc fusion proteins that are tested in clinical trials for the treatment of various types of anemias, including renal anemia. The mechanism of the action of the novel drugs is incompletely understood, but it seems to be based on the inactivation of soluble proteins of the transforming growth factor-ß (TGFß) family. This review considers pros and cons of the clinical use of the drugs in reference to the current therapy with recombinant erythropoiesis-stimulating agents (ESAs). RECENT FINDINGS One or more activin type-II receptor (ActRII) ligands appear to inhibit erythroid precursors, for example growth and differentiation factor 11. Trapping of these ligands by the recombinant ActRII fusion proteins, sotatercept and luspatercept increases red blood cell numbers and hemoglobin levels in humans. Reportedly, the novel compounds were well tolerated in trials on healthy volunteers and patients suffering from anemia due to chronic kidney disease or malignancies. On approval, the drugs may prove particularly useful in patients suffering from ineffective erythropoiesis, such as in myelodysplastic syndrome, multiple myeloma or ß-thalassemia, where ESAs are of little use. Independent of their effect on erythropoiesis, ActRII ligand traps were found to exert beneficial effects on renal tissue in experimental animals. SUMMARY ESAs are likely to remain standard of care in renal anemia. There is a need for a better understanding of the effects of ActRII ligand traps on TGFß-like proteins. The novel drugs have not been approved for sale as therapeutics so far. Their long-term efficacy and safety still needs to be proven, particularly with respect to immunogenicity. Antifibrotic effects may be worthy to be investigated in humans.
Collapse
|
33
|
Pham TND, Ma W, Miller D, Kazakova L, Benchimol S. Erythropoietin inhibits chemotherapy-induced cell death and promotes a senescence-like state in leukemia cells. Cell Death Dis 2019; 10:22. [PMID: 30622244 PMCID: PMC6325163 DOI: 10.1038/s41419-018-1274-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/03/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022]
Abstract
There are conflicting reports on the adverse effects of erythropoietin (EPO) for the management of cancer-associated anemia. The recognition that erythropoietin receptors (EPORs) are expressed outside the erythroid lineage and concerns that erythropoiesis-stimulating agents (ESAs) may cause tumors to grow and increase the risk of venous thromboembolism have resulted in substantially fewer cancer patients receiving ESA therapy to manage myelosuppressive chemotherapy. In this study, we found that EPO suppresses p53-dependent apoptosis induced by genotoxic (daunorubicin, doxorubicin, and γ-radiation) and non-genotoxic (nutlin-3a) agents and induces a senescence-like state in myeloid leukemia cells. EPO interferes with stress-dependent Mdm2 downregulation and leads to the destabilization of p53 protein. EPO selectively modulates the expression of p53 target genes in response to DNA damage preventing the induction of a number of noncoding RNAs (ncRNAs) previously associated with p53-dependent apoptosis. EPO also enhances the expression of the cyclin-dependent kinase inhibitor p21WAF1 and promotes recruitment of p53 to the p21 promoter. In addition, EPO antagonizes Mcl-1 protein degradation in daunorubicin-treated cells. Hence, EPO signaling targets Mcl-1 expression and the p53-Mdm2 network to promote tumor cell survival.
Collapse
Affiliation(s)
| | - Weili Ma
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - David Miller
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Lidia Kazakova
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Samuel Benchimol
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
34
|
Red blood cell transfusion and its alternatives in oncologic surgery-A critical evaluation. Crit Rev Oncol Hematol 2018; 134:1-9. [PMID: 30771868 DOI: 10.1016/j.critrevonc.2018.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/14/2018] [Accepted: 11/29/2018] [Indexed: 01/28/2023] Open
Abstract
Although blood transfusions have been used for more than 100 years and their potential to save lives is indisputable, there is still limited data on medium- and long-term outcomes after hemotherapy. Until recently, red blood cell transfusions represented the most commonly employed treatment for cancer anemia. As transfusions have been related to worse patient outcome in oncologic surgery, preventive strategies and alternative treatment approaches in the perioperative setting are warranted. This review aims to evaluate the evidence concerning the impact of transfusion on the course of malignant diseases with a focus on oncologic surgery and to provide a bundle of measures to improve patient care. The perioperative period is pivotal in determining long-term cancer outcome. An increasingly recognized area for improvement during this highly sensitive period is the treatment of anemia for three main reasons: Firstly, anemia has been recognized as an independent predictor of poor prognosis in cancer patients. Secondly, anemia is largely undertreated. Thirdly and probably most importantly, anemia therapy relied and often still relies heavily on red blood cell (RBC) transfusions, which may be an often suboptimal stopgap treatment. Perioperative RBC transfusions should be kept to a minimum due to growing concerns regarding the associated risks, which this review tries to clarify by providing an update of recent literature. This review furthermore discusses treatments for anemia and provides best-practice approaches to improve perioperative management of oncology patients undergoing surgery.
Collapse
|
35
|
Aapro M, Beguin Y, Bokemeyer C, Dicato M, Gascón P, Glaspy J, Hofmann A, Link H, Littlewood T, Ludwig H, Österborg A, Pronzato P, Santini V, Schrijvers D, Stauder R, Jordan K, Herrstedt J. Management of anaemia and iron deficiency in patients with cancer: ESMO Clinical Practice Guidelines. Ann Oncol 2018; 29:iv96-iv110. [PMID: 29471514 DOI: 10.1093/annonc/mdx758] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Affiliation(s)
- M Aapro
- Genolier Cancer Centre, Clinique de Genolier, Genolier, Switzerland
| | - Y Beguin
- University of Liège, Liège
- CHU of Liège, Liège, Belgium
| | - C Bokemeyer
- Department of Oncology, Hematology and BMT with Section Pneumology, University of Hamburg, Hamburg, Germany
| | - M Dicato
- Hématologie-Oncologie, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - P Gascón
- Department of Haematology-Oncology, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - J Glaspy
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - A Hofmann
- Medical Society for Blood Management, Laxenburg, Austria
| | - H Link
- Klinik für Innere Medizin I, Westpfalz-Klinikum, Kaiserslautern, Germany
| | - T Littlewood
- Department of Haematology, John Radcliffe Hospital, Oxford, UK
| | - H Ludwig
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - A Österborg
- Karolinska Institute and Karolinska Hospital, Stockholm, Sweden
| | - P Pronzato
- Medica Oncology, IRCCS Asiana Pedaliter Universitaria San Martino - IST, Institutor Nazionale per la Ricercars sol Chancre, Genova
| | - V Santini
- Department of Experimental and Clinical Medicine, Haematology, University of Florence, Florence, Italy
| | - D Schrijvers
- Department of Medical Oncology, Ziekenhuisnetwerk Antwerpen, Antwerp, Belgium
| | - R Stauder
- Department of Internal Medicine V (Haematology and Oncology), Innsbruck Medical University, Innsbruck, Austria
| | - K Jordan
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - J Herrstedt
- Department of Oncology, Zealand University Hospital Roskilde, Roskilde
- University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Busti F, Marchi G, Ugolini S, Castagna A, Girelli D. Anemia and Iron Deficiency in Cancer Patients: Role of Iron Replacement Therapy. Pharmaceuticals (Basel) 2018; 11:E94. [PMID: 30274354 PMCID: PMC6315653 DOI: 10.3390/ph11040094] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022] Open
Abstract
Anemia in cancer patients is quite common, with remarkable negative impacts on quality of life and overall prognosis. The pathogenesis is complex and typically multifactorial, with iron deficiency (ID) often being a major and potentially treatable contributor. In turn, ID in cancer patients can be due to multiple concurring mechanisms, including bleeding (e.g., in gastrointestinal cancers or after surgery), malnutrition, medications, and hepcidin-driven iron sequestration into macrophages with subsequent iron-restricted erythropoiesis. Indeed, either absolute or functional iron deficiency (AID or FID) can occur. While for absolute ID there is a general consensus regarding the laboratory definition (that is ferritin levels <100 ng/mL ± transferrin saturation (TSAT) <20%), a shared definition of functional ID is still lacking. Current therapeutic options in cancer anemia include iron replacement, erythropoietic stimulating agents (ESAs), and blood transfusions. The latter should be kept to a minimum, because of concerns regarding risks, costs, and limited resources. Iron therapy has proved to be a valid approach to enhance efficacy of ESAs and to reduce transfusion need. Available guidelines focus mainly on patients with chemotherapy-associated anemia, and generally suggest intravenous (IV) iron when AID or FID is present. However, in the case of FID, the upper limit of ferritin in association with TSAT <20% at which iron should be prescribed is a matter of controversy, ranging up to 800 ng/mL. An increasingly recognized indication to IV iron in cancer patients is represented by preoperative anemia in elective oncologic surgery. In this setting, the primary goal of treatment is to decrease the need of blood transfusions in the perioperative period, rather than improving anemia-related symptoms as in chemotherapy-associated anemia. Protocols are mainly based on experiences of Patient Blood Management (PBM) in non-oncologic surgery, but no specific guidelines are available for oncologic surgery. Here we discuss some possible approaches to the management of ID in cancer patients in different clinical settings, based on current guidelines and recommendations, emphasizing the need for further research in the field.
Collapse
Affiliation(s)
- Fabiana Busti
- Department of Medicine, Section of Internal Medicine, University of Verona, and EuroBloodNet Referral Center for Iron Disorders, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico G.B. Rossi, 37134 Verona, Italy.
| | - Giacomo Marchi
- Department of Medicine, Section of Internal Medicine, University of Verona, and EuroBloodNet Referral Center for Iron Disorders, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico G.B. Rossi, 37134 Verona, Italy.
| | - Sara Ugolini
- Department of Medicine, Section of Internal Medicine, University of Verona, and EuroBloodNet Referral Center for Iron Disorders, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico G.B. Rossi, 37134 Verona, Italy.
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine, University of Verona, and EuroBloodNet Referral Center for Iron Disorders, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico G.B. Rossi, 37134 Verona, Italy.
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine, University of Verona, and EuroBloodNet Referral Center for Iron Disorders, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico G.B. Rossi, 37134 Verona, Italy.
| |
Collapse
|
37
|
Zhang YH, Lu Y, Lu H, Zhang MW, Zhou YM, Li XL, Lv P, Zhao XY. Pre-treatment hemoglobin levels are an independent prognostic factor in patients with non-small cell lung cancer. Mol Clin Oncol 2018; 9:44-49. [PMID: 29896399 DOI: 10.3892/mco.2018.1628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 05/09/2018] [Indexed: 12/19/2022] Open
Abstract
To date, few studies have reported the prognostic value of pre-treatment hemoglobin levels in patients with non-small cell lung cancer (NSCLC). In the present study, 416 patients with NSCLC were retrospectively reviewed. Univariate Cox proportional hazards regression analysis demonstrated that patients with normal pre-treatment hemoglobin (NPHb) levels had a greater chance of surviving for longer period, than did patients with low pre-treatment hemoglobin (LPHb) levels (HR, 2.05; 95% CI, 1.63-2.57; P<0.001). After adjustment for age, sex, tumor-node-metastasis stage, Karnofsky performance status, lung lobectomy, chemotherapy and radiotherapy, multivariate Cox proportional hazards regression analysis revealed that LPHb was an independent predictor for the poor prognosis of patients with NSCLC (HR, 1.86; 95% CI, 1.47-2.36; P<0.001). Estimation of the cumulative survival revealed that the overall survival of NPHb patients was significantly higher than that for LBHb patients (P<0.05), independent of whether the patients had received lung lobectomy or chemotherapy treatments. In conclusion, low pre-treatment hemoglobin levels were demonstrated to be an independent biomarker for poor prognosis in patients with NSCLC.
Collapse
Affiliation(s)
- Yue-Hua Zhang
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China.,Department of Oncology, The First Affiliated Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Yuquan Lu
- International Joint Research Laboratory for Cell Medical Engineering of Henan, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Hong Lu
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Meng-Wei Zhang
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Yue-Min Zhou
- International Joint Research Laboratory for Cell Medical Engineering of Henan, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China.,Center for Translational Medicine, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Xiang-Lei Li
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Pin Lv
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Xiao-Yan Zhao
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| |
Collapse
|
38
|
Pretreatment Hemoglobin Level Is an Independent Prognostic Factor in Patients with Lung Adenocarcinoma. Can Respir J 2018; 2018:6328127. [PMID: 29887927 PMCID: PMC5977049 DOI: 10.1155/2018/6328127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/06/2018] [Indexed: 12/17/2022] Open
Abstract
Background/Aim Few studies have reported the prognostic value of pretreatment hemoglobin levels in patients with lung adenocarcinoma (LA). In the present study, we retrospectively reviewed 306 LA patients for their prognosis associated with the pretreatment hemoglobin levels. Methods Person-years and case fatality rate (CFR) were calculated from May 2010 to June 2017. Hazard ratio (HR) and 95% confidence intervals (CIs) were estimated using the Cox proportional hazards regression analysis. Survival curves were generated using the Kaplan–Meier analysis. Results Patients with low pretreatment hemoglobin (LPHb) levels had a higher CFR than did patients with normal pretreatment hemoglobin (NPHb) levels (HR = 1.48, 95% CI = 1.06–2.08, and P=0.023). Overall survival of NPHb patients was significantly higher than that of LPHb patients (P < 0.05). Conclusion Low pretreatment hemoglobin level was demonstrated to be an independent biomarker for poor prognosis in patients with LA.
Collapse
|
39
|
Zhou Y, Xu H, Ding Y, Lu Q, Zou MH, Song P. AMPKα1 deletion in fibroblasts promotes tumorigenesis in athymic nude mice by p52-mediated elevation of erythropoietin and CDK2. Oncotarget 2018; 7:53654-53667. [PMID: 27449088 PMCID: PMC5288212 DOI: 10.18632/oncotarget.10687] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/07/2016] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis is essential for tumor development. Accumulating evidence suggests that adenosine monophosphate-activated protein kinase (AMPK), an energy sensor and redox modulator, is associated with cancer development. However, the effect of AMPK on tumor development is controversial, and whether AMPK affects tumor angiogenesis has not been resolved. We show that deletion of AMPKα1, but not AMPKα2, upregulates non-canonical nuclear factor kappa B2 (NF-κB2)/p52-mediated cyclin-dependent kinase 2 (CDK2), which is responsible for the anchorage-independent cell growth of immortalized mouse embryo fibroblasts (MEFs). Co-culture with AMPKα1 knockout MEFs (or their conditioned medium) enhances the migration and network formation of human microvascular endothelial cells, which is dependent on p52-upregulated erythropoietin (Epo). AMPKα1 deletion stimulates cellular proliferation of allograft MEFs, angiogenesis, and tumor development in athymic nu/nu mice, which is partly ameliorated by antibody-mediated Epo neutralization. Therefore, the AMPKα1-p52-Epo pathway may be involved in stromal fibroblast-mediated angiogenesis and tumorigenesis.
Collapse
Affiliation(s)
- Yanhong Zhou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA.,Key Laboratory of Hubei Province on Cardio-Cerebral Diseases, Hubei University of Science and Technology, Xianning, Hubei 437100, China
| | - Hairong Xu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA.,School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Qiulun Lu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
40
|
Cortinovis D, Beretta G, Piazza E, Luchena G, Aglione S, Bertolini A, Buzzoni R, Cabiddu M, Carnaghi C, Danova M, Farina G, Ferrari V, Frascaroli M, Reni M, Tansini G. Chemotherapy-Induced Anemia and Oncologist Perception on Treatment: Results of a Web-Based Survey. TUMORI JOURNAL 2018; 99:45-50. [DOI: 10.1177/030089161309900108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and background Anemia prevalence and incidence in chemotherapy-treated patients is high. Erythropoiesis-stimulating agents (ESAs) are frequently employed in the management of chemotherapy-induced anemia. However, other treatments such as red blood transfusion or iron supplementation are normally used. Recent international guidelines raised some concern about ESAs employment with a possible impact in chemotherapy-induced anemia management and changes in clinical practice behavior. Methods To evaluate opinions about chemotherapy-induced anemia clinical management preference, the Associazione Italiana Oncologia Medica (AIOM) Lombardy section coordinators sent via email a 12-item questionnaire about their knowledge on CIA and usual therapeutic strategies to manage this adverse event to AIOM Lombardy onco-hematologist members. Results From January 2011 to March 2011, 81 questionnaires were collected with an approximated share of 30%. The survey was completed mainly by oncologists (91%) aged 35–50 years (50%). Chemotherapy-induced anemia was considered to have clinical impact in changing cancer therapeutic strategy by nearly 60% of the respondents. ESAs were administered largely (80%) with concomitant iron supplementation in 52%; 38% jointly used blood transfusion as part of the therapy. Nearly 20% of those who replied correctly employed transferrin saturation levels as a marker to guide iron supplementation. Physician prescribers strictly followed the guidelines to start and stop ESAs even if 14% were negatively influenced by new ASCO recommendations. ESA biosimilars were considered future substitutes of originators in 45% of the cases. Conclusions Chemotherapy-induced anemia was perceived as an adverse event with a mild impact on clinical practice. ESAs were largely employed, however the number of transfusions and lack of employment of markers of iron depletion suggested that adherence to guidelines could be theoretically met but with some discordances regarding the most appropriate strategies in daily clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Roberto Buzzoni
- DH & Terapia Ambulatoriale Oncologica, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - Mary Cabiddu
- Oncologia Medica, Ospedale Treviglio-Caravaggio, Bergamo
| | - Carlo Carnaghi
- Oncologia Medica, Humanitas Mater Domini, Castellanza (VA), Onco-Ematologia, Humanitas Cancer Center, Rozzano (MI)
| | - Marco Danova
- Medicina oncologica, Ospedale Civile, Vigevano (PV)
| | | | | | | | | | | |
Collapse
|
41
|
Widick P, Brunner AM, Schiffman F. Hematologic Manifestations of Malignancy. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00155-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
42
|
Erythropoietin promoted the proliferation of hepatocellular carcinoma through hypoxia induced translocation of its specific receptor. Cancer Cell Int 2017; 17:119. [PMID: 29238266 PMCID: PMC5725980 DOI: 10.1186/s12935-017-0494-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022] Open
Abstract
Background Erythropoietin (EPO) is a hypoxia-inducible stimulator of erythropoiesis. Besides its traditional application in anemia therapy, it offers an effective treatment in the cancer patients, especially those who receive chemotherapy. Several reports indicated that it could promote the tumor cell proliferation through its specific receptor (EPOR). Unfortunately, the role of EPO/EPOR in hepatocellular carcinoma (HCC) progressing is still uncertain. Methods Protein in tumor tissue from HCC patients or H22 tumor-bearing mice was detected with immunohistochemistry. Cells were cultured under 1% oxygen to establish hypoxia. RT-PCR and western blotting were used to measure mRNA and protein of EPO/EPOR, respectively. MTT, flow cytometry and PCNA staining were used to detect cell proliferation. Immunofluorescence staining was applied to study the expression and location of cellular EPOR. The EPOR binding studies were performed with 125I-EPO radiolabeling assay. Results EPO and EPOR protein were up-regulated in HCC tissue of patients and H22-bearing mice. These were positively correlated with hypoxia-inducible factor -1 α and ki-67. Hypoxia up-regulated the expression of EPO and EPOR in HepG2 cells. It also induced the proliferation and increased the percentage of divided cells after 24, 48 and 72 h treatment. These were inhibited in cells pre-treated with 0.5 μg/mL soluble-EPOR. Immunofluorescence staining presented that EPOR was obviously translocated from nucleus to cytoplasm and membrane under hypoxia. EPOR binding activity was also increased after exposure to hypoxia. Recombinant human erythropoietin obviously elevated cell proliferation rate and the percentage of divided under hypoxia but not normoxia, which were also inhibited by soluble-EPOR. Conclusions Our result indicated for the first time that EPO promoted the proliferation of HCC cells through hypoxia induced translocation of it specific receptor. Trial registration TJC20141113, retrospectively registered
Collapse
|
43
|
He L, Wu S, Hao Q, Dioum EM, Zhang K, Zhang C, Li W, Zhang W, Zhang Y, Zhou J, Pang Z, Zhao L, Ma X, Li M, Zhang Q. Local blockage of self-sustainable erythropoietin signaling suppresses tumor progression in non-small cell lung cancer. Oncotarget 2017; 8:82352-82365. [PMID: 29137269 PMCID: PMC5669895 DOI: 10.18632/oncotarget.19354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/30/2017] [Indexed: 12/18/2022] Open
Abstract
Functional significance of co-expressed erythropoietin (EPO) and its receptor (EPOR) in non-small cell lung cancer (NSCLC) had been under debate. In this study, co-overexpression of EPO/EPOR was confirmed to be positively associated with poor survival in NSCLC. The serum EPO in 14 of 35 enrolled NSCLC patients were found elevated significantly and decreased to normal level after tumor resection. With primary tumor cell culture and patient-derived tumor xenograft (PDX) mouse model, the EPO secretion from the tumors of these 14 patients was verified. Then, we proved the patient derived serum EPO was functionally active and had growth promotion effect in EPO/EPOR overexpressed but not in EPO/EPOR under-expressed NSCLC cells. We also illustrated EPO promoted NSCLC cell proliferation through an EPOR/Jak2/Stat5a/cyclinD1 pathway. In xenograft mouse model, we proved local application of EPO neutralizing antibody and short hairpin RNA (shRNA) against EPOR effectively inhibited the growth of EPO/EPOR overexpressed NSCLC cells and prolonged survivals of the mice. Finally, EPO/EPOR/Jak2/Stat5a/cyclinD1 signaling was found to be a mediator of hypoxia induced growth in EPO/EPOR overexpressed NSCLC. Our results illustrated a subgroup of NSCLC adapt to hypoxia through self-sustainable EPO/EPOR signaling and suggest local blockage of EPO/EPOR as potential therapeutic method in this distinct NSCLC population.
Collapse
Affiliation(s)
- Lei He
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Shouzhen Wu
- Shaanxi Institute of Pediatric Diseases, Xi'an Children's Hospital, Xi'an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Elhadji M Dioum
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Current/Present address: Diabetes Department, Nestle Institute of Health Science, EPFL Campus, Lausanne, Switzerland
| | - Kuo Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Wei Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jiming Zhou
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Zhijun Pang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Lijuan Zhao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Xiaowen Ma
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Qiuyang Zhang
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Current/Present address: Center for Esophageal Research, Baylor University Medical Center, Dallas, Texas, USA
| |
Collapse
|
44
|
Bonnas C, Wüstefeld L, Winkler D, Kronstein-Wiedemann R, Dere E, Specht K, Boxberg M, Tonn T, Ehrenreich H, Stadler H, Sillaber I. EV-3, an endogenous human erythropoietin isoform with distinct functional relevance. Sci Rep 2017; 7:3684. [PMID: 28623280 PMCID: PMC5473850 DOI: 10.1038/s41598-017-03167-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/25/2017] [Indexed: 01/14/2023] Open
Abstract
Generation of multiple mRNAs by alternative splicing is well known in the group of cytokines and has recently been reported for the human erythropoietin (EPO) gene. Here, we focus on the alternatively spliced EPO transcript characterized by deletion of exon 3 (hEPOΔ3). We show co-regulation of EPO and hEPOΔ3 in human diseased tissue. The expression of hEPOΔ3 in various human samples was low under normal conditions, and distinctly increased in pathological states. Concomitant up-regulation of hEPOΔ3 and EPO in response to hypoxic conditions was also observed in HepG2 cell cultures. Using LC-ESI-MS/MS, we provide first evidence for the existence of hEPOΔ3 derived protein EV-3 in human serum from healthy donors. Contrary to EPO, recombinant EV-3 did not promote early erythroid progenitors in cultures of human CD34+ haematopoietic stem cells. Repeated intraperitoneal administration of EV-3 in mice did not affect the haematocrit. Similar to EPO, EV-3 acted anti-apoptotic in rat hippocampal neurons exposed to oxygen-glucose deprivation. Employing the touch-screen paradigm of long-term visual discrimination learning, we obtained first in vivo evidence of beneficial effects of EV-3 on cognition. This is the first report on the presence of a naturally occurring EPO protein isoform in human serum sharing non-erythropoietic functions with EPO.
Collapse
Affiliation(s)
| | - Liane Wüstefeld
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine and DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Daniela Winkler
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine and DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Romy Kronstein-Wiedemann
- German Red Cross Blood Donor Service North-East, Institute of Transfusion Medicine, Dresden, Germany
| | - Ekrem Dere
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine and DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Katja Specht
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Torsten Tonn
- German Red Cross Blood Donor Service North-East, Institute of Transfusion Medicine, Dresden, Germany
- Department of Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, Technische Universität Desden, Dresden, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine and DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | | | | |
Collapse
|
45
|
Losem C, Koenigsmann M, Rudolph C. Biosimilar Retacrit ® (epoetin zeta) in the treatment of chemotherapy-induced symptomatic anemia in hematology and oncology in Germany (ORHEO) - non-interventional study. Onco Targets Ther 2017; 10:1295-1305. [PMID: 28280364 PMCID: PMC5338968 DOI: 10.2147/ott.s122427] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Symptomatic anemia is a frequent and severe complication of chemotherapy that is commonly treated with erythropoiesis-stimulating agents. The primary objective of this study was to assess the change in hemoglobin levels in patients with chemotherapy-induced anemia (CIA) following treatment with biosimilar Retacrit® (epoetin zeta). Secondary objectives included changes in hematologic parameters and tolerability. Methods This was a non-interventional, multicenter, long-term observational study that is part of an ongoing surveillance program for epoetin zeta. Adult patients (N=291) with solid tumors, malignant lymphomas or multiple myeloma, and chemotherapy-induced symptomatic anemia, who were eligible for treatment with biosimilar epoetin zeta, were enrolled. Patients were evaluated at enrollment, 3 months, and 6 months. Results Evaluable patients had lymphoma or myeloma (n=30) or solid tumors (n=260). At 3 months, patients with lymphoma and myeloma showed the greatest increase in mean (SD) hemoglobin from 9.2 (0.9) to 11.0 (1.8) g/dL, whereas patients with breast cancer showed the smallest increase from 10.0 (1.0) to 11.1 (1.2) g/dL. At 6 months, the greatest mean increase occurred in patients with lymphoma or myeloma from 11.0 (1.8) to 11.7 (2.3) g/dL, and the smallest in patients with other solid tumors from 10.9 (1.4) to 11.1 (1.5) g/dL. Patient evaluation of epoetin zeta therapy was positive, as most patients expressed satisfaction with epoetin zeta treatment during the study, compliance with treatment was high, and most indicated their willingness to be retreated if necessary. Epoetin zeta was also well tolerated; overall, in 25 patients (8.6%), there were 31 adverse events. Conclusion Despite variability among different disease groups, epoetin zeta was effective and well tolerated in patients with different types of solid tumors and hematologic malignancies.
Collapse
|
46
|
Miao S, Wang SM, Cheng X, Li YF, Zhang QS, Li G, He SQ, Chen XP, Wu P. Erythropoietin promoted the proliferation of hepatocellular carcinoma through hypoxia induced translocation of its specific receptor. Cancer Cell Int 2017. [PMID: 29238266 DOI: 10.1186/s12935-017-04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Erythropoietin (EPO) is a hypoxia-inducible stimulator of erythropoiesis. Besides its traditional application in anemia therapy, it offers an effective treatment in the cancer patients, especially those who receive chemotherapy. Several reports indicated that it could promote the tumor cell proliferation through its specific receptor (EPOR). Unfortunately, the role of EPO/EPOR in hepatocellular carcinoma (HCC) progressing is still uncertain. METHODS Protein in tumor tissue from HCC patients or H22 tumor-bearing mice was detected with immunohistochemistry. Cells were cultured under 1% oxygen to establish hypoxia. RT-PCR and western blotting were used to measure mRNA and protein of EPO/EPOR, respectively. MTT, flow cytometry and PCNA staining were used to detect cell proliferation. Immunofluorescence staining was applied to study the expression and location of cellular EPOR. The EPOR binding studies were performed with 125I-EPO radiolabeling assay. RESULTS EPO and EPOR protein were up-regulated in HCC tissue of patients and H22-bearing mice. These were positively correlated with hypoxia-inducible factor -1 α and ki-67. Hypoxia up-regulated the expression of EPO and EPOR in HepG2 cells. It also induced the proliferation and increased the percentage of divided cells after 24, 48 and 72 h treatment. These were inhibited in cells pre-treated with 0.5 μg/mL soluble-EPOR. Immunofluorescence staining presented that EPOR was obviously translocated from nucleus to cytoplasm and membrane under hypoxia. EPOR binding activity was also increased after exposure to hypoxia. Recombinant human erythropoietin obviously elevated cell proliferation rate and the percentage of divided under hypoxia but not normoxia, which were also inhibited by soluble-EPOR. CONCLUSIONS Our result indicated for the first time that EPO promoted the proliferation of HCC cells through hypoxia induced translocation of it specific receptor. Trial registration TJC20141113, retrospectively registered.
Collapse
Affiliation(s)
- Shuo Miao
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Su-Mei Wang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xue Cheng
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yao-Feng Li
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Qing-Song Zhang
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technolgy, Wuhan, 430030 China
| | - Gang Li
- Department of Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Song-Qing He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Xiao-Ping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technolgy, Wuhan, 430030 China
| | - Ping Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
47
|
Våtsveen TK, Sponaas AM, Tian E, Zhang Q, Misund K, Sundan A, Børset M, Waage A, Brede G. Erythropoietin (EPO)-receptor signaling induces cell death of primary myeloma cells in vitro. J Hematol Oncol 2016; 9:75. [PMID: 27581518 PMCID: PMC5007700 DOI: 10.1186/s13045-016-0306-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 08/24/2016] [Indexed: 01/04/2023] Open
Abstract
Background Multiple myeloma is an incurable complex disease characterized by clonal proliferation of malignant plasma cells in a hypoxic bone marrow environment. Hypoxia-dependent erythropoietin (EPO)-receptor (EPOR) signaling is central in various cancers, but the relevance of EPOR signaling in multiple myeloma cells has not yet been thoroughly investigated. Methods Myeloma cell lines and malignant plasma cells isolated from bone marrow of myeloma patients were used in this study. Transcript levels were analysed by quantitative PCR and cell surface levels of EPOR in primary cells by flow cytometry. Knockdown of EPOR by short interfering RNA was used to show specific EPOR signaling in the myeloma cell line INA-6. Flow cytometry was used to assess viability in primary cells treated with EPO in the presence and absence of neutralizing anti-EPOR antibodies. Gene expression data for total therapy 2 (TT2), total therapy 3A (TT3A) trials and APEX 039 and 040 were retrieved from NIH GEO omnibus and EBI ArrayExpress. Results We show that the EPOR is expressed in myeloma cell lines and in primary myeloma cells both at the mRNA and protein level. Exposure to recombinant human EPO (rhEPO) reduced viability of INA-6 myeloma cell line and of primary myeloma cells. This effect could be partially reversed by neutralizing antibodies against EPOR. In INA-6 cells and primary myeloma cells, janus kinase 2 (JAK-2) and extracellular signal regulated kinase 1 and 2 (ERK-1/2) were phosphorylated by rhEPO treatment. Knockdown of EPOR expression in INA-6 cells reduced rhEPO-induced phospo-JAK-2 and phospho-ERK-1/2. Co-cultures of primary myeloma cells with bone marrow-derived stroma cells did not protect the myeloma cells from rhEPO-induced cell death. In four different clinical trials, survival data linked to gene expression analysis indicated that high levels of EPOR mRNA were associated with better survival. Conclusions Our results demonstrate for the first time active EPOR signaling in malignant plasma cells. EPO-mediated EPOR signaling reduced the viability of myeloma cell lines and of malignant primary plasma cells in vitro. Our results encourage further studies to investigate the importance of EPO/EPOR in multiple myeloma progression and treatment. Trial registration [Trial registration number for Total Therapy (TT) 2: NCT00083551 and TT3: NCT00081939].
Collapse
Affiliation(s)
- Thea Kristin Våtsveen
- K.G. Jebsen Centre of Myeloma Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Present Address: Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Present Address: Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Anne-Marit Sponaas
- K.G. Jebsen Centre of Myeloma Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Erming Tian
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Qing Zhang
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kristine Misund
- K.G. Jebsen Centre of Myeloma Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anders Sundan
- K.G. Jebsen Centre of Myeloma Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, N-7491, Trondheim, Norway
| | - Magne Børset
- K.G. Jebsen Centre of Myeloma Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Immunology and Transfusion Medicine, St Olavs University Hospital, Trondheim, Norway
| | - Anders Waage
- K.G. Jebsen Centre of Myeloma Research, Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Haematology, St. Olavs University Hospital, Trondheim, Norway
| | - Gaute Brede
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.
| |
Collapse
|
48
|
Gemici C, Yetmen O, Yaprak G, Ozden S, Tepetam H, Ozyurt H, Mayadagli A. Is there any role of intravenous iron for the treatment of anemia in cancer? BMC Cancer 2016; 16:661. [PMID: 27542823 PMCID: PMC4992337 DOI: 10.1186/s12885-016-2686-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 08/07/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Anemia is a major cause of morbidity in patients with cancer resulting in poor physical performance, prognosis and therapy outcome. The aim of this study is to assess the efficacy of intravenous (iv) iron administration for the correction of anemia, for the prevention of exacerbation of anemia, for decreasing blood transfusion rates, and for the survival of cancer patients. METHODS Patients with different solid tumor diagnosis who received iv iron during their cancer treatment were evaluated retrospectively. Sixty-three patients with hemoglobin (Hgb) levels between ≥ 9 g/dL, and ≤ 10 g/dL, and no urgent need for red blood cell transfusion were included in this retrospective analysis. The aim of cancer treatment was palliative for metastatic patients (36 out of 63), or adjuvant or curative for patients with localized disease (27 out of 63). All the patients received 100 mg of iron sucrose which was delivered intravenously in 100 mL of saline solution, infused within 30 min, 5 infusions every other day. Complete blood count, serum iron, and ferritin levels before and at every 1 to 3 months subsequently after iv iron administration were followed regularly. RESULTS Initial mean serum Hgb, serum ferritin and serum iron levels were 9.33 g/dL, 156 ng/mL, and 35.9 μg/dL respectively. Mean Hgb, ferritin, and iron levels 1 to 3 months, and 6 to 12 months after iv iron administration were 10.4 g/dL, 11.2 g/dL, 298.6 ng/mL, 296.7 ng/mL, and 71.6 μg/dL, 67.7 μg/dL respectively with a statistically significant increase in the levels (p < 0.001). Nineteen patients (30 %) however had further decrease in Hgb levels despite iv iron administration, and blood transfusion was necessary in 18 of these 19 patients (28.5 %). The 1-year overall survival rates differed in metastatic cancer patients depending on their response to iv iron; 61.1 % in responders versus 35.3 % in non-responders, (p = 0.005), furthermore response to iv iron correlated with tumor response to cancer treatment, and this relation was statistically significant, (p < 0.001). CONCLUSIONS Iv iron administration in cancer patients undergoing active oncologic treatment is an effective and safe measure for correction of anemia, and prevention of worsening of anemia. Amelioration of anemia and increase in Hgb levels with iv iron administration in patients with disseminated cancer is associated with increased tumor response to oncologic treatment and overall survival. Response to iv iron may be both a prognostic and a predictive factor for response to cancer treatment and survival.
Collapse
Affiliation(s)
- Cengiz Gemici
- Department of Oncology, Dr. Lutfi Kirdar Kartal Education and Research Hospital, Cevizli, Istanbul, Turkey.
| | - Ozlem Yetmen
- Department of Oncology, Dr. Lutfi Kirdar Kartal Education and Research Hospital, Cevizli, Istanbul, Turkey
| | - Gokhan Yaprak
- Department of Oncology, Dr. Lutfi Kirdar Kartal Education and Research Hospital, Cevizli, Istanbul, Turkey
| | - Sevgi Ozden
- Department of Oncology, Dr. Lutfi Kirdar Kartal Education and Research Hospital, Cevizli, Istanbul, Turkey
| | - Huseyin Tepetam
- Department of Oncology, Dr. Lutfi Kirdar Kartal Education and Research Hospital, Cevizli, Istanbul, Turkey
| | - Hazan Ozyurt
- Department of Oncology, Dr. Lutfi Kirdar Kartal Education and Research Hospital, Cevizli, Istanbul, Turkey
| | | |
Collapse
|
49
|
Launay-Vacher V, Janus N, Deray G, Scotté F. Results from the LIDO anemia survey: adherence to EORTC guidelines in cancer patients in France. Support Care Cancer 2016; 24:3347-51. [DOI: 10.1007/s00520-016-3162-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/07/2016] [Indexed: 10/22/2022]
|
50
|
Yigit M, Sogut O, Yigit E, Turkdogan KA, Kaplan O, Dur A, Sonmez E, Demirel B. The relationship between anemia and recurrence of ischemic stroke in patients with Trousseau's syndrome: A retrospective cross-sectional study. Turk J Emerg Med 2016; 16:65-68. [PMID: 27896324 PMCID: PMC5121256 DOI: 10.1016/j.tjem.2015.11.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 11/09/2015] [Accepted: 11/27/2015] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES The relationship between cancer and thrombosis was first recognized by the French internist Armand Trousseau in 1865. Trousseau's syndrome is a spectrum of symptoms that result from recurrent thromboembolism associated with cancer or malignancy-related hypercoagulability. In this study, we investigated whether demographics, clinical features, or laboratory findings were able to predict recurrent stroke episodes in patients with Trousseau's syndrome. METHODS In total, 178 adult patients were enrolled in this retrospective cross-sectional study. All patients had been admitted to the emergency room of our hospital between January 2011 and September 2014 and were diagnosed with acute ischemic stroke. Patients were divided into two groups: patients with malignancy (Trousseau's syndrome), and patients without malignancy. RESULTS There were several significant differences between the laboratory results of the two patient groups. For patients with Trousseau's, the hemoglobin levels for those with one stroke was 12.29 ± 1.81, while those in patients who had experienced more than one stroke was 10.94 ± 2.14 (p = 0.004). CONCLUSIONS Trousseau's syndrome is a cancer-associated coagulopathy associated with high morbidity and mortality rates. In this study, anemia was associated with increased stroke recurrence in patients with malignancy (Trousseau's syndrome).
Collapse
Affiliation(s)
- Mehmet Yigit
- Department of Emergency Medicine, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Ozgur Sogut
- Department of Emergency Medicine, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Eda Yigit
- Department of Emergency Medicine, Sisli Etfal Training and Research Hospital, Istanbul, Turkey
| | | | - Onur Kaplan
- Department of Emergency Medicine, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Ali Dur
- Department of Emergency Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ertan Sonmez
- Department of Emergency Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Bulut Demirel
- Department of Emergency Medicine, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| |
Collapse
|