pubmed_id
stringlengths
41
43
abstract
stringlengths
3
18.8k
http://www.ncbi.nlm.nih.gov/pubmed/28923211
1. Semin Oncol. 2017 Apr;44(2):132-135. doi: 10.1053/j.seminoncol.2017.06.007. Epub 2017 Jul 13. Nivolumab and pembrolizumab: Monoclonal antibodies against programmed cell death-1 (PD-1) that are interchangeable. Prasad V(1), Kaestner V(2). Author information: (1)Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR; Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR; Senior Scholar in the Center for Health Care Ethics, Oregon Health & Science University, Portland, OR. Electronic address: [email protected]. (2)Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR. Electronic address: [email protected]. Nivolumab (Opdivo, Bristol Meyer Squibb, New York, NY) and pembrolizumab (Keytruda, Merck, Kenilworth, NJ) are the first two US Food and Drug Administration (FDA)-approved monoclonal antibodies targeting programmed death-1 (PD-1). Nivolumab and pembrolizumab work by interfering with the interaction between PD-1 and programmed death ligand-1 (PD-L1), whose unimpeded interaction downregulates T cells allowing cancer cells to evade immune surveillance. These drugs have earned a series of FDA approvals for melanoma, non-small cell lung cancer (NSCLC), head and neck squamous cell cancer (HNSCC), urothelial cancer, classical Hodgkin lymphoma, and renal cell cancer. In this review we will summarize the data for efficacy and toxicity for these two agents. We conclude that they represent two valuable but interchangeable alternatives to target their approved indications. We will discuss how this can help global payers seeking to contain the cost of cancer therapeutics that continues to spiral out of control. Copyright © 2017. Published by Elsevier Inc. DOI: 10.1053/j.seminoncol.2017.06.007 PMID: 28923211 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27058852
1. Clin Adv Hematol Oncol. 2015 Dec;13(12):858-68. Programmed death 1 immune checkpoint inhibitors. Trivedi MS(1), Hoffner B(2), Winkelmann JL(1), Abbott ME(1), Hamid O(2), Carvajal RD(1). Author information: (1)Columbia University Medical Center, New York, New York. (2)The Angeles Clinic and Research Institute, Los Angeles, California. Programmed death 1 (PD-1) is an immune checkpoint that provides inhibitory signals to the immune system in order to modulate the activity of T cells in peripheral tissues and maintain self-tolerance in the setting of infection and inflammation. In cancer, the immune checkpoints are exploited so that the tumor cells are able to evade the immune system. Immune checkpoint inhibitors are a type of cancer immunotherapy that targets pathways such as PD-1 in order to reinvigorate and enhance the immune response against tumor cells. The US Food and Drug Administration (FDA) has approved 2 PD-1 inhibitors, nivolumab and pembrolizumab, and several others are under investigation. Although PD-1 inhibitors have demonstrated activity in many different types of malignancies, FDA approval has been granted only in melanoma and in non-small cell lung cancer (NSCLC). Identifying biomarkers that can predict response to PD-1 inhibitors is critical to maximizing the benefit of these agents. Future directions for PD-1 inhibitors include investigation of combination therapies, use in malignancies other than melanoma and NSCLC, and refinement of biomarkers. PMID: 27058852 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27795534
1. Rinsho Ketsueki. 2016;57(10):2224-2231. doi: 10.11406/rinketsu.57.2224. [Immune checkpoint inhibitors targeting programmed cell death-1 (PD-1) in cancer therapy]. [Article in Japanese] Hatae R(1), Chamoto K. Author information: (1)Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University. Immune checkpoint inhibitors, especially anti-programmed cell death-1 (PD-1) antibodies, have revolutionized cancer therapy. A PD-1 antibody, nivolumab, was the first of these agents to be approved by the Pharmaceuticals and Medical Devices Agency (PMDA) of Japan, as a new cancer drug for melanoma, in July 2014. While PD-1 mAb therapy has so far been approved only for untreated malignant melanomas and non-small cell lung cancer, many clinical studies on various types of cancer have been conducted worldwide. Immune checkpoint inhibitors target lymphocytes rather than cancer cells, and evoke an anti-tumor immune reaction. Since the activated lymphocytes recognize various tumor-associated antigens including a mutated antigen, immune checkpoint inhibitors exhibit continuous long-term effectiveness, despite the generation of genetic mutations in cancer cells. As compared with previous cancer treatments, immune checkpoint inhibitors show superior efficacy against tumors with fewer side effects. Therefore, these novel immune checkpoint inhibitor agents are anticipated to become a 4th cancer treatment option following surgery, chemotherapy, and radiation therapy. Herein, we review the main clinical results of PD-1 mAb cancer immunotherapy obtained to date and discuss issues relevant to administering this form of treatment. DOI: 10.11406/rinketsu.57.2224 PMID: 27795534 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30304963
1. Expert Rev Anticancer Ther. 2018 Dec;18(12):1169-1175. doi: 10.1080/14737140.2018.1535315. Epub 2018 Oct 22. Nivolumab for the treatment of hepatocellular carcinoma. Finkelmeier F(1), Waidmann O(1), Trojan J(1). Author information: (1)a Medizinische Klinik 1 , Universitätsklinikum Frankfurt, Goethe-Universität , Frankfurt/Main , Germany. T-cell checkpoint inhibition as a cancer treatment approach has been the main breakthrough in cancer treatment during the last years. Since the approval of the first commercial CTLA-4 antibody ipilimumab in 2011 for the treatment of melanoma, research and drug development in this field has accelerated massively. In 2014, the US Food and Drug Administration (FDA) approved the first PD-1 targeting agent, namely pembrolizumab, shortly followed by nivolumab. Areas covered: Nivolumab is a fully human immunoglobulin G4 anti-PD-1 monoclonal antibody which is approved for multiple advanced malignancies, including melanoma, non-small cell lung cancer, renal cell cancer, Hodgkin's lymphoma, squamous head and neck cancer, and urothelial carcinoma. In September 2017, nivolumab was approved by the FDA for liver cancer as a second line treatment after failure of sorafenib based on the data of the multi-cohort phase 1/2 trial CheckMate-040. This article reviews the concept of immunotherapy in liver cancer with focus on nivolumab. Expert commentary: Immunotherapy in hepatocellular carcinoma is safe and is a new treatment option for patients with advanced stage disease besides sorafenib and regorafenib in the US. Randomized phase III trials of nivolumab, pembrolizumab, atezolizumab, durvalumab and tislelizumab as mono- or combination-therapy are ongoing. DOI: 10.1080/14737140.2018.1535315 PMID: 30304963 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33686894
1. Expert Opin Emerg Drugs. 2021 Jun;26(2):79-92. doi: 10.1080/14728214.2021.1901884. Epub 2021 Mar 19. Emerging PD-1/PD-L1 antagonists for the treatment of malignant melanoma. Vanella V(1), Festino L(1), Vitale MG(1), Alfano B(1), Ascierto PA(1). Author information: (1)Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy. INTRODUCTION: Increased understanding of the interactive mechanisms between tumors and the immune system led to the development of immune checkpoint inhibitors, which have revolutioned the treatment of metastatic melanoma and subsequently many other tumors. In 2014, nivolumab and pembrolizumab, two checkpoint inhibitors binding to PD-1, were approved for the treatment of metastatic melanoma. Since then, a plethora of new molecules have enriched the armamentarium against melanoma. AREAS COVERED: This review summarizes the last updates about treatment with nivolumab and pembrolizumab, data on other PD-1/PDL-1 agents such as spartalizumab and atezolizumab and emerging compounds, new combinations with NKTR-214, anti LAG-3, anti IDO-1 and TVEC, new checkpoint inhibitors (e.g. TIM-3 or TIGIT) and other new molecules for the treatment of metastatic melanoma. EXPERT OPINION: Currently, several ongoing clinical trials are investigating novel molecules, or immunotherapy combinations, in order to achieve even better survival outcomes for patients, overcoming resistance mechanisms and improving toxicity profiles. The challenge in the near future will be to select the most appropriate treatments according to the specific characteristics of the patients. DOI: 10.1080/14728214.2021.1901884 PMID: 33686894 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32863353
1. Jpn J Infect Dis. 2021 Mar 24;74(2):102-109. doi: 10.7883/yoken.JJID.2020.444. Epub 2020 Aug 31. Growth Characteristics of Rickettsia Species LON Strains Closely Related to Rickettsia japonica Isolated from Haemaphysalis longicornis Ticks in Mouse Derived L929 and Human-Derived THP-1 Host Cell Lines. Tai H(1), Su H(1), Takamoto N(1), Fujita H(1)(2), Takano A(3), Oishi S(4), Abe F(4), Ando S(5), Ohashi N(1). Author information: (1)Graduate Program in Pharmaceutical and Nutritional Sciences, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Japan. (2)Mahara Institute of Medical Acarology, Japan. (3)Department of Veterinary Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Japan. (4)Department of Microbiology, Shizuoka Institute of Environment and Hygiene, Japan. (5)Department of Virology I, National Institute of Infectious Diseases, Japan. Non-pathogenic Rickettsia species LON strains closely related to an agent of Japanese spotted fever (JSF), R. japonica, were isolated in Japan from Haemaphysalis longicornis ticks in 2001. However, the biological properties of LONs in mammalian host cells are poorly understood. In this study, microscopic analysis showed that LONs in a mouse-derived L929 host cell line were rod shaped with sizes of 0.3-0.5 × 0.5-2.0 μm. Molecular analysis revealed the existence of a LON-specific disrupted open reading frame in R. japonica-related group-specific DNA regions. Growth kinetics of LON-2 and LON-13 strains analyzed by a quantitative real-time PCR showed 100-fold or more increment of LONs cultured in L929 host cells at 30°C and slightly less increment at 33°C, and 25-fold increment in human-derived THP-1 host cells at 35°C on day 7 (168 h) post infection. The generation times of the two LON strains cultured in L929 and THP-1 were estimated to be 9.4-12.9 h and 9.6-10.9 h, respectively. To our knowledge, this is the first report on the biological characteristics of Rickettsia sp. LON strains in mammalian cells, which may provide significant information for the experimental approaches for other rickettsiae. DOI: 10.7883/yoken.JJID.2020.444 PMID: 32863353 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33808008
1. Pharmaceutics. 2021 Mar 5;13(3):341. doi: 10.3390/pharmaceutics13030341. Dermatillomania: Strategies for Developing Protective Biomaterials/Cloth. Ravipati P(1), Conti B(1), Chiesa E(1), Andrieux K(2). Author information: (1)Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy. (2)Department of Pharmacie, Université Paris Descartes, 75006 Paris, France. Dermatillomania or skin picking disorder (SPD) is a chronic, recurrent, and treatment resistant neuropsychiatric disorder with an underestimated prevalence that has a concerning negative impact on an individual's health and quality of life. The current treatment strategies focus on behavioral and pharmacological therapies that are not very effective. Thus, the primary objective of this review is to provide an introduction to SPD and discuss its current treatment strategies as well as to propose biomaterial-based physical barrier strategies as a supporting or alternative treatment. To this end, searches were conducted within the PubMed database and Google Scholar, and the results obtained were organized and presented as per the following categories: prevalence, etiology, consequences, diagnostic criteria, and treatment strategies. Furthermore, special attention was provided to alternative treatment strategies and biomaterial-based physical treatment strategies. A total of six products with the potential to be applied as physical barrier strategies in supporting SPD treatment were shortlisted and discussed. The results indicated that SPD is a complex, underestimated, and underemphasized neuropsychiatric disorder that needs heightened attention, especially with regard to its treatment and care. Moreover, the high synergistic potential of biomaterials and nanosystems in this area remains to be explored. Certain strategies that are already being utilized for wound healing can also be further exploited, particularly as far as the prevention of infections is concerned. DOI: 10.3390/pharmaceutics13030341 PMCID: PMC8001957 PMID: 33808008 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/36016429
1. Viruses. 2022 Aug 18;14(8):1807. doi: 10.3390/v14081807. Clinical Differentiation of Severe Fever with Thrombocytopenia Syndrome from Japanese Spotted Fever. Nakada N(1)(2), Yamamoto K(1), Tanaka M(1), Ashizawa H(1), Yoshida M(3), Umemura A(3), Fukuda Y(3), Katoh S(4), Sumiyoshi M(5), Mihara S(5), Kobayashi T(6), Ito Y(1), Ashizawa N(1)(7), Takeda K(1), Ide S(6)(8), Iwanaga N(1), Takazono T(1)(7), Tashiro M(7), Tanaka T(7), Nakamichi S(2), Morimoto K(9), Ariyoshi K(9), Morita K(10), Kurihara S(11), Yanagihara K(12), Furumoto A(5)(8), Izumikawa K(7), Mukae H(1). Author information: (1)Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki 852-8102, Japan. (2)Health Center, Nagasaki University, Nagasaki 852-8521, Japan. (3)Department of Respiratory Medicine, Sasebo City General Hospital, Sasebo 857-8511, Japan. (4)Department of General Internal Medicine, Nagasaki Rosai Hospital, Sasebo 857-0134, Japan. (5)Department of Respiratory Medicine, Isahaya General Hospital, Isahaya 854-8501, Japan. (6)Department of Respiratory Medicine, Sasebo Chuo Hospital, Sasebo 857-1195, Japan. (7)Department of Infection Control and Education Center, Nagasaki University Hospital, Nagasaki 852-8102, Japan. (8)Infectious Disease Experts Training Center, Nagasaki University Hospital, Nagasaki 852-8102, Japan. (9)Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan. (10)Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan. (11)Department of Medical Safety, Nagasaki University Hospital, Nagasaki 852-8102, Japan. (12)Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki 852-8102, Japan. Severe fever with thrombocytopenia syndrome (SFTS) and Japanese spotted fever (JSF; a spotted fever group rickettsiosis) are tick-borne zoonoses that are becoming a significant public health threat in Japan and East Asia. Strategies for treatment and infection control differ between the two; therefore, initial differential diagnosis is important. We aimed to compare the clinical characteristics of SFTS and JSF based on symptomology, physical examination, laboratory data, and radiography findings at admission. This retrospective study included patients with SFTS and JSF treated at five hospitals in Nagasaki Prefecture, western Japan, between 2013 and 2020. Data from 23 patients with SFTS and 38 patients with JSF were examined for differentiating factors and were divided by 7:3 into a training cohort and a validation cohort. Decision tree analysis revealed leukopenia (white blood cell [WBC] < 4000/μL) and altered mental status as the best differentiating factors (AUC 1.000) with 100% sensitivity and 100% specificity. Using only physical examination factors, absence of skin rash and altered mental status resulted in the best differentiating factors with AUC 0.871, 71.4% sensitivity, and 90.0% specificity. When treating patients with suspected tick-borne infection, WBC < 4000/µL, absence of skin rash, and altered mental status are very useful to differentiate SFTS from JSF. DOI: 10.3390/v14081807 PMCID: PMC9415593 PMID: 36016429 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35507925
1. Trop Biomed. 2022 Mar 1;39(1):55-59. doi: 10.47665/tb.39.1.007. A new record of Rickettsia japonica in ticks infesting a Burmese ferret-badger in Thailand. Hirunkanokpun S(1), Ahantarig A(2), Baimai B(2), Pramual P(3), Trinachartvanit W(2). Author information: (1)Department of Biology, Faculty of Science, Ramkhamhaeng University, Bangkok, 10240, Thailand. (2)Biodiversity Research Cluster, Department of Biology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand. (3)Department of Biology, Faculty of science, Mahasarakham University, Maha Sarakham, 44150, Thailand. Ticks are important vectors of arthropod-borne diseases and they can transmit a wide variety of zoonotic pathogens to humans, domestic and wild animals. Rickettsia japonica is a member of SFG rickettsiae causing Japanese spotted fever (JSF) and can transmit to humans via infected ticks. In this study, we report the first case of Rickettsia japonica in Haemaphysalis hystricis tick collected from a roadkill Burmese ferret-badger ( Melogale personata ) in Loei province, northeastern Thailand. According to the DNA sequences and phylogenetic analyses of the outer membrane protein A and B genes ( ompA and ompB), the detected R. japonica was identical to those found in JSF patients in Korea, Japan, and China, and closely related to Rickettsia detected by ompA in a tick from Thailand. Further study on the prevalence of R. japonica and diversity of mammalian reservoir hosts will be useful to gain a better understanding of JSF epidemiology. DOI: 10.47665/tb.39.1.007 PMID: 35507925 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35668012
1. J Geriatr Oncol. 2022 Sep;13(7):997-1002. doi: 10.1016/j.jgo.2022.05.013. Epub 2022 Jun 3. Real-life safety of PD-1 and PD-L1 inhibitors in older patients with cancer: An observational study. Storm BN(1), Abedian Kalkhoran H(2), Wilms EB(2), Brocken P(3), Codrington H(3), Houtsma D(4), Portielje JEA(5), de Glas N(5), van der Ziel D(5), van den Bos F(6), Visser LE(7). Author information: (1)Department of Pharmacy, Haga Teaching Hospital, The Hague, the Netherlands. Electronic address: [email protected]. (2)Department of Pharmacy, Haga Teaching Hospital, The Hague, the Netherlands. (3)Department of Pulmonary Diseases - Pulmonic Oncology, Haga Teaching Hospital, The Hague, the Netherlands. (4)Department of Internal Medicine - Medical Oncology, Haga Teaching Hospital, The Hague, the Netherlands. (5)Department of Internal Medicine - Medical Oncology, University Medical Centre Leiden, Leiden, the Netherlands. (6)Department of Gerontology & Geriatrics, University Medical Centre Leiden, Leiden, the Netherlands. (7)Department of Pharmacy, Haga Teaching Hospital, The Hague, the Netherlands; Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands. INTRODUCTION: To compare the real-world safety profile of programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1) inhibitors between younger and older patients. MATERIALS AND METHODS: All patients receiving pembrolizumab, nivolumab, atezolizumab or durvalumab between September 2016 and September 2019 at Haga Teaching Hospital, The Hague, The Netherlands were included in this retrospective study. Immune-related adverse drug reactions (irADRs) were manually retrieved from the electronic patient files. The cumulative incidence of irADRs were compared between younger (<65 years) and older (≥65 years) patients using a Pearsons Chi-square test. RESULTS: We identified 217 patients who were treated with at least one dose of PD-(L)1 inhibitor. 58% were 65 years or older at the start of immunotherapy. 183 patients (84.3%) received monotherapy PD-(L)1 inhibitors and 34 (15.7%) received chemo-immunotherapy. A total of 278 irADRs were registered. Cutaneous irADRs (53.9%), thyroid gland disorders (20.3%), and non-infectious diarrhoea/colitis (17.5%) were the most frequently reported irADRs. The majority of the irADRs were mild to moderate and no fatal irADRs were observed. 61 (21.9%) of the irADRs needed systemic treatment, of which 19 (6.8%) required treatment with corticosteroids. 18 irADRs (6.5%) were severe and resulted in hospitalisation. The cumulative incidence of cutaneous irADRs was different between the age groups: 45.7% of the patients <65 years and in 60.0% of the patients ≥65 years (p = 0.036). No statistical difference was found in the cumulative incidence of other irADRs between the two age groups. DISCUSSION: Advanced age is not associated with immune-related adverse drug reactions of PD-1 and PD-L1 inhibitors. Copyright © 2022 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.jgo.2022.05.013 PMID: 35668012 [Indexed for MEDLINE] Conflict of interest statement: Declaration of Competing Interest None.
http://www.ncbi.nlm.nih.gov/pubmed/20587542
1. Int Immunol. 2010 Aug;22(8):651-60. doi: 10.1093/intimm/dxq049. Epub 2010 Jun 29. PD-L1 and PD-L2 differ in their molecular mechanisms of interaction with PD-1. Ghiotto M(1), Gauthier L, Serriari N, Pastor S, Truneh A, Nunès JA, Olive D. Author information: (1)Institut National de la Santé et de la Recherche Médicale, Unité 891, Centre de Recherche en Cancérologie de Marseille, Marseille, France. The programmed death-1 (PD-1) molecule is involved in peripheral tolerance and in the immune escape mechanisms during chronic viral infections and cancer. PD-1 interacts with two ligands, PD-L1 and PD-L2. We have investigated the molecular mechanisms of PD-1 interactions with its ligands by surface plasmon resonance and cell surface binding as well as the ability of the two ligands to compete for PD-1 binding. PD-L1 and PD-L2 bound PD-1 with comparable affinities, but striking differences were observed at the level of the association and dissociation characteristics. PD-L1, but not PD-L2, had a delayed interaction reminiscent of a phenomenon of conformational transition. These mechanisms were confirmed by using PD-L1 mAbs that delayed the dissociation of PD-L1 from PD-1. This mechanism was not restricted to PD-1 binding since PD-L1 behaved in a similar manner with its second ligand, CD80. Finally, we could demonstrate that PD-L1 and PD-L2 competed for PD-1 binding and conversely, an antagonist PD-1 mAb blocked both PD-L1 and PD-L2 binding to PD-1 and strongly enhanced T-cell proliferation. These data further emphasize the differential molecular mechanisms of interaction of PD-L1 and PD-L2 with PD-1, and suggest possible new approach for the therapy of chronic infection, cancer and transplantation. DOI: 10.1093/intimm/dxq049 PMCID: PMC3168865 PMID: 20587542 [Indexed for MEDLINE] Conflict of interest statement: CONFLICT OF INTEREST The authors declare no financial or commercial conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/30851544
1. Leuk Res. 2019 Apr;79:52-59. doi: 10.1016/j.leukres.2019.02.010. Epub 2019 Feb 28. Programmed Cell Death Receptor (PD-1) Ligand (PD-L1) expression in Philadelphia chromosome-negative myeloproliferative neoplasms. Wang JC(1), Chen C(2), Kundra A(2), Kodali S(2), Pandey A(2), Wong C(2), Cheung T(2), Gotlieb V(2), Joseph G(2), Tribie S(2). Author information: (1)Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA. Electronic address: [email protected]. (2)Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA. Programmed Cell Death Receptor (PD-1) and its Ligand (PD-L1) pathway inhibitor therapy has been explored in the field of oncology treatment mainly for solid tumors. In hematologic malignancies, there is limited information except for Hodgkin's lymphoma, and there is even less information regarding myeloproliferative neoplasm (MPN). Therefore, we explored this by first measuring PD-1 and PD-L1 levels (percentage of positive cells) in 63 patients with Philadelphia chromosome-negative MPN (Ph(-) MPN), including 16 MF (12 PMF, 2 post-PV-MF, 2 post-ET-MF), 29 ET, and 18 PV. We found there was no significant difference in PD-1 or PD-L1 levels between the different MPN groups but that there was a significant difference when PV, ET and MF were grouped as MPN and compared with controls, of all immune cells including CD4+, CD8+, CD14+ and CD34+ progenitor cells. We further found a higher incidence of higher expression levels (more than 50% of cells with positive expression) of PD-1 and PD-L1 (20% and 26%, respectively) in the CD34+ cells; in contrast, we found a low incidence (0.08-1.8%) in the immune cells in MPN patients. PD-1 and PD-L1 levels were also measured by MFI methods, and we obtained similar results except the measurements by percentage appeared to be more sensitive than the MFI methods. We found no correlation between PD-1 and PD-L1 expression levels and clinical features including WBC, platelet counts, hemoglobin levels, presence or absence of the JAK2, MPL, or CALR gene mutation, or splenomegaly. Since MPN represents stem cell disorders, the presence of elevated expression of PD-1 and PD-L1 in these cells suggests that the exploration of PD-1 and PD-L1 pathway inhibitor therapy may be worthwhile in Ph(-) MPN. Copyright © 2019 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.leukres.2019.02.010 PMID: 30851544 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31436392
1. Cancer Med. 2019 Oct;8(13):5969-5978. doi: 10.1002/cam4.2510. Epub 2019 Aug 22. The effects and safety of PD-1/PD-L1 inhibitors on head and neck cancer: A systematic review and meta-analysis. Wang BC(1), Cao RB(1), Li PD(1), Fu C(2). Author information: (1)Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (2)Department of Dermatology, The First Hospital of Wuhan, Wuhan, China. BACKGROUND: Inhibitors of programmed cell death-1 (PD-1) and its ligand (PD-L1) have been increasingly used in head and neck cancer therapy and reported to improve the outcomes with an acceptable safety profile. This systematic review and meta-analysis was conducted to assess the benefit and risk of PD-1/PD-L1 inhibitors in patients with head and neck cancer. METHOD: The PubMed, Cochrane Library, EMBASE and Web of Science databases were systematically searched to find potentially eligible studies up to May 30, 2019. Primary outcomes were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR) and adverse events. RESULTS: Overall, this analysis consisted of nine eligible studies, with two randomized controlled trials and seven single arm trials. In the treatment of recurrent or metastatic head and neck cancer, PD-1 inhibitors showed significantly lower relative risk of death than standard-of-care therapy (odds ratio [OR] = 0.60, 95% confidence interval [CI]: 0.44-0.82, I2  = 0%, P = .001). Programmed cell death-1 inhibitors also decreased the risk of disease progression, however, there was no statistically significant difference of PFS between the treatments (OR = 0.69, 95% CI: 0.48-1.01, I2  = 0%, P = .05). Subgroup analysis showed that human papillomavirus (HPV) positive patients had higher response rates than HPV negative patients in PD-1/PD-L1 inhibitors-treated population (ORR: 18.8% vs 12.2%; DCR: 42.8% vs 34.4%). The most common any-grade and grade ≥3 treatment-related adverse events were fatigue (14.7%, 95% CI: 12.3%-17.1%) and aspartate aminotransferase increased (1.6%, 95% CI: 0.3%-2.9%), respectively. CONCLUSION: Programmed cell death-1 inhibitors prolonged OS in comparison with standard-of-care therapy in recurrent or metastatic head and neck cancer patients. Human papillomavirus positive patients were superior to HPV negative patients in the treatment of PD-1/PD-L1 inhibitors. More phase III randomized controlled trials are warranted to confirm our findings. © 2019 The Authors. Cancer Medicine published by John Wiley & Sons Ltd. DOI: 10.1002/cam4.2510 PMCID: PMC6792498 PMID: 31436392 [Indexed for MEDLINE] Conflict of interest statement: None declared.
http://www.ncbi.nlm.nih.gov/pubmed/27903604
1. J Exp Med. 2016 Dec 12;213(13):2835-2840. doi: 10.1084/jem.20161462. Epub 2016 Nov 30. What does PD-L1 positive or negative mean? Ribas A(1)(2), Hu-Lieskovan S(3)(2). Author information: (1)Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095 [email protected]. (2)Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles (UCLA), Los Angeles, CA 90095. (3)Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095. Expression of the programmed death-1 (PD-1) ligand 1 (PD-L1) is used to select patients and analyze responses to anti-PD-1/L1 antibodies. The expression of PD-L1 is regulated in different ways, which leads to a different significance of its presence or absence. PD-L1 positivity may be a result of genetic events leading to constitutive PD-L1 expression on cancer cells or inducible PD-L1 expression on cancer cells and noncancer cells in response to a T cell infiltrate. A tumor may be PD-L1 negative because it has no T cell infiltrate, which may be reversed with an immune response. Finally, a tumor that is unable to express PD-L1 because of a genetic event will always be negative for PD-L1 on cancer cells. © 2016 Ribas and Hu-Lieskovan. DOI: 10.1084/jem.20161462 PMCID: PMC5154949 PMID: 27903604 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30519815
1. Target Oncol. 2018 Dec;13(6):769-777. doi: 10.1007/s11523-018-0610-1. PD-1 and PD-L1 Expression in Male Breast Cancer in Comparison with Female Breast Cancer. Manson QF(1), Ter Hoeve ND(1), Buerger H(2), Moelans CB(1), van Diest PJ(3). Author information: (1)Department of Pathology, University Medical Center Utrecht, PO Box 85500, 3508 GA, Utrecht, The Netherlands. (2)Institute of Pathology Paderborn/Höxter, Cooperative Breast Center, Paderborn, Germany. (3)Department of Pathology, University Medical Center Utrecht, PO Box 85500, 3508 GA, Utrecht, The Netherlands. [email protected]. BACKGROUND: Male breast cancer is rare, as it represents less than 1% of all breast cancer cases. In addition, male breast cancer appears to have a different biology than female breast cancer. Programmed death-1 (PD-1) and its ligand, programmed death-ligand 1 (PD-L1), seem to have prognostic and predictive values in a variety of cancers, including female breast cancer. However, the role of PD-1 and PD-L1 expression in male breast cancer has not yet been studied. OBJECTIVES: To compare PD-1 and PD-L1 expression in male breast cancer to female breast cancer and to evaluate prognostic values in both groups. PATIENTS AND METHODS: Tissue microarrays from formalin-fixed paraffin-embedded resection material of 247 female and 164 male breast cancer patients were stained for PD-1 and PD-L1 by immunohistochemistry. RESULTS: PD-1 expression on tumor-infiltrating lymphocytes was significantly less frequent in male than in female cancers (48.9 vs. 65.3%, p = 0.002). In contrast, PD-L1 expression on tumor and immune cells did not differ between the two groups. In male breast cancer, PD-1 and tumor PD-L1 were associated with grade 3 tumors. In female breast cancer, PD-1 and PD-L1 were associated with comparably worse clinicopathological variables. In a survival analysis, no prognostic value was observed for PD-1 and PD-L1 in either male and female breast cancer. In a subgroup analysis, female patients with grade 3/tumor PD-L1-negative or ER-negative/immune PD-L1-negative tumors had worse overall survival. CONCLUSIONS: PD-1 seems to be less often expressed in male breast cancer compared to female breast cancer. Although PD-1 and PD-L1 are not definite indicators for good or bad responses, male breast cancer patients may therefore respond differently to checkpoint immunotherapy with PD-1 inhibitors than female patients. DOI: 10.1007/s11523-018-0610-1 PMCID: PMC6297201 PMID: 30519815 [Indexed for MEDLINE] Conflict of interest statement: Quirine F. Manson, Natalie D. ter Hoeve, Horst Buerger, Cathy B. Moelans, and Paul J. van Diest, declare that they have no conflicts of interest that might be relevant to the contents of this manuscript.
http://www.ncbi.nlm.nih.gov/pubmed/21118528
1. Crit Care. 2010;14(6):R220. doi: 10.1186/cc9354. Epub 2010 Nov 30. PD-L1 blockade improves survival in experimental sepsis by inhibiting lymphocyte apoptosis and reversing monocyte dysfunction. Zhang Y(1), Zhou Y, Lou J, Li J, Bo L, Zhu K, Wan X, Deng X, Cai Z. Author information: (1)Clinical Research Center, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, PR China. INTRODUCTION: Lymphocyte apoptosis and monocyte dysfunction play a pivotal role in sepsis-induced immunosuppression. Programmed death-1 (PD1) and its ligand programmed death ligand-1 (PD-L1) exert inhibitory function by regulating the balance among T cell activation, tolerance, and immunopathology. PD-1 deficiency or blockade has been shown to improve survival in murine sepsis. However, PD-L1 and PD-1 differ in their expression patterns and the role of PD-L1 in sepsis-induced immunosuppression is still unknown. METHODS: Sepsis was induced in adult C57BL/6 male mice via cecal ligation and puncture (CLP). The expression of PD-1 and PD-L1 expression on peripheral T cells, B cells and monocytes were measured 24 hours after CLP or sham surgery. Additionally, the effects of anti-PD-L1 antibody on lymphocyte number, apoptosis of spleen and thymus, activities of caspase-8 and caspase-9, cytokine production, bacterial clearance, and survival were determined. RESULTS: Expression of PD-1 on T cells, B cells and monocytes and PD-L1 on B cells and monocytes were up-regulated in septic animals compared to sham-operated controls. PD-L1 blockade significantly improved survival of CLP mice. Anti-PD-L1 antibody administration prevented sepsis-induced depletion of lymphocytes, increased tumor necrosis factor (TNF)-α and interleukin (IL)-6 production, decreased IL-10 production, and enhanced bacterial clearance. CONCLUSIONS: PD-L1 blockade exerts a protective effect on sepsis at least partly by inhibiting lymphocyte apoptosis and reversing monocyte dysfunction. Anti-PD-L1 antibody administration may be a promising therapeutic strategy for sepsis-induced immunosuppression. DOI: 10.1186/cc9354 PMCID: PMC3220038 PMID: 21118528 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16606670
1. J Exp Med. 2006 Apr 17;203(4):883-95. doi: 10.1084/jem.20051776. Epub 2006 Apr 10. Tissue expression of PD-L1 mediates peripheral T cell tolerance. Keir ME(1), Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA, Koulmanda M, Freeman GJ, Sayegh MH, Sharpe AH. Author information: (1)Department of Pathology, Brigham and Women's Hospital and Children's Hospital Boston, MA 02115, USA. Comment in J Exp Med. 2006 Apr 17;203(4):817-20. doi: 10.1084/jem.20060219. Programmed death 1 (PD-1), an inhibitory receptor expressed on activated lymphocytes, regulates tolerance and autoimmunity. PD-1 has two ligands: PD-1 ligand 1 (PD-L1), which is expressed broadly on hematopoietic and parenchymal cells, including pancreatic islet cells; and PD-L2, which is restricted to macrophages and dendritic cells. To investigate whether PD-L1 and PD-L2 have synergistic or unique roles in regulating T cell activation and tolerance, we generated mice lacking PD-L1 and PD-L2 (PD-L1/PD-L2(-/-) mice) and compared them to mice lacking either PD-L. PD-L1 and PD-L2 have overlapping functions in inhibiting interleukin-2 and interferon-gamma production during T cell activation. However, PD-L1 has a unique and critical role in controlling self-reactive T cells in the pancreas. Our studies with bone marrow chimeras demonstrate that PD-L1/PD-L2 expression only on antigen-presenting cells is insufficient to prevent the early onset diabetes that develops in PD-L1/PD-L2(-/-) non-obese diabetic mice. PD-L1 expression in islets protects against immunopathology after transplantation of syngeneic islets into diabetic recipients. PD-L1 inhibits pathogenic self-reactive CD4+ T cell-mediated tissue destruction and effector cytokine production. These data provide evidence that PD-L1 expression on parenchymal cells rather than hematopoietic cells protects against autoimmune diabetes and point to a novel role for PD-1-PD-L1 interactions in mediating tissue tolerance. DOI: 10.1084/jem.20051776 PMCID: PMC2118286 PMID: 16606670 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31876895
1. JAMA Oncol. 2020 Mar 1;6(3):375-384. doi: 10.1001/jamaoncol.2019.5367. Use of Immunotherapy With Programmed Cell Death 1 vs Programmed Cell Death Ligand 1 Inhibitors in Patients With Cancer: A Systematic Review and Meta-analysis. Duan J(1), Cui L(2), Zhao X(2), Bai H(1), Cai S(2), Wang G(2), Zhao Z(2), Zhao J(2), Chen S(2), Song J(2), Qi C(2), Wang Q(2), Huang M(2), Zhang Y(2), Huang D(2), Bai Y(2), Sun F(3), Lee JJ(4), Wang Z(1), Wang J(1). Author information: (1)State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. (2)The Medical Department, 3D Medicines Inc, Shanghai, China. (3)School of Public Health, Department of Epidemiology and Biostatistics, Peking University Health Science Centre, Beijing, China. (4)Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston. Comment in JAMA Oncol. 2020 Jul 1;6(7):1116-1117. doi: 10.1001/jamaoncol.2020.0646. JAMA Oncol. 2020 Jul 1;6(7):1115-1116. doi: 10.1001/jamaoncol.2020.0637. JAMA Oncol. 2020 Jul 1;6(7):1115. doi: 10.1001/jamaoncol.2020.0631. JAMA Oncol. 2020 Jul 1;6(7):1114-1115. doi: 10.1001/jamaoncol.2020.0628. JAMA Oncol. 2020 Jul 1;6(7):1113-1114. doi: 10.1001/jamaoncol.2020.0625. IMPORTANCE: Immune checkpoint inhibitors of programmed cell death 1 (PD-1) and its ligand (PD-L1) have led to a paradigm shift in cancer treatment. Understanding the clinical efficacy and safety profile of these drugs is necessary for treatment strategy in clinical practice. OBJECTIVE: To assess the differences between anti-PD-1 and anti-PD-L1 regarding efficacy and safety shown in randomized clinical trials across various tumor types. DATA SOURCES: Systematic searches of PubMed, Cochrane CENTRAL, and Embase were conducted from January 1, 2000, to March 1, 2019. In addition, abstracts and presentations from all major conference proceedings were reviewed. STUDY SELECTION: All randomized clinical trials that compared anti-PD-1 and anti-PD-L1 with standard treatment in patients with cancer were selected as candidates. Retrospective studies, single-arm phase 1/2 studies, and trials comparing anti-PD-1 and anti-PD-L1 with other immunotherapies were excluded. Studies of anti-PD-1 and anti-PD-L1 therapy were screened and paired by the matching of clinical characteristics as mirror groups. DATA EXTRACTION AND SYNTHESIS: Three investigators independently extracted data from each study following the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-analyses) guideline. Trial names, first author, year of publication, study design, National Clinical Trial identifier number, blinding status, study phase, pathologic characteristics, number of patients, patients' age and sex distribution, Eastern Cooperative Oncology Group Performance Status, lines of treatment, study drugs, biomarker status, follow-up time, incidence of adverse events, and hazard ratios (HRs) with 95% CIs for overall survival and progression-free survival were extracted. A random-effects model was applied for data analysis. MAIN OUTCOMES AND MEASURES: Differences in OS between anti-PD-1 and anti-PD-L1 across different cancer types were assessed. An effect size was derived from each mirror group and then pooled across all groups using a random-effects model. RESULTS: Nineteen randomized clinical trials involving 11 379 patients were included in the meta-analysis. Overall, anti-PD-1 exhibited superior overall survival (HR, 0.75; 95% CI, 0.65-0.86; P < .001) and progression-free survival (HR, 0.73; 95% CI, 0.56-0.96; P = .02) compared with anti-PD-L1. No significant difference was observed in their safety profiles. Sensitivity analysis presented consistency in the overall estimates across these analyses. Consistent results were observed through frequentist and bayesian approaches with the same studies. CONCLUSIONS AND RELEVANCE: Comprehensive analysis suggests that anti-PD-1 exhibited favorable survival outcomes and a safety profile comparable to that of anti-PD-L1, which may provide a useful guide for clinicians. DOI: 10.1001/jamaoncol.2019.5367 PMCID: PMC6990765 PMID: 31876895 [Indexed for MEDLINE] Conflict of interest statement: Conflict of Interest Disclosures: Drs Cui, X. Zhao, Cai, G. Wang, Z. Zhao, J. Zhao, Chen, Song, Qi, Q. Wang, M. Huang, Zhang, D. Huang, and Bai are employees of 3D Medicines Inc. No other conflicts were reported.
http://www.ncbi.nlm.nih.gov/pubmed/21097698
1. J Leukoc Biol. 2011 Apr;89(4):507-15. doi: 10.1189/jlb.0610327. Epub 2010 Nov 19. Expression of PD-L1 and PD-L2 on human macrophages is up-regulated by HIV-1 and differentially modulated by IL-10. Rodríguez-García M(1), Porichis F, de Jong OG, Levi K, Diefenbach TJ, Lifson JD, Freeman GJ, Walker BD, Kaufmann DE, Kavanagh DG. Author information: (1)Ragon Institute of MGH, MIT and Harvard University, 149 13th St., Charlestown, MA 02129, USA. Comment in J Leukoc Biol. 89:495. PD-1 plays an important role in T cell exhaustion during HIV infection. PD-1 has two ligands: PD-L1, expressed on hematopoietic and nonhematopoietic cells, and PD-L2, limited to DCs and macrophages. Little is known about PD-L1 expression and regulation in human macrophages. Previous reports have found few immediate effects of macrophage exposure to HIV, suggesting that macrophages lack PRRs for this virus. Using quantitative confocal microscopy and a multiplexed cytokine bead array, we measured induction of PD-L1, PD-L2, and innate response cytokines in human MDMs in response to chemically inactivated HIV virions. Consistent with previous reports, no cytokines were induced by HIV virion exposure. Whereas PD-L1 and PD-L2 had low baseline expression, TLR ligands (LPS and CL097) up-regulated PD-L1 but not PD-L2. Unlike what we found for cytokine expression, PD-L1 and PD-L2 were up-regulated in response to exposure with inactivated HIV virions or with replication-competent HIV. Expression of PD-L1 was differentially modulated by IL-10, which induced up-regulation of PD-L1 but not of PD-L2, and IL-10 blockade enhanced only PD-L2 expression. We discuss implications for innate recognition of HIV by macrophages and potential, different roles for PD-L1 and PD-L2 in immunity and pathogenesis. DOI: 10.1189/jlb.0610327 PMCID: PMC3058820 PMID: 21097698 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28960263
1. Cancer. 2018 Jan 15;124(2):271-277. doi: 10.1002/cncr.31043. Epub 2017 Sep 28. Comparison of the toxicity profile of PD-1 versus PD-L1 inhibitors in non-small cell lung cancer: A systematic analysis of the literature. Pillai RN(1), Behera M(1), Owonikoko TK(1), Kamphorst AO(2), Pakkala S(1), Belani CP(3), Khuri FR(1), Ahmed R(2), Ramalingam SS(1). Author information: (1)Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia. (2)Emory Vaccine Center, Department of Microbiology and Immunology, Emory University, Atlanta, Georgia. (3)Pennsylvania University, Penn State Hershey Cancer Institute, Hershey, Pennsylvania. Comment in J Thorac Dis. 2018 Nov;10(Suppl 33):S4034-S4037. doi: 10.21037/jtd.2018.09.46. J Thorac Dis. 2018 Nov;10(Suppl 33):S4065-S4068. doi: 10.21037/jtd.2018.09.83. J Thorac Dis. 2018 Nov;10(Suppl 33):S4069-S4072. doi: 10.21037/jtd.2018.09.102. J Thorac Dis. 2018 Nov;10(Suppl 33):S4082-S4084. doi: 10.21037/jtd.2018.09.103. BACKGROUND: Monoclonal antibodies against programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) are effective therapies in patients with non-small cell lung cancer (NSCLC). Herein, the authors performed a systematic review investigating differences in the toxicities of PD-1 and PD-L1 inhibitors. METHODS: An electronic literature search was performed of public databases (MEDLINE, Excerpta Medica dataBASE [EMBASE], and Cochrane) and conference proceedings for trials using PD-1 inhibitors (nivolumab and pembrolizumab) and PD-L1 inhibitors (atezolizumab, durvalumab, and avelumab) in patients with NSCLC. A formal systematic analysis was conducted with Comprehensive Meta-Analysis software (version 2.2). Clinical and demographic characteristics, response, and toxicity data were compared between both groups. RESULTS: A total of 23 studies reported between 2013 and 2016 were eligible for analysis. The total number of patients evaluated for toxicities was 3284 patients in the PD-1 group and 2460 patients in the PD-L1 group. The baseline patient characteristics of the 2 groups were similar, although there was a trend toward increased squamous histology in the group treated with PD-L1 (32% vs 25%; P = .6). There was no difference in response rate noted between PD-1 (19%) and PD-L1 (18.6%) inhibitors (P = .17). The incidence of overall adverse events (AEs) was comparable between the PD-1 and PD-L1 inhibitors (64% [95% confidence interval (95% CI), 63%-66%] vs 66% [95% CI, 65%-69%]; P = .8). Fatigue was the most frequently reported AE with both classes of drugs. Patients treated with PD-1 inhibitors were found to have a slightly increased rate of immune-related AEs (16% [95% CI, 14%-17%] vs 11% [95% CI, 10%-13%]; P = .07) and pneumonitis (4% [95% CI, 3%-5%] vs 2% [95% CI, 1%-3%]; P = .01) compared with patients who received PD-L1 inhibitors. CONCLUSIONS: In this systematic review involving 5744 patients with NSCLC, the toxicity and efficacy profiles of PD-1 and PD-L1 inhibitors appear to be similar. Cancer 2018;124:271-7. © 2017 American Cancer Society. © 2017 American Cancer Society. DOI: 10.1002/cncr.31043 PMCID: PMC5761314 PMID: 28960263 [Indexed for MEDLINE] Conflict of interest statement: Disclosures: None of the authors have any conflicts of interest to report.
http://www.ncbi.nlm.nih.gov/pubmed/17924994
1. Am J Transplant. 2007 Dec;7(12):2683-92. doi: 10.1111/j.1600-6143.2007.01999.x. Epub 2007 Oct 9. Striking dichotomy of PD-L1 and PD-L2 pathways in regulating alloreactive CD4(+) and CD8(+) T cells in vivo. Habicht A(1), Kewalaramani R, Vu MD, Demirci G, Blazar BR, Sayegh MH, Li XC. Author information: (1)Transplantation Research Center, Brigham and Women's Hospital and the Children's Hospital of Boston, Boston, MA, USA. Programmed death-1 (PD-1) is a recently identified coinhibitory molecule that belongs to the CD28 superfamily. PD-1 has two ligands PD-L1 and PD-L2. There is some evidence that PD-L1 and PD-L2 serve distinct functions, but their exact function in alloimmunity remains unclear. In the present study, we used a GVHD-like model that allows detailed analyses of T-cell activation at a single cell level in vivo to examine the role of PD-1/PD-L1 and PD-1/PD-L2 interactions in regulating proliferation of CD4(+) and CD8(+) T cells in response to alloantigen stimulation. We found that both CD4(+) and CD8(+) T cells proliferated vigorously in vivo and that PD-L1 and PD-L2 exhibit strikingly different effect on T-cell proliferation. While blocking PD-L1 did not affect the in vivo proliferation of CD4(+) and CD8(+) T cells regardless of CD28 costimulation, blocking PD-L2 resulted in a marked increase in the responder frequency of CD8(+) T-cells in vivo. The effect of PD-L2 on the CD8(+) T-cell proliferation is regulated by CD28 costimulation and by the CD4(+) T cells. We conclude that PD-L1 and PD-L2 function differently in regulating alloreactive T-cell activation in vivo, and PD-L2 is predominant in this model in limiting alloreactive CD8(+) T-cell proliferation. DOI: 10.1111/j.1600-6143.2007.01999.x PMID: 17924994 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30547271
1. Clin Exp Metastasis. 2019 Feb;36(1):29-37. doi: 10.1007/s10585-018-9950-6. Epub 2018 Dec 13. Frequent discordance in PD-1 and PD-L1 expression between primary breast tumors and their matched distant metastases. Manson QF(1), Schrijver WAME(1), Ter Hoeve ND(1), Moelans CB(1), van Diest PJ(2). Author information: (1)Department of Pathology, University Medical Center Utrecht, PO Box 85500, 3508 GA, Utrecht, The Netherlands. (2)Department of Pathology, University Medical Center Utrecht, PO Box 85500, 3508 GA, Utrecht, The Netherlands. [email protected]. Programmed death-1 (PD-1) is an immune checkpoint that is able to inhibit the immune system by binding to its ligand programmed death-ligand 1 (PD-L1). In many cancer types, among which breast cancer, prognostic and/or predictive values have been suggested for both PD-1 and PD-L1. Previous research has demonstrated discrepancies in PD-L1 expression between primary breast tumors and distant metastases, however data so far have been scarce. We therefore evaluated immunohistochemical expression levels of PD-1 and PD-L1 in primary breast tumors and their paired distant metastases, and evaluated prognostic values. Tissue microarrays from formalin-fixed paraffin-embedded resection specimens of primary breast cancers and their matched distant metastases were immunohistochemically stained for PD-1 and PD-L1. PD-1 was available in both primary tumor and metastasis in 82 patients, and PD-L1 in 49 patients. PD-1 was discrepant between primary tumor and metastasis in half of the patients (50%), PD-L1 on tumor cells was discrepant in 28.5%, and PD-L1 on immune cells in 40.8% of the patients. In primary tumors there was a correlation between PD-1 positivity and a higher tumor grade, and between immune PD-L1 and ER negativity. In survival analyses, a significantly better overall survival was observed for patients with PD-L1 negative primary breast tumors that developed PD-L1 positive distant metastases (HR 3.013, CI 1.201-7.561, p = 0.019). To conclude, PD-1 and tumor and immune PD-L1 seem to be discordantly expressed between primary tumors and their matched distant metastases in about one-third to a half of the breast cancer patients. Further, gained expression of PD-L1 in metastases seems to indicate better survival. This illustrates the need of reassessing PD-1 and PD-L1 expression on biopsies of distant metastases to optimize the usefulness of these biomarkers. DOI: 10.1007/s10585-018-9950-6 PMCID: PMC6394593 PMID: 30547271 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/12697896
1. Proc Natl Acad Sci U S A. 2003 Apr 29;100(9):5336-41. doi: 10.1073/pnas.0931259100. Epub 2003 Apr 15. PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Loke P(1), Allison JP. Author information: (1)Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA. PD-L1 and PD-L2 are ligands for PD-1, a costimulatory molecule that plays an inhibitory role in regulating T cell activation in the periphery. We find that PD-L1 is highly expressed on inflammatory macrophages as compared with resident peritoneal macrophages but can be induced on resident macrophages by classical activation stimuli such as lipopolysaccharide, IFN-gamma, and polyinosinic-polycytidylic acid. Further up-regulation of PD-L1 on inflammatory macrophages can also be induced by subsequent exposure to lipopolysaccharide and IFN-gamma. In contrast, PD-L2 is not expressed on inflammatory macrophages but can be induced by alternative activation via IL-4. Although PD-L1 is highly inducible on a variety of antigen-presenting cell lines as well as resident macrophages, PD-L2 is most significantly inducible only on inflammatory macrophages. PD-L1 up-regulation depends on TLR4 and STAT1, whereas PD-L2 expression depends on IL-4R alpha and STAT6. Consistent with these results, T helper 1T helper 2 (Th1/Th2) cells also differentially up-regulate PD-L1 and PD-L2 expression on inflammatory macrophages. Hence, Th1 cells as well as microbial products can enhance PD-L1 expression on many different macrophage populations, whereas Th2 cells instruct only inflammatory macrophages to up-regulate PD-L2. These results suggest that PD-L1 and PD-L2 might have different functions in regulating type 1 and type 2 responses. DOI: 10.1073/pnas.0931259100 PMCID: PMC154346 PMID: 12697896 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32770212
1. Adv Nutr. 2020 Nov 16;11(6):1405-1413. doi: 10.1093/advances/nmaa089. The "Virtual Digital Twins" Concept in Precision Nutrition. Gkouskou K(1)(2), Vlastos I(1), Karkalousos P(3), Chaniotis D(3), Sanoudou D(4)(5)(6), Eliopoulos AG(1)(5)(6). Author information: (1)Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece. (2)Embiodiagnostics, Biology Research Company, Heraklion, Crete, Greece. (3)Department of Biomedical Sciences, University of West Attica, Athens, Greece. (4)Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece. (5)Center for New Biotechnologies and Precision Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece. (6)Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece. Nutritional and lifestyle changes remain at the core of healthy aging and disease prevention. Accumulating evidence underscores the impact of genetic, metabolic, and host gut microbial factors on individual responses to nutrients, paving the way for the stratification of nutritional guidelines. However, technological advances that incorporate biological, nutritional, lifestyle, and health data at an unprecedented scale and depth conceptualize a future where preventative dietary interventions will exceed stratification and will be highly individualized. We herein discuss how genetic information combined with longitudinal metabolomic, immune, behavioral, and gut microbial parameters, and bioclinical variables could define a digital replica of oneself, a "virtual digital twin," which could serve to guide nutrition in a personalized manner. Such a model may revolutionize the management of obesity and its comorbidities, and provide a pillar for healthy aging. Copyright © The Author(s) on behalf of the American Society for Nutrition 2020. DOI: 10.1093/advances/nmaa089 PMCID: PMC7666894 PMID: 32770212 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34536404
1. Lancet Neurol. 2021 Oct;20(10):821-831. doi: 10.1016/S1474-4422(21)00242-8. Safety and efficacy of oral levosimendan in people with amyotrophic lateral sclerosis (the REFALS study): a randomised, double-blind, placebo-controlled phase 3 trial. Cudkowicz M(1), Genge A(2), Maragakis N(3), Petri S(4), van den Berg L(5), Aho VV(6), Sarapohja T(6), Kuoppamäki M(6), Garratt C(6), Al-Chalabi A(7); REFALS investigators. Collaborators: Kiernan M, Mathers S, Henderson R, Needham M, Schultz D, Löscher W, Mitrovic N, Rath J, Damme PV, De Bleecker JL, Delstanche S, Johnston W, Zinman L, O'Connell C, Matte G, Dionne A, Korngut L, Turnbull J, Laaksovirta H, Jokela M, Tapiola T, Soriani MH, Couratier P, Camu W, Corcia P, Ludolph A, Großkreutz J, Meyer T, Boentert M, Schrank B, Prudlo J, Untucht R, Hardiman O, Siciliano G, Chio' A, Mazzini L, Inghilleri M, Caponnetto C, Mora G, Mora Pardina JS, Farrero Munoz E, Vázquez Costa JF, Aguera Morales E, Varona L, Andersen P, Ingre C, Johansson R, Radunovic A, Young C, Babu S, Shaibani A, Staff N, Vu T, Rivner M, Scelsa S, Sivakumar K, Waheed W, Heitzman D, Rana S, Pattee G, Ajroud-Driss S, Bayat E, Kasarskis E, Lange DJ, Elliott M, Harris B, Felice K, Pulley MT, Kwan J, Brown M, Ravits J, Burford M, Karam C, Miller T, Andrews J, Levine T, Locatelli E, Wymer J, Bedlack R, Fee D, Goyal N, Oskarsson B, McCluskey L, Caress J, Weiss M, Quick A, Bromberg M, Lacomis D, Goutman S, Rezania K, Guliani G, Goslin K, Katz JS. Author information: (1)Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA. Electronic address: [email protected]. (2)Clinical Research Unit and ALS clinic, Montreal Neurological Institute and Hospital, Montreal, QC, Canada. (3)Department of Neurology, Johns Hopkins University, Baltimore, MD, USA. (4)Medizinische Hochschule Hannover, Hannover, Germany. (5)Department of Neurology, University Medical Center Utrecht, Utrecht, Netherlands. (6)Orion Corporation, Espoo, Finland. (7)Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, and Department of Neurology, King's College Hospital, King's College London, London, UK. Comment in Nat Rev Neurol. 2021 Nov;17(11):660. doi: 10.1038/s41582-021-00569-7. Comment on Lancet Neurol. 2021 Oct;20(10):775-777. doi: 10.1016/S1474-4422(21)00255-6. BACKGROUND: There is an urgent unmet need for new therapies in amyotrophic lateral sclerosis. In a clinical study with healthy volunteers, levosimendan, a calcium sensitiser, was shown to improve neuromechanical efficiency and contractile function of the human diaphragm. We aimed to evaluate the safety and efficacy of oral levosimendan in people with amyotrophic lateral sclerosis, with a focus on respiratory function. METHODS: The REFALS study is a randomised, double-blind, placebo-controlled phase 3 trial at 99 amyotrophic lateral sclerosis specialist centres in 14 countries worldwide. People with amyotrophic lateral sclerosis were eligible for participation if they were at least 18 years of age and had a sitting slow vital capacity (SVC) of 60-90% predicted. Participants were randomly assigned (2:1) by interactive web-response system to receive either levosimendan or placebo. The capsules for oral administration were identical in appearance to maintain blinding of participants and investigators. The primary endpoint was the change from baseline in supine SVC at 12 weeks, assessed as the percentage of predicted normal sitting SVC. The key secondary endpoint was the combined assessment of function and survival (CAFS) up to 48 weeks. Analyses were done in the intention-to-treat population, comprising all participants who were randomly assigned. This trial is registered at ClinicalTrials.gov (NCT03505021) and has been completed. An extension study (REFALS-ES; NCT03948178) has also been completed, but will be reported separately. FINDINGS: Between June 21, 2018, and June 28, 2019, 871 people were screened for the study, of whom 496 were randomly assigned either levosimendan (n=329) or placebo (n=167). Participants were followed up between June 27, 2018 and June 26, 2020, for a median duration of 50·1 (IQR 37·5-51·1) weeks. The median duration of treatment was 47·9 (IQR 26·4-48·1) weeks. Change from baseline in supine SVC at 12 weeks was -6·73% with levosimendan and -6·99% with placebo, with no significant difference between the treatments (estimated treatment difference 0·26%, 95% CI -2·03 to 2·55, p=0·83). Similarly, at week 48, CAFS did not differ between treatment groups (least squares mean change from baseline 10·69, 95% CI -15·74 to 37·12; nominal p value=0·43). The most frequent adverse events were increased heart rate (106 [33%] of 326 receiving levosimendan vs 12 [7%] of 166 receiving placebo), fall (85 [26%] vs 48 [29%]), headache (93 [29%] vs 36 [22%]), and dyspnoea (59 [18%] vs 32 [19%]). 33 (10%) participants allocated levosimendan and 20 (12%) assigned placebo died during the trial, mainly due to respiratory failure or progression of amyotrophic lateral sclerosis. INTERPRETATION: Levosimendan was not superior to placebo in maintaining respiratory function in a broad population with amyotrophic lateral sclerosis. Although levosimendan was generally well tolerated, increased heart rate and headache occurred more frequently with levosimendan than with placebo. The possibility of a clinically relevant subgroup of responsive individuals requires further evaluation. FUNDING: Orion Corporation. Copyright © 2021 Elsevier Ltd. All rights reserved. DOI: 10.1016/S1474-4422(21)00242-8 PMID: 34536404 [Indexed for MEDLINE] Conflict of interest statement: Declaration of interests MC reports consultancy fees from Biohaven Pharmaceuticals, Eli Lilly, Orion Corporation, Revelescio, Wave Pharmaceuticals, Mitsubishi Tanabe Pharma, Anelixis, Aclipse, Cytokinetics, Avexis, Sunovian, Disarm, ALS Pharmaceuticals, RRD, Biogen, Takeda, outside the submitted work. AG reports personal fees from Mitsubishi Tanabe Pharma America, Sanofi Genzyme, ALS Pharma, AB Sciences, Biogen, Novartis, CSL Behring, Anavex, Avexis, Alexion, Revalesio, Roche, Cytokinetics, Orion Corporation, and Akcea, outside the submitted work. NM reports personal fees from Orion Corporation, during the conduct of the study; personal fees from Massachusetts General Hospital, Cytokinetics, Amylyx, and Orphazyme, outside the submitted work; and clinical trial site principle investigator fees from Biogen Idec, Medicinova, Anelixis, Apellis, and Helixmith. SP reports grants from the German Neuromuscular Society, Federal Ministry of Education and Research, German Israeli Foundation for scientific research and development, and EU Joint Programme for Neurodegenerative Disease Research; and speaker or medical expert honoraria from Cytokinetics, Desitin Pharma, Biogen, Novartis, Teva Pharmaceuticals, and Roche, outside of the submitted work. LvdB is on the advisory board Orion, with no financial compensation. LvdB reports personal fees from Denali, Calico, Biogen, Ferrer, and Orphazyme; and grants from Takeda, outside the submitted work. VVA, TS and CG are Employees of Orion Corporation; MK is an employee of Orion Corporation and owns Orion Corporation stocks. AA-C reports non-financial support from Orion Corporation, during the conduct of the study; consultancy fees from Mitsubishi Tanabe Pharma, Biogen Idec, Cytokinetics, Wave Pharmaceuticals, Apellis, Amylyx, Novartis, and Eli Lilly, outside the submitted work.
http://www.ncbi.nlm.nih.gov/pubmed/31315908
1. J Neurol Neurosurg Psychiatry. 2019 Oct;90(10):1165-1170. doi: 10.1136/jnnp-2018-320288. Epub 2019 Jul 17. Oral levosimendan in amyotrophic lateral sclerosis: a phase II multicentre, randomised, double-blind, placebo-controlled trial. Al-Chalabi A(1)(2), Shaw P(3), Leigh PN(4), van den Berg L(5), Hardiman O(6), Ludolph A(7), Aho VV(8), Sarapohja T(9), Kuoppamäki M(10)(11). Author information: (1)Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK. (2)Department of Neurology, King's College Hospital, London, UK. (3)Sheffield Institute for Translational Neuroscience and NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, UK. (4)Department of Neuroscience Brighton and Sussex Medical School, Trafford Centre for Biomedical Science, Falmer, Brighton, UK. (5)Departmentof Neurology, University Medical Center Utrecht, Utrecht, The Netherlands. (6)Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland. (7)Department of Neurology, University of Ulm, Ulm, Germany. (8)Orion Pharma, Orion Corporation, Turku, Finland [email protected]. (9)Orion Pharma, Orion Corporation, Espoo, Finland. (10)Orion Pharma, Orion Corporation, Turku, Finland. (11)Lundbeck, Copenhagen, Denmark. OBJECTIVE: To evaluate the efficacy and safety of oral levosimendan in patients with amyotrophic lateral sclerosis (ALS). This phase II, randomised, double-blind, placebo-controlled, crossover, three-period study with 6 months open-label follow-up enrolled adults with ALS and sitting slow vital capacity (SVC) 60%-90 % of predicted from 11 sites in four countries. METHODS: Patients received levosimendan 1 mg daily, 1 mg two times a day or placebo during three 14-day crossover periods and levosimendan 1-2 mg daily during open-label follow-up. Primary endpoint was sitting SVC; secondary endpoints included supine SVC, ALS Functional Rating Scale-Revised (ALSFRS-R), tolerability and safety. RESULTS: Of 66 patients randomised, 59 contributed to the double-blind results and 50 entered open-label follow-up. Sitting SVC was not significantly different between the treatments. In post hoc analysis using period-wise baselines, supine SVC favoured levosimendan over placebo, estimated mean differences from baseline being -3.62% on placebo, +0.77% on levosimendan 1 mg daily (p=0.018) and +2.38% on 1 mg two times a day (p=0.001). Headache occurred in 16.7% of patients during levosimendan 1 mg daily (p=0.030), 28.6% during 1 mg two times a day (p=0.002) and 3.3% during placebo. The respective frequencies for increased heart rate were 5.1% (p=0.337), 18.5% (p=0.018) and 1.7%. No significant differences between the treatments were seen for other adverse events. CONCLUSIONS: Levosimendan did not achieve the primary endpoint of improving sitting SVC in ALS. Headache and increased heart rate were increased on levosimendan, although it was otherwise well tolerated. A phase III study to evaluate the longer term effects of oral levosimendan in ALS is ongoing. © Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. DOI: 10.1136/jnnp-2018-320288 PMCID: PMC6817985 PMID: 31315908 [Indexed for MEDLINE] Conflict of interest statement: Competing interests: VVA and TS are employees of Orion Corporation. Other authors have no conflicts to disclose.
http://www.ncbi.nlm.nih.gov/pubmed/24031027
1. Carcinogenesis. 2014 Feb;35(2):424-31. doi: 10.1093/carcin/bgt305. Epub 2013 Sep 12. PD-1 blockage delays murine squamous cell carcinoma development. Belai EB(1), de Oliveira CE, Gasparoto TH, Ramos RN, Torres SA, Garlet GP, Cavassani KA, Silva JS, Campanelli AP. Author information: (1)Department of Biological Sciences and. Engagement of programmed death-1 (PD-1) with its two ligands [programmed death ligand-1 (PD-L1) and PD-L2] has been associated with the suppression of tumor-reactive T cells; however, the underlying mechanism for this T-cell dysfunction is not clear. We hypothesized that PD-1 and PD-L1 signals are, in part, responsible for squamous cell carcinoma (SCC) escape from immune antitumor regulation by modulation of the tumor environment. In the present study, we used a multistage model of SCC to examine the role of PD-1/PD-L1 activation during tumor development. Tumor sites presented an increased percentage of CD4(+) and CD8(+) T cells expressing PD-1 when compared with non-tumorigenic control mice, whereas the expression of PD-L1 was particularly increased in F4/80(+) macrophages in tumor sites. Further, the systemic immune neutralization of PD-1 resulted in a decreased number and delayed incidence rate of papillomas followed by a differential expression of cytokeratins, suggesting that the PD-1-PD-L1 interaction contributes to the progression of SCC by downregulation of antitumor responses. In fact, blocking PD-1 increased the percentage of CD8(+) and CD4(+) T cells, and the levels of interferon-γ in the tumor sites. Our results indicated involvement of PD-1(+) T cells in SCC development and in the modulation of the inflammatory immune response. DOI: 10.1093/carcin/bgt305 PMID: 24031027 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36362876
1. Life (Basel). 2022 Oct 27;12(11):1721. doi: 10.3390/life12111721. Three-Country Snapshot of Ornithine Transcarbamylase Deficiency. Seker Yilmaz B(1), Baruteau J(1)(2)(3), Arslan N(4), Aydin HI(5), Barth M(6), Bozaci AE(7), Brassier A(8), Canda E(9), Cano A(10), Chronopoulou E(11), Connolly GM(12), Damaj L(13), Dawson C(14), Dobbelaere D(15), Douillard C(15), Eminoglu FT(16), Erdol S(17), Ersoy M(18), Fang S(3), Feillet F(19), Gokcay G(20), Goksoy E(21), Gorce M(22), Inci A(23), Kadioglu B(24), Kardas F(25), Kasapkara CS(26), Kilic Yildirim G(27), Kor D(28), Kose M(29), Marelli C(30)(31), Mundy H(32), O'Sullivan S(33), Ozturk Hismi B(34), Ramachandran R(35), Roubertie A(30)(31), Sanlilar M(36), Schiff M(8), Sreekantam S(37), Stepien KM(38), Uzun Unal O(39), Yildiz Y(40), Zubarioglu T(41), Gissen P(1)(2)(3). Author information: (1)Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK. (2)National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK. (3)Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK. (4)Paediatric Metabolic Medicine Department, Dokuz Eylul University Faculty of Medicine, Izmir 35340, Turkey. (5)Paediatric Metabolic Medicine Department, Baskent University Faculty of Medicine, Ankara 06490, Turkey. (6)Centre de Référence des Maladies Héréditaires du Métabolisme, CHU Angers, 4 rue Larrey, CEDEX 9, 49933 Angers, France. (7)Paediatric Metabolic Medicine Department, Diyarbakir Children's Hospital, Diyarbakir 21100, Turkey. (8)Reference Center for Inborn Errors of Metabolism, Necker University Hospital, APHP and University of Paris Cité, 75015 Paris, France. (9)Paediatric Metabolic Medicine Department, Ege University Faculty of Medicine, Izmir 35100, Turkey. (10)Reference Center of Inherited Metabolic Disorders, Timone Enfants Hospital, 264 rue Saint-Pierre, 13005 Marseille, France. (11)Department of Inherited Metabolic Disease, Division of Women's and Children's Services, University Hospitals Bristol NHS Foundation Trust, Bristol BS1 3NU, UK. (12)Belfast Health and Social Care Trust, Belfast BT9 7AB, UK. (13)Centre de Compétence Maladies Héréditaires du Métabolisme, CHU Hôpital Sud, CEDEX 2, 35203 Rennes, France. (14)Metabolic Medicine Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, UK. (15)Medical Reference Center for Inherited Metabolic Diseases, Jeanne de Flandre University Hospital and RADEME Research Team for Rare Metabolic and Developmental Diseases, EA 7364 CHRU Lille, 59000 Lille, France. (16)Paediatric Metabolic Medicine Department, Ankara University Faculty of Medicine, Ankara 06080, Turkey. (17)Paediatric Metabolic Medicine Department, Uludag University Faculty of Medicine, Bursa 16059, Turkey. (18)Paediatric Metabolic Medicine Department, Dr Sadi Konuk Reseach & Training Hospital, Istanbul 34450, Turkey. (19)Centre de Référence des Maladies Métaboliques de Nancy, CHU Brabois Enfants, 5 Rue du Morvan, 54500 Vandœuvre-lès-Nancy, France. (20)Paediatric Metabolic Medicine Department, Istanbul University Istanbul Faculty of Medicine, Istanbul 34093, Turkey. (21)Paediatric Metabolic Medicine Department, Cengiz Gokcek Children's Hospital, Gaziantep 27010, Turkey. (22)Centre de Référence des Maladies Rares du Métabolisme, Hôpital des Enfants-CHU Toulouse, 330 Avenue de Grande-Bretagne, CEDEX 9, 31059 Toulouse, France. (23)Paediatric Metabolic Medicine Department, Gazi University Faculty of Medicine, Ankara 06500, Turkey. (24)Paediatric Metabolic Medicine Department, Konya City Hospital, Konya 42020, Turkey. (25)Paediatric Metabolic Medicine Department, Erciyes University Faculty of Medicine, Kayseri 38030, Turkey. (26)Paediatric Metabolic Medicine Department, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara 06800, Turkey. (27)Paediatric Metabolic Medicine Department, Osmangazi University Faculty of Medicine, Eskisehir 26480, Turkey. (28)Paediatric Metabolic Medicine Department, Cukurova University Faculty of Medicine, Adana 01250, Turkey. (29)Paediatric Metabolic Medicine Department, Faculty of Medicine, Izmir Katip Celebi University, Izmir 35620, Turkey. (30)MMDN, University Montpellier, EPHE, INSERM, 34090 Montpellier, France. (31)Expert Center for Metabolic and Neurogenetic Diseases, Centre Hospitalier Universitaire (CHU), 34090 Montpellier, France. (32)Evelina Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London SE1 7EH, UK. (33)Royal Belfast Hospital for Sick Children, Belfast BT12 6BA, UK. (34)Paediatric Metabolic Medicine Department, Marmara University Faculty of Medicine, Istanbul 34854, Turkey. (35)Guy's and St Thomas' NHS Foundation Trust, London SE1 7EH, UK. (36)Paediatric Metabolic Medicine Department, Antalya Training and Research Hospital, Antalya 07100, Turkey. (37)Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham B4 6NH, UK. (38)Adult Inherited Metabolic Diseases, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK. (39)Paediatric Metabolic Medicine Department, Kocaeli University Faculty of Medicine, Kocaeli 41380, Turkey. (40)Paediatric Metabolic Medicine Department, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey. (41)Paediatric Metabolic Medicine Department, Istanbul University-Cerrahpasa Faculty of Medicine, Istanbul 34096, Turkey. X-linked ornithine transcarbamylase deficiency (OTCD) is the most common urea cycle defect. The disease severity ranges from asymptomatic carrier state to severe neonatal presentation with hyperammonaemic encephalopathy. We audited the diagnosis and management of OTCD, using an online 12-question-survey that was sent to 75 metabolic centres in Turkey, France and the UK. Thirty-nine centres responded and 495 patients were reported in total. A total of 208 French patients were reported, including 71 (34%) males, 86 (41%) symptomatic and 51 (25%) asymptomatic females. Eighty-five Turkish patients included 32 (38%) males, 39 (46%) symptomatic and 14 (16%) asymptomatic females. Out of the 202 UK patients, 66 (33%) were male, 83 (41%) asymptomatic and 53 (26%) symptomatic females. A total of 19%, 12% and 7% of the patients presented with a neonatal-onset phenotype in France, Turkey and the UK, respectively. Vomiting, altered mental status and encephalopathy were the most common initial symptoms in all three countries. While 69% in France and 79% in Turkey were receiving protein restriction, 42% were on a protein-restricted diet in the UK. A total of 76%, 47% and 33% of patients were treated with ammonia scavengers in Turkey, France and the UK, respectively. The findings of our audit emphasize the differences and similarities in manifestations and management practices in three countries. DOI: 10.3390/life12111721 PMCID: PMC9695856 PMID: 36362876 Conflict of interest statement: PG is an academic co-founder of Bloomsbury Genetic Therapies, UCL spinout developing a gene programme in OTC deficiency.
http://www.ncbi.nlm.nih.gov/pubmed/36217298
1. Ann Clin Transl Neurol. 2022 Nov;9(11):1715-1726. doi: 10.1002/acn3.51668. Epub 2022 Oct 10. Predicting the disease severity in male individuals with ornithine transcarbamylase deficiency. Scharre S(#)(1), Posset R(#)(1), Garbade SF(1), Gleich F(1), Seidl MJ(1), Druck AC(1), Okun JG(1), Gropman AL(2), Nagamani SCS(3), Hoffmann GF(1), Kölker S(1), Zielonka M(1)(4); Urea Cycle Disorders Consortium (UCDC) and the European registry and network for Intoxication type Metabolic Diseases (E-IMD) Consortia Study Group. Collaborators: Ah Mew N, Baumgartner MR, Berry GT, Berry SA, Burrage L, Diaz GA, Ficicioglu C, Kisin G, Konczal L, Lam C, McCandless SE, Merritt JL, Schulze A, Walter ME, Wilson A, Wong D, Arnaudo F, Augoustides-Savvopoulou P, Barić I, Bosch AM, Cano A, Chien YH, Dionisi-Vici C, Dobbelaere D, Eyskens F, Freisinger P, Garcia-Cazorla A, Honzik T, Karall D, Lund AM, Murphy E, Santer R, Schiff M, Skouma A, Sykut-Cegielska J, Wijburg FA, Zeman J. Author information: (1)Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany. (2)Division of Neurodevelopmental Pediatrics and Neurogenetics, Children's National Health System and The George Washington School of Medicine, Washington, District of Columbia, USA. (3)Department of Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA. (4)Heidelberg Research Center for Molecular Medicine (HRCMM), Heidelberg, Germany. (#)Contributed equally OBJECTIVE: Ornithine transcarbamylase deficiency (OTC-D) is an X-linked metabolic disease and the most common urea cycle disorder. Due to high phenotypic heterogeneity, ranging from lethal neonatal hyperammonemic events to moderate symptoms and even asymptomatic individuals, the prediction of the disease course at an early disease stage is very important to individually adjust therapies such as medical treatment or liver transplantation. In this translational study, we developed a severity-adjusted classification system based on in vitro residual enzymatic OTC activity. METHODS: Applying a cell-based expression system, residual enzymatic OTC activities of 71 pathogenic OTC variants were spectrophotometrically determined and subsequently correlated with clinical and biochemical outcome parameters of 119 male individuals with OTC-D (mOTC-D) as reported in the UCDC and E-IMD registries. RESULTS: Integration of multiple data sources enabled the establishment of a robust disease prediction model for mOTC-D. Residual enzymatic OTC activity not only correlates with age at first symptoms, initial peak plasma ammonium concentration and frequency of metabolic decompensations but also predicts mortality. The critical threshold of 4.3% residual enzymatic activity distinguishes a severe from an attenuated phenotype. INTERPRETATION: Residual enzymatic OTC activity reliably predicts the disease severity in mOTC-D and could thus serve as a tool for severity-adjusted evaluation of therapeutic strategies and counselling patients and parents. © 2022 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association. DOI: 10.1002/acn3.51668 PMCID: PMC9639638 PMID: 36217298 [Indexed for MEDLINE] Conflict of interest statement: SK received EU funding for the European registry and network for Intoxication type Metabolic Diseases (E‐IMD; CHAFEA agreement no. 2010 12 01). SK receives funding from Immedica Pharma AB for the European Post‐Authorization Registry for Ravicti® (glycerol phenylbutyrate) oral liquid in partnership with the E‐IMD (RRPE) (EU PAS Register no. EUPAS17267; http://www.encepp.eu/). SK and GFH receive funding from the Dietmar Hopp Foundation (St. Leon‐Rot, Germany) for a pilot study on extended newborn screening evaluating the technical feasibility, diagnostic process quality and health benefits for 28 inherited metabolic diseases including UCDs (NBS 2025, project no. 1DH1911376, 1DH2011117). GFH received lecture fees from Swedish Orphan Biovitrum GmbH. RP receives consultancy fees from Immedica Pharma AB. The sponsors have in no way influenced the design, conductance, analysis and report of the present study. All other authors declare that they have no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/3945292
1. N Engl J Med. 1986 Feb 27;314(9):541-7. doi: 10.1056/NEJM198602273140903. Natural history of symptomatic partial ornithine transcarbamylase deficiency. Rowe PC, Newman SL, Brusilow SW. We reviewed the natural history and differential diagnosis of ornithine transcarbamylase deficiency (an X-linked inborn error of urea synthesis) in 13 symptomatic female heterozygotes. The patients presented as early as the first week of life or as late as the sixth year. The most common symptoms before diagnosis were nonspecific: episodic extreme irritability (100 percent), episodic vomiting and lethargy (100 percent), protein avoidance (92 percent), ataxia (77 percent), Stage II coma (46 percent), delayed physical growth (38 percent), developmental delay (38 percent), and seizures (23 percent). Including the proband, 42 percent of the female members of the 13 families studied had symptoms. The median interval between the onset of major symptoms (vomiting and lethargy, seizures, and coma) and diagnosis was 16 months (range, 1 to 142). Five patients had IQ scores below 70 at the time of diagnosis. We suggest that careful evaluation of the family history, the dietary history, the episodic nature of the nonspecific symptoms, the response of these symptoms to the withdrawal of protein, and their frequent onset at the time of weaning from breast milk will permit early diagnosis and might thereby reduce the risk of death or neurologic impairment in female patients with partial ornithine transcarbamylase deficiency. DOI: 10.1056/NEJM198602273140903 PMID: 3945292 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17845164
1. Pediatr Dermatol. 2007 Jul-Aug;24(4):394-6. doi: 10.1111/j.1525-1470.2007.00457.x. Acrodermatitis enteropathica-like dermatosis associated with ornithine transcarbamylase deficiency. Pascual JC(1), Matarredona J, Mut J. Author information: (1)Department of Dermatology, Hospital General Universitario de Elche, Elche, Spain. [email protected] The urea cycle is the major metabolic pathway for excretion of waste nitrogen. Ornithine transcarbamylase deficiency is the most frequent urea cycle disorder. It is a hereditary-X-linked disease with over 150 mutations described. Ornithine transcarbamylase deficiency causes vomiting, lethargy, hyperventilation, and even death, mainly in the neonatal period. Ammonia, an extremely toxic molecule for the organism, is generated during protein catabolism and is accumulated in patients with this deficiency. Part of the treatment consists of a low-protein diet, to avoid hyperammonemia episodes, which can even have a fatal outcome. Patients can become deficient in several amino acids, either through the low-protein diet or directly through the primary enzyme deficiency; this in turn can cause an acrodermatitis enteropathica-like dermatosis. DOI: 10.1111/j.1525-1470.2007.00457.x PMID: 17845164 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36394206
1. Clin Transl Med. 2022 Nov;12(11):e1113. doi: 10.1002/ctm2.1113. Alternative splicing of HSPA12A pre-RNA by SRSF11 contributes to metastasis potential of colorectal cancer. Pan YJ(1), Huo FC(1), Kang MJ(1), Liu BW(2), Wu MD(1), Pei DS(1). Author information: (1)Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China. (2)Department of General Surgery, Xuzhou Medical University, Xuzhou, China. BACKGROUND: Dysregulation of alternative splicing (AS) induced by serine/arginine-rich proteins has recently been linked to cancer metastasis. Nonetheless, as a member of the serine/arginine-rich protein family, the involvement of SRSF11 in colorectal cancer (CRC) is unknown. METHODS: The TCGA dataset and clinical samples were used to assess SRSF11 expression levels in CRC. For SRSF11, functional experiments were conducted both in vitro and in vivo. RNA-seq technology was used to analyze and screen SRSF11-triggered AS events, which were then confirmed by in vivo UV crosslinking and immunoprecipitation (CLIP) and mini-gene reporter assays. Jalview software was used to determine the preferential binding motif with relation to exon skipping (ES) events. Furthermore, coimmunoprecipitation (Co-IP) and Phospho-tag SDS-PAGE experiments were used to investigate PAK5-mediated phosphorylation regulation on SRSF11, and in vitro kinase experiments validated the interaction. RESULTS: In CRC, SRSF11 was discovered to be overexpressed and associated with a poor prognosis. And SRSF11 played a pro-metastatic role in vitro and in vivo. By screening SRSF11-regulated AS events, we identified the binding motif of SRSF11-triggered splicing-switching of HSPA12A AS, which specifically regulated HSPA12A AS by directly binding to a motif in exon 2. Mechanistically, the HSPA12A transcript with exon 2 retention increased N-cadherin expression by promoting RNA stability. Furthermore, the oncogenic kinase PAK5 phosphorylated SRSF11 at serine 287, protecting it from ubiquitination degradation. CONCLUSIONS: SRSF11 exerts pro-metastatic effects in CRC by inhibiting the AS of HSPA12A pre-RNA. Our findings point to SRSF11-regulated HSPA12A splicing as a novel relationship between SRSF11-regulated splicing and CRC metastasis and suggest a PAK5/SRSF11/HSPA12A axis as a potential therapeutic target and prognostic biomarker in CRC. © 2022 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics. DOI: 10.1002/ctm2.1113 PMCID: PMC9670187 PMID: 36394206 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no potential conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/36001712
1. N Engl J Med. 2022 Aug 25;387(8):715-726. doi: 10.1056/NEJMoa2201302. Olokizumab versus Placebo or Adalimumab in Rheumatoid Arthritis. Smolen JS(1), Feist E(1), Fatenejad S(1), Grishin SA(1), Korneva EV(1), Nasonov EL(1), Samsonov MY(1), Fleischmann RM(1); CREDO2 Group. Collaborators: Everding A, Khariouzov A, Koenig R, Kuehne C, Wassenberg S, Kurthen R, Bortlik L, Dokoupilova E, Horvath R, Lapcikova A, Prochazkova L, Simkova G, Urbanova Z, Vitek P, Svobodova M, Blahova M, Petrikova A, Sleglova O, Machkova M, Rosa J, Novosad L, Houzarova A, Stejfova Z, Gollerova V, Dvorak Z, Drescher E, Nagy M, Simoncsics E, Sulyok G, Somos E, Takacs K, Kovacs A, Rapolthy I, Scheinberg MA, Cauceglia Melazzi AC, D'Andrea Marcolino FM, Ferreira Fernandes AM, Bohn JM, Radominski SC, Scafuto Scotton A, Waldemar Keiserman M, Ximenes AC, da Silva AC, Emerich de Abreu CR, de Vasconcelos V, Lima SM, Martins de Mello F, Mihaela Ramazan AM, Asnal C, Constanza Subils G, Gulin JP, Del Valle Lucero E, Mannucci Walter PA, Moreno JLC, Spindler AJ, Testa GA, Venarotti HO, Velasco Zamora BJ, Kerzberg E, Tate P, Velasco Zamora JL, Gallo R, Carrio J, Alvarellos AJ, Basijokiene V, Bukauskiene L, Lauciuviene R, Kvedaraviciene R, Dambrauskiene J, Ranceva J, Chen HA, Liou LB, Chalem Choueka MR, Arbelaez Cortes A, Velez Sanchez PJ, Jaller Raad JJ, Saaibi Solano DL, Ong V, Pakozdi A, Williams E, George E, Gwynne C, Batley M, Savi T, Müller R, Ju JH, Lee SS, Park YB, Kim J, Jung SY, Lim MK, De la Garza Ramos EH, Garcia Valladares I, Jara Quezada L, Duran Barragan S, Vicente Vicente Gonzalez V, Enriquez Sosa FE, Perez EMV, Flores Alvarado DE, Rizo Rodriguez JC, Munoz Lopez S, Pacheco Tena C, Saulite-Kandevica D, Rehman Q, Neuwelt CM, Schnitz W, Kutner M, Diegel R, Najam S, Sunkureddi P, Waller P, Ayesu K, Bunch T, Wolfe S, Pick M, Mallepalli J, Mabaquiao A, Paez H, Edgerton C, Churchill M, Forstot J, Chohan S, Bhadbhade P, Kenney H, Jesus A, Eisenberg M, El-Kadi H, Goddard D, Saadeh C, Goldberger E, Fraser A, Scoville C, Howell M, Arne ET, Smith K, Mehta C, Gladstein G, Harris M, Baraf H, Mishra N, Codding C, Lee E, Neal-Kraal R, LaGrone R, Metyas S, Murphy F, Babajanians A, Antolini C, Cedeno JE, Querubin R, Vasandani J, King C, Snow D, Iglesias N, Diri E, Firooz N, Karrar A, Brionez T, Shaikh AA, Khan A, Soloman N, Diaz-Secades L, Kreutz D, Basu D, Rivera T, Zagar K, Kumar V, Su TK, Javed S, Bognar M, Tsoneva K, Todorov S, Toncheva A, Dimitrov E, Kapandjieva N, Kopcheva S, Bichovska D, Stoilov R, Penev D, Petranova T, Zielinska A, Rell-Bakalarska M, Burczynska J, Jeka S, Pawtel A, Dankiewicz-Fares I, Dudek A, Dworak K, Kolacinski R, Racewicz A, Olechnowicz-Tietz S, Kaminski A, Kuc K, Strzelecka A, Wojtecka-Grabka M, Rapa A, Malys Brylka A, Rowinska-Osuch A, Wronisz M, Vezikova N, Gordeev I, Evstigneeva L, Izmozherova N, Kamalova R, Krechikova D, Matsievskaya G, Plaksina T, Smolyarchuk E, Stanislav M. Author information: (1)From the Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna (J.S.S.); Helios Fachklinik Vogelsang-Gommern, Vogelsang-Gommern, Germany (E.F.); SFC Medica, Charlotte, NC (S.F.); R-Pharm (S.A.G., E.V.K., M.Y.S.), V.A. Nasonova Research Institute of Rheumatology (E.L.N.), and Sechenov Medical University (M.Y.S.) - all in Moscow; and the University of Texas Southwestern Medical Center at Dallas and Metroplex Clinical Research Center - both in Dallas (R.M.F.). BACKGROUND: The cytokine interleukin-6 is involved in the pathogenesis of rheumatoid arthritis. Olokizumab, a humanized monoclonal antibody targeting the interleukin-6 cytokine directly, is being tested for the treatment of rheumatoid arthritis. METHODS: In a 24-week, phase 3, multicenter, placebo- and active-controlled trial, we randomly assigned (in a 2:2:2:1 ratio) patients with rheumatoid arthritis and an inadequate response to methotrexate to receive subcutaneous olokizumab at a dose of 64 mg every 2 or 4 weeks, adalimumab (40 mg every 2 weeks), or placebo; all patients continued methotrexate therapy. The primary end point was an American College of Rheumatology 20 (ACR20) response (≥20% fewer tender and swollen joints and ≥20% improvement in three of five other domains) at week 12, with each olokizumab dose tested for superiority to placebo. We also tested the noninferiority of each olokizumab dose to adalimumab with respect to the percentage of patients with an ACR20 response (noninferiority margin, -12 percentage points in the lower boundary of the 97.5% confidence interval for the difference between groups). RESULTS: A total of 464 patients were assigned to receive olokizumab every 2 weeks, 479 to receive olokizumab every 4 weeks, 462 to receive adalimumab, and 243 to receive placebo. An ACR20 response at week 12 occurred in 44.4% of the patients receiving placebo, in 70.3% receiving olokizumab every 2 weeks (difference vs. placebo, 25.9 percentage points; 97.5% confidence interval [CI], 17.1 to 34.1), in 71.4% receiving olokizumab every 4 weeks (difference vs. placebo, 27.0 percentage points; 97.5% CI, 18.3 to 35.2), and in 66.9% receiving adalimumab (difference vs. placebo, 22.5 percentage points; 95% CI, 14.8 to 29.8) (P<0.001 for the superiority of each olokizumab dose to placebo). Both olokizumab doses were noninferior to adalimumab with respect to the percentage of patients with an ACR20 response at week 12 (difference, 3.4 percentage points [97.5% CI, -3.5 to 10.2] with olokizumab every 2 weeks and 4.5 percentage points [97.5% CI, -2.2 to 11.2] with olokizumab every 4 weeks). Adverse events, most commonly infections, occurred in approximately 70% of the patients who received olokizumab. Antibodies against olokizumab were detected in 3.8% of the patients receiving the drug every 2 weeks and in 5.1% of those receiving it every 4 weeks. CONCLUSIONS: In patients with rheumatoid arthritis who were receiving maintenance methotrexate, olokizumab was superior to placebo and noninferior to adalimumab in producing an ACR20 response at 12 weeks. Larger and longer trials are required to determine the efficacy and safety of olokizumab in patients with rheumatoid arthritis. (Supported by R-Pharm; CREDO2 ClinicalTrials.gov number, NCT02760407.). Copyright © 2022 Massachusetts Medical Society. DOI: 10.1056/NEJMoa2201302 PMID: 36001712 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29956806
1. Mol Med Rep. 2018 Sep;18(3):3020-3026. doi: 10.3892/mmr.2018.9230. Epub 2018 Jun 27. Decreased expression of TROAP suppresses cellular proliferation, migration and invasion in gastric cancer. Jing K(1), Mao Q(1), Ma P(1). Author information: (1)Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China. Trophinin associated protein (TROAP) is a cytoplasmic protein required for spindle assembly and cell invasion; however, its biological function in cancer remains to be elucidated. In the present study, by analyzing three independent datasets from the Oncomine database, it was identified that TROAP mRNA expression was upregulated in gastric cancer (GC) tissues compared with normal counterparts. Furthermore, elevated expression of TROAP was associated with poor survival in patients with GC, as predicted using Kaplan‑Meier analysis. TROAP was knocked down to verify its functional role in gastric cancer cell lines, SGC‑7901 and MGC80‑3. MTT assay was used to analyze cell proliferation. Cell cycle progression, and migration and invasion were determined using flow cytometry and Transwell assay, respectively. In vitro experiments demonstrated that knockdown of TROAP significantly suppressed cell proliferation, G1 to S cell cycle transition, and the migration and invasion ability of GC cells. The results of the present study suggest that TROAP is overexpressed in GC and serves an oncogenic role in gastric cancer by affecting cell proliferation and invasion. DOI: 10.3892/mmr.2018.9230 PMID: 29956806 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34077623
1. CNS Neurosci Ther. 2021 Jun 2;27(9):1064-76. doi: 10.1111/cns.13688. Online ahead of print. TROAP regulates cell cycle and promotes tumor progression through Wnt/β-Catenin signaling pathway in glioma cells. Zhao ZQ(1)(2), Wu XJ(1)(2), Cheng YH(1)(2), Zhou YF(2), Ma XM(2), Zhang J(1)(2), Heng XY(1)(2), Feng F(1)(2)(3). Author information: (1)Department of Neurosurgery, Linyi People's Hospital, Linyi, China. (2)Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China. (3)Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China. AIMS: Experimental evidence demonstrated a crucial role of TROAP (Trophinin-associated protein) in regulating the cell proliferation of multiple tumors, while TROAP expression and function were largely unknown in glioma. We aimed to investigate the oncogenic role of TROAP and its potential mechanisms in gliomagenesis. METHODS: Four gene expression databases (GEO, TCGA, GTEx and CCLE) were enrolled in our study and used for TROAP expression and survival analysis. TROAP expression was quantified by qRT-PCR, western blot and immunohistochemistry assays in glioma tissues and cell lines. TROAP knockdown and overexpression vector were constructed and transfected into glioma cells. CCK-8, colony formation, transwell, and wound healing assays were used to evaluate cell viability, migration and invasion, flow cytometry to determine cell cycle arrest. Gene set enrichment analysis (GSEA) was conducted to screen the pathway involved in TROAP-high phenotype. The expression of cell cycle and Wnt/β-Catenin signaling proteins were analyzed by immunofluorescence and western blot. RESULTS: Based on the bioinformatic analysis and a series of functional assays, we found the TROAP was enriched in glioma tissues and cell lines, its overexpression was correlated with the clinicopathologic characteristics and poor prognosis. TROAP knockdown inhibited cell proliferation, migration, invasion, and G1/S cell cycle arrest compared with control group in glioma. Mechanism analysis revealed that TROAP activated Wnt/β-Catenin pathway and upregulated its downstream targets expression, while silencing β-Catenin or Axin2 could reverse the tumor-promoting effects caused by TROAP, confirming that TROAP-induced malignant phenotype and tumorigenesis via Wnt/β-Catenin signaling pathway. CONCLUSION: The present study found that TROAP accelerated the progression of gliomagenesis through Wnt/β-Catenin pathway, and TROAP might be considered as a novel target for glioma therapy. © 2021 The Authors. CNS Neuroscience & Therapeutics Published by John Wiley & Sons Ltd. DOI: 10.1111/cns.13688 PMCID: PMC8339535 PMID: 34077623 Conflict of interest statement: No potential conflicts of interest were disclosed.
http://www.ncbi.nlm.nih.gov/pubmed/30854102
1. J Cancer. 2019 Jan 29;10(4):957-967. doi: 10.7150/jca.26666. eCollection 2019. The Upregulation of Trophinin-Associated Protein (TROAP) Predicts a Poor Prognosis in Hepatocellular Carcinoma. Hu H(1), Xu L(2), Chen Y(3), Luo SJ(1), Wu YZ(3), Xu SH(1), Liu MT(1), Lin F(2), Mei Y(2), Yang Q(2), Qiang YY(2), Lin YW(1), Deng YJ(1), Lin T(1), Sha YQ(1), Huang BJ(2), Zhang SJ(1). Author information: (1)Department of Chinese Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, P. R. China. (2)State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P. R. China. (3)Department of Chinese Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, P. R. China. Purpose: Trophinin-associated protein (TROAP) is a cytoplasmic protein that plays a significant role in the processes of embryo transplantation and microtubule regulation. However, the relevant survival analysis and cancer progression analysis have not yet been reported. Methods: Eighteen matched pairs of tumor and adjacent non-tumor samples were evaluated to detect the TROAP mRNA level. Immunohistochemistry (IHC) was used to evaluate the TROAP expression in 108 hepatocellular carcinoma patients who underwent surgical resection. Meanwhile, data from the TCGA database was statistically evaluated. Results: In the present study, we detected a significant increase in the TROAP mRNA level in tumor tissues when compared with adjacent non-tumor tissues. Moreover, the upregulation of TROAP was associated with increased serum AFP and GGT; the greater the tumor number was, the larger the tumor size, differentiation grade, and cancer embolus in clinical analysis. In HCC patients, elevated TROAP expression in the primary tumor was positively related to clinical severity, such as poor overall survival and disease-free survival. In addition, both univariate and multivariate survival analysis validated that TROAP expression was a promising independent risk factor for overall survival and disease-free survival in HCC patients. Furthermore, the results derived from the analysis of data from the TCGA database were consistent with previous results. Altogether, our results show that TROAP is a novel crucial regulator of HCC progression and is a potential therapeutic biomarker for HCC patients. Conclusions: Elevated TROAP expression predicted a poor prognosis, and TROAP may serve as a potential biomarker for application in oncotherapy. DOI: 10.7150/jca.26666 PMCID: PMC6400818 PMID: 30854102 Conflict of interest statement: Competing Interests: The authors have declared that no competing interest exists.
http://www.ncbi.nlm.nih.gov/pubmed/30284652
1. Dig Dis Sci. 2019 Jan;64(1):137-143. doi: 10.1007/s10620-018-5315-x. Epub 2018 Oct 4. High Trophinin-Associated Protein Expression Is an Independent Predictor of Poor Survival in Liver Cancer. Jiao Y(1), Li Y(2), Lu Z(3), Liu Y(4). Author information: (1)Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China. (2)Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China. (3)Department of General Surgery, The Second Hospital of Jilin University, Changchun, 130041, Jilin, People's Republic of China. (4)Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China. [email protected]. BACKGROUND: Trophinin-associated protein (TROAP) is a cytoplasmic protein that functions as an adhesion molecule in processes such as embryo implantation, spindle formation, and cancer. OBJECTIVE: To evaluate the relationship of TROAP expression in hepatocellular carcinoma (HCC) tissue with clinicopathologic parameters and survival time in liver cancer patients based on an analysis of The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) data. METHODS: RNA-sequencing (RNA-Seq) expression data and clinical information were downloaded for the TCGA-LIHC cohort. Associations between TROAP expression in HCC tissues and clinical parameters were evaluated by Chi-square tests. Differences in survival between high and low expression groups (median expression cutoff) from Cox regression analysis were compared, and P values were calculated by a log-rank test. Kaplan-Meier curves were compared with the log-rank test. RESULTS: Analysis of RNA-Seq gene expression data for 373 patients with primary tumors revealed overexpression of TROAP in liver cancer. High TROAP expression was associated with survival status (P = 0.015), T stage (P = 0.049), clinical stage (P = 0.048), and gender (P = 0.033). Patients with high TROAP-expressing liver cancers had a shorter median overall survival of 3.83 years compared with 5.80 years for patients with low TROAP-expressing liver cancers (P = 0.00422). Multivariate analysis identified TROAP expression as an independent prognostic variable for overall survival in liver cancer patients. CONCLUSION: TROAP expression is an independent predictor of poor survival in liver cancer. DOI: 10.1007/s10620-018-5315-x PMID: 30284652 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34344706
1. Ann Rheum Dis. 2022 Apr;81(4):469-479. doi: 10.1136/annrheumdis-2021-219876. Epub 2021 Aug 3. Olokizumab, a monoclonal antibody against interleukin 6, in combination with methotrexate in patients with rheumatoid arthritis inadequately controlled by methotrexate: efficacy and safety results of a randomised controlled phase III study. Nasonov E(1), Fatenejad S(2), Feist E(3), Ivanova M(4), Korneva E(5), Krechikova DG(6), Maslyanskiy AL(7), Samsonov M(5), Stoilov R(4), Zonova EV(8), Genovese M(9). Author information: (1)Research Institute of Rheumatology of RAMS, Moskva, Russian Federation. (2)SFC Medica, Charlotte, North Carolina, USA. (3)Department for Rheumatology, HELIOS Specialist Hospital Vogelsang/Gommern Clinic for Rheumatology, Gommern, Germany. (4)University Hospital St Ivan Rilski Rheumatology Clinic, Sofia, Bulgaria. (5)Medical, CJSC R-Pharm, Moskow, Russian Federation. (6)Non-state Healthcare Institution Regional Clinical Hospital at Smolensk Station, Smolensk, Russian Federation. (7)Medical Research Centre, Federal Almazov North West Medical Research Centre, Saint-Petersburg, Russian Federation. (8)Novosibirsk State Medical University, Novosibirsk, Russian Federation. (9)Division of Immunology and Rheumatology, Stanford University, Stanford, California, USA [email protected]. OBJECTIVE: To evaluate the efficacy and safety of olokizumab (OKZ) in patients with active rheumatoid arthritis despite treatment with methotrexate (MTX). METHODS: In this 24-week multicentre, placebo-controlled, double-blind study, patients were randomised 1:1:1 to receive subcutaneously administered OKZ 64 mg once every 2 weeks, OKZ 64 mg once every 4 weeks, or placebo plus MTX. The primary efficacy endpoint was the proportion of patients achieving an American College of Rheumatology 20% (ACR20) response at week 12. The secondary efficacy endpoints included percentage of subjects achieving Disease Activity Score 28-joint count based on C reactive protein <3.2, Health Assessment Questionnaire Disability Index at week 12, ACR50 response and Clinical Disease Activity Index ≤2.8 at week 24. Safety and immunogenicity were assessed throughout the study. RESULTS: A total of 428 patients were randomised. ACR20 responses were more frequent with OKZ every 2 weeks (63.6%) and OKZ every 4 weeks (70.4%) than placebo (25.9%) (p<0.0001 for both comparisons). There were significant differences in all secondary efficacy endpoints between OKZ-treated arms and placebo. Treatment-emergent serious adverse events (TESAEs) were reported by more patients in the OKZ groups compared with placebo. Infections were the most common TESAEs. No subjects developed neutralising antidrug antibodies. CONCLUSIONS: Treatment with OKZ was associated with significant improvement in signs, symptoms and physical function of rheumatoid arthritis without discernible differences between the two regimens. Safety was as expected for this class of agents. Low immunogenicity was observed. Trial registration number NCT02760368. © Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. DOI: 10.1136/annrheumdis-2021-219876 PMCID: PMC8921576 PMID: 34344706 [Indexed for MEDLINE] Conflict of interest statement: Competing interests: EN: speakers’ bureau for AbbVie, Eli Lilly, Janssen, Novartis, Pfizer. SF: consulting fees from R-Pharm International, ICON and PPD contract research organisations, shareholder of Pfizer, INC stocks. EF: research grants from BMS, Eli Lilly, Novartis, Roche; consulting fees from AbbVie, BMS, Eli Lilly, Gilead Sciences, Galapagos, Novartis, Roche, Sanofi, Sobi; speakers’ bureau for AbbVie, BMS, Eli Lilly, Gilead Sciences, Galapagos, Medac, Novartis, Roche, Sanofi, Sobi. MI: speakers’ bureau for AbbVie, Eli Lilly, MSD, Novartis, Pfizer, UCB. EK: employee of R-Pharm, with no R-Pharm stock. DGK: speakers’ bureau for Bayer, Boehringer Ingelheim, UCB; research grants from BMS, Eli Lilly, Janssen, Pfizer, R-Pharm. ALM: consulting fees from R-Pharm; speakers’ bureau for AbbVie, Boehringer Ingelheim, Novartis, R-Pharm; other activities for AbbVie, Johnson, MSD, Novartis, Roche, outside the submitted work. MS: employee of R-Pharm, with R-Pharm stock. RS: speakers’ bureau for AbbVie, Boehringer Ingelheim, Eli Lilly, Janssen, MSD, Novartis, Pfizer, UCB. EVZ: research grants from AbbVie, Celgene, Janssen, Novartis, Amgen, Pfizer, speakers’ bureau and consultant for AbbVie, Celgene, Janssen, Novartis, Pfizer, Sanofi, Boehringer Ingelheim, Bayer, Sandoz. MG: employee of Gilead Sciences, with Gilead Sciences stock.
http://www.ncbi.nlm.nih.gov/pubmed/29117881
1. Oncol Res. 2018 Jun 11;26(5):691-701. doi: 10.3727/096504017X15101398724809. Epub 2017 Nov 8. Downregulated Trophinin-Associated Protein Plays a Critical Role in Human Hepatocellular Carcinoma Through Upregulation of Tumor Cell Growth and Migration. Lian Y(1), Fan W(2), Huang Y(3), Wang H(1), Wang J(1), Zhou L(3), Wu X(3), Deng M(2), Huang Y(1). Author information: (1)Guangdong Province Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China. (2)Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China. (3)Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China. Trophinin-associated protein (TROAP) was a protein first identified to mediate the process of embryo transplantation and later found to be involved in microtubule regulation. However, little is known about the role of TROAP in hepatocellular carcinoma (HCC). In the present study, we reported that both TROAP mRNA and protein expressions were downregulated in human HCC samples as well as cell lines. A high level of TROAP was associated with small tumor size (p < 0.05), minor tumor nodules (p < 0.01), and mild vein invasion (p < 0.05). We further constructed in vitro TROAP depletion and overexpression HCC cell models. TROAP depletion significantly enhanced the proliferation and colony formation abilities, whereas TROAP overexpression had an inhibitory effect on the growth of HCC cells. The G1/S phase arrest by TROAP overexpression correlated with increased cell cycle inhibitors p21 and p27, and declined cell cycle promoting kinase complex CDK6/cyclin D1. Depressed TROAP expression enhanced the migration ability, while the opposite influence was observed in TROAP-overexpressed HCC cells. Taken together, these results indicate that TROAP suppresses cellular growth and migration in HCC. This discovery will further our understanding of the pathogenic mechanisms of human HCC. DOI: 10.3727/096504017X15101398724809 PMCID: PMC7844635 PMID: 29117881 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/33500384
1. Cell Death Dis. 2021 Jan 26;12(1):125. doi: 10.1038/s41419-021-03422-3. TROAP switches DYRK1 activity to drive hepatocellular carcinoma progression. Li L(#)(1)(2)(3), Wei JR(#)(4), Song Y(#)(5), Fang S(#)(6), Du Y(#)(6), Li Z(7), Zeng TT(7), Zhu YH(7), Li Y(7), Guan XY(8)(9)(10). Author information: (1)State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China. [email protected]. (2)Department of Clinical Oncology, State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China. [email protected]. (3)Department of Clinical Oncology Center, The University of Hongkong-Shenzhen Hospital, 518053, Shenzhen, China. [email protected]. (4)State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 510060, Guangzhou, China. (5)Affiliated Cancer Hospital & Institutes of Guangzhou Medical University, Guangzhou Key Medical Discipline Construction Project, 510095, Guangzhou, China. (6)The Seventh Affiliated Hospital, Sun Yat-sen University, 518100, Shenzhen, China. (7)State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China. (8)State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China. [email protected]. (9)Department of Clinical Oncology, State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China. [email protected]. (10)Department of Clinical Oncology Center, The University of Hongkong-Shenzhen Hospital, 518053, Shenzhen, China. [email protected]. (#)Contributed equally Hepatocellular carcinoma (HCC) is one of the common malignancy and lacks effective therapeutic targets. Here, we demonstrated that ectopic expression of trophinin-associated protein (TROAP) dramatically drove HCC cell growth assessed by foci formation in monolayer culture, colony formation in soft agar and orthotopic liver transplantation in nude mice. Inversely, silencing TROAP expression with short-hairpin RNA attenuated the malignant proliferation of HCC cells in vitro and in vivo. Next, mechanistic investigation revealed that TROAP directly bound to dual specificity tyrosine phosphorylation regulated kinase 1A/B (DYRK1A/B), resulting in the cytoplasmic retention of proteins DYRK1A/B and promoting cell cycle process via activation of Akt/GSK-3β signaling. Combination of cisplatin with an inhibitor of DYRK1 AZ191 effectively inhibited tumor growth in mouse model for HCC cells with high level of TROAP. Clinically, TROAP was significantly upregulated by miR-142-5p in HCC tissues, which predicted the poor survival of patients with HCC. Therefore, TROAP/DYRK1/Akt axis may be a promising therapeutic target and prognostic indicator for patients with HCC. DOI: 10.1038/s41419-021-03422-3 PMCID: PMC7838256 PMID: 33500384 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/31198787
1. Biomed Res Int. 2019 May 6;2019:6140951. doi: 10.1155/2019/6140951. eCollection 2019. TROAP Promotes Breast Cancer Proliferation and Metastasis. Li K(1), Zhang R(2), Wei M(3), Zhao L(1), Wang Y(4), Feng X(5), Yang Y(5), Yang S(6), Zhang L(5). Author information: (1)Department of Oncology I, Shengjing Hospital of China Medical University, Shenyang, China. (2)Shuwen Biotech Co. Ltd., Deqing, China. (3)School of Pharmacy, China Medical University, Shenyang, China. (4)The 4 Affiliated Hospital of Harbin Medical University, Harbin, China. (5)Department of Pathology, Harbin Medical University, Harbin, China. (6)Department of Anatomy, Basic Medical Science College, Harbin Medical University, Harbin, China. Trophinin-associated protein (TROAP) is a cytoplasmic protein required for microtubular cytoskeleton regulation and spindle assembly, and its expression plays a critical role in the initiation and progression of various types of cancer. However, little is known about the role of TROAP in breast cancer (BC). TROAP mRNA expression levels and clinical data from Gene Expression Omnibus (GEO) datasets (GSE42568, 104 BC patients; GSE1456, 159 BC patients; and GSE21653, 266 BC patients) were analyzed by the R2: Genomics Analysis and Visualization Platform to estimate overall survival (OS). We also analyzed the genes correlated with TROAP by gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis to predict potential relationships between TROAP and other genes in BC. Our study verified that both TROAP mRNA and protein expression levels were upregulated in human BC samples and cell lines. In vitro experiments demonstrated that TROAP knockdown significantly inhibited cell proliferation, the G1 to S phase transition, and the migration and invasion abilities of BC cells. The present study suggests that TROAP plays an important role in promoting the proliferation, invasion, and metastasis of BC. DOI: 10.1155/2019/6140951 PMCID: PMC6526557 PMID: 31198787 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30431120
1. Oncol Rep. 2019 Feb;41(2):1169-1179. doi: 10.3892/or.2018.6854. Epub 2018 Nov 9. TROAP regulates prostate cancer progression via the WNT3/survivin signalling pathways. Ye J(1), Chu C(1), Chen M(2), Shi Z(3), Gan S(1), Qu F(1), Pan X(1), Yang Q(1), Tian Y(1), Wang L(1), Yang W(1), Cui X(1). Author information: (1)Department of Urology, Third Affiliated Hospital, The Second Military Medical University, Shanghai 201805, P.R. China. (2)Department of Urology, Affiliated Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China. (3)Department of Urology, Taizhou First People's Hospital, Taizhou, Zhejiang 318020, P.R. China. Prostate cancer (PCa) is one of the most commonly diagnosed malignancies, and 90% of advanced prostate cancer patients relapse after therapy. Trophinin associated protein (TROAP) is essential for centrosome integrity and proper bipolar organisation of spindle assembly during mitosis and plays an essential role in proliferation. We found that TROAP expression correlates with patient survival and speculated that it may be involved in PCa progression. The Oncomine database tool (http://www.oncomine.org) was used to analyse TROAP mRNA expression from microarray data, and patient survival analysis for target genes was performed using the PROGgeneV2 Database (http://watson.compbio.iupui.edu). Gene interference with lentivirus was used to silence TROAP expression in PCa cells and knockdown efficiency was detected by qRT-PCR and western blot analysis. Cell viability, colony formation, cell cycle and apoptosis were then assessed to determine the function of TROAP in PCa cells. Markers of cell cycle and apoptosis were tested by western blotting. The correlation between WNT3 or survivin expression and TROAP transcripts in prostate cancer tissues was analysed using GEPIA (http://gepia.cancer-pku.cn) and validated by western blotting. The in vivo role of TROAP was investigated using xenografts. This protein was overexpressed in PCa, and exhibited relatively higher expression in PCa cell lines, DU145 and 22Rv1. Importantly, analysing human cancer databases available from PROGgeneV2 showed that higher expression of TROAP is associated with shorter overall survival in prostate cancer patients. TROAP knockdown inhibited cell proliferation and led to cell cycle arrest at S phase in 22Rv1 and DU145 cells. Cell cycle arrest resulted in apoptosis in both cell lines via the cyclin A2-cyclin B1-caspase pathway. WNT3 and survivin expression levels were found to correlate with TROAP in PCa, and in vivo xenograft assays revealed that silencing of TROAP inhibited PCa tumour growth. Therefore, TROAP might represent a novel predictive marker to guide therapeutic intervention. DOI: 10.3892/or.2018.6854 PMID: 30431120 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31285897
1. J Thorac Dis. 2019 May;11(5):2043-2050. doi: 10.21037/jtd.2019.04.86. Trophinin-associated protein expression is an independent prognostic biomarker in lung adenocarcinoma. Chen Z(1), Zhou Y(1), Luo R(1), Liu K(1), Chen Z(1). Author information: (1)Department of Thoracic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China. BACKGROUND: Lung cancer is the leading cause of cancer-related deaths worldwide, with lung adenocarcinoma (LAC) representing the most common subtype. Trophinin-associated protein (TROAP) is a cytoplasmic protein first identified to mediate the process of embryo transplantation, which has been recently found to be involved in microtubule regulation. However, limited information about the role of TROAP in LAC is available. METHODS: We evaluated the relationship of TROAP expression in LAC tissues with clinical pathologic parameters and the survival time in LAC patients based on a statistical analysis of The Cancer Genome Atlas (TCGA) lung cancer data (N=528). Differences in survival between high and low expression groups (median expression cutoff) from the Cox univariate/multivariate regression analysis were then compared. RESULTS: According to the Chi-square tests, we found high TROAP expression correlated with younger age (≤60) (P=0.047), male sex (P<0.005), an earlier T-stage (P=0.011), N-stage (P=0.017), M-stage (P=0.022), TNM (P=0.007), and a longer smoking history (>30 pack-year) (P<0.001). A Kaplan-Meier analysis demonstrated that high TROAP expression may correspond with poor overall survival of LAC patients in T3 stage (P=0.0013), N0 stage (P=0.014), and M0 stage (P=0.0023). Multivariate analysis confirmed that TROAP expression was related to overall survival in LAC patients independently [hazard ratio (HR): 1.784, 95% confidence interval (CI): 1.072-2.968, P=0.026]. CONCLUSIONS: Our results suggested that TROAP is an independent prognostic biomarker of poor survival in LAC. DOI: 10.21037/jtd.2019.04.86 PMCID: PMC6588746 PMID: 31285897 Conflict of interest statement: Conflicts of Interest: The authors have no conflicts of interest to declare.
http://www.ncbi.nlm.nih.gov/pubmed/33692939
1. Front Oncol. 2021 Feb 22;10:592239. doi: 10.3389/fonc.2020.592239. eCollection 2020. EZH2-TROAP Pathway Promotes Prostate Cancer Progression Via TWIST Signals. Jin L(1), Zhou Y(1), Chen G(2), Dai G(1), Fu K(1), Yang D(1), Zhu J(1). Author information: (1)Department of Urology, Second Affiliated Hospital of Soochow University, Suzhou, China. (2)Department of Radiology, Second Affiliated Hospital of Soochow University, Suzhou, China. Trophinin-associated protein (TROAP) has been shown to be overexpressed and promotes tumor progression in some tumors. We performed this study to assess the biological and clinical significance of TROAP in prostate cancer. We downloaded TROAP mRNA expression data from TCGA and GEO databases. We analyzed expressions of TROAP and other genes in prostate cancer tumors at different stages and assessed Gleason scores. We used Celigo image, Transwell, and rescue assays, and flow cytometry detection to assess growth, apoptosis, proliferation, migration, and invasion of the prostate cancer cells. We identified and validated up- and down-stream genes in the TROAP pathway. The mRNA data suggested that TROAP expression was markedly upregulated in prostate cancer compared with its expression in normal tissues, especially in cancers with high stages and Gleason scores. Moreover, a high TROAP expression was associated with poor patient survival. Results of our in vitro assay showed that TROAP knockdown inhibited DU145 and PC3 cell proliferation and viability via cell apoptosis and S phase cycle arrest. The Transwell assay showed that TROAP knockdown inhibited cell migration and invasion, probably through MMP-9 and E-Cadherin modulation. Overexpression of TWIST partially abrogated the inhibitory effects of TROAP knockdown on prostate cancer cells. Our integrative mechanism dissection revealed that TROAP is in a pathway downstream of EZH2 and that it activates the TWIST/c-Myc pathway to regulate prostate cancer progression. In all, we identified TROAP as a driver of prostate cancer development and progression, providing a novel target for prostate cancer treatments. Copyright © 2021 Jin, Zhou, Chen, Dai, Fu, Yang and Zhu. DOI: 10.3389/fonc.2020.592239 PMCID: PMC7938320 PMID: 33692939 Conflict of interest statement: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35708862
1. Mol Biol Rep. 2022 Aug;49(8):7899-7909. doi: 10.1007/s11033-022-07622-8. Epub 2022 Jun 16. Trophinin-associated protein expression correlates with shorter survival of patients with glioma: a study based on multiple data fusion analysis. Sun Y(#)(1), Liu ZD(#)(2), Liu RZ(#)(2), Lian XY(#)(2), Cheng XB(2), Jia YL(3), Liu BF(2), Gao YZ(4), Wang X(5). Author information: (1)Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China. (2)Department of Surgery of Spine and Spinal Cord, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital; People's Hospital of Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China. (3)Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, China. (4)Department of Surgery of Spine and Spinal Cord, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital; People's Hospital of Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China. [email protected]. (5)Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China. [email protected]. (#)Contributed equally BACKGROUND: Trophinin-associated protein (TROAP) mediates embryonic transfer, regulates microtubules, and is associated with the biological behavior of various cancers. However, there is limited information on the role of TROAP in glioma. METHODS AND RESULTS: We obtained clinical information on 1948 patients with glioma from The Cancer Genome Atlas, Gene Expression Omnibus and the Chinese Glioma Genome Atlas. Basal assays were used to measure changes in TROAP expression levels in high-grade glioma cell lines and in normal human astrocytes. Quantitative reverse transcription polymerase chain reaction assays showed that TROAP expression was higher in glioma cell lines than in normal astrocytes. The expression level of TROAP in 749 glioma was significantly higher than that in 228 normal brain tissues using Student's t test. The expression of TROAP has a positive relationship with the clinical characteristics of poor prognosis, such as WHO grade, age and has negatively correlated with the indicators of beneficial prognosis, such as IDH mutation and 1p19q co-deletion. Kaplan-Meier survival curves, single multifactor analysis were used to analyze correlations between TROAP and clinical features and prognosis of gliomas. In addition, TROAP overexpression was an independent risk factor for glioma and was associated with reduced overall survival of patients with glioma particularly in patients with WHO grade III and grade IV glioma. Gene set enrichment analysis showed that homologous recombination, cell cycle, and p53 signaling pathways were enriched in samples overexpressing TROAP. CONCLUSION: TROAP is a potential risk factor associated with poor prognosis in patients with glioma and may act as a highly specific biomarker, offering the possibility of individualized glioma treatment. © 2022. The Author(s), under exclusive licence to Springer Nature B.V. DOI: 10.1007/s11033-022-07622-8 PMID: 35708862 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34287099
1. Cell Cycle. 2021 Aug;20(16):1578-1588. doi: 10.1080/15384101.2021.1953767. Epub 2021 Jul 21. MiR-532-3p suppresses cell viability, migration and invasion of clear cell renal cell carcinoma through targeting TROAP. Gao B(1), Wang L(1), Zhang Y(1), Zhang N(1), Han M(1), Liu H(1), Sun D(1), Liu Y(1). Author information: (1)Department of Urology, Tangshan Central Hospital, Tangshan, Hebei, P.R. China. Clear cell renal cell carcinoma (ccRCC) is a subtype of renal cell cancer with the highest mortality, infiltration, and metastasis rate, threatening human health. Despite oncogenic role of TROAP in various cancers, its function in ccRCC remains to be unraveled. The differentially expressed mRNAs (DEmRNAs) and miRNAs (DEmiRNAs) were obtained by analyzing the related data sets of ccRCC in TCGA. The expression levels of mRNAs and miRNAs in the cell were detected by qRT-PCR, while the protein levels were characterized by western blot. The viability, migratory and invasive abilities of ccRCC cells were determined by MTT, wound healing and cell invasion assays. The combination of miRNA target site prediction and dual-luciferase reporter gene assay verified the binding relationship between miR-532-3p and TROAP. Research on ccRCC displayed that TROAP expression was upregulated, while miR-532-3p was down-regulated. Besides, upregulation of TROAP could accelerate viability, migratory and invasive potentials of ccRCC cells. On the contrary, miR-532-3p could downregulate TROAP level, but TROAP upregulation reversed the viability, migration, and invasion of ccRCC cells. MiR-532-3p could attenuate the viability, migration and invasion of ccRCC cells by targeting TROAP. This may generate novel insights into molecular therapeutic targets for ccRCC. DOI: 10.1080/15384101.2021.1953767 PMCID: PMC8409774 PMID: 34287099 [Indexed for MEDLINE] Conflict of interest statement: No potential conflict of interest was reported by the author(s).
http://www.ncbi.nlm.nih.gov/pubmed/36394206
1. Clin Transl Med. 2022 Nov;12(11):e1113. doi: 10.1002/ctm2.1113. Alternative splicing of HSPA12A pre-RNA by SRSF11 contributes to metastasis potential of colorectal cancer. Pan YJ(1), Huo FC(1), Kang MJ(1), Liu BW(2), Wu MD(1), Pei DS(1). Author information: (1)Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China. (2)Department of General Surgery, Xuzhou Medical University, Xuzhou, China. BACKGROUND: Dysregulation of alternative splicing (AS) induced by serine/arginine-rich proteins has recently been linked to cancer metastasis. Nonetheless, as a member of the serine/arginine-rich protein family, the involvement of SRSF11 in colorectal cancer (CRC) is unknown. METHODS: The TCGA dataset and clinical samples were used to assess SRSF11 expression levels in CRC. For SRSF11, functional experiments were conducted both in vitro and in vivo. RNA-seq technology was used to analyze and screen SRSF11-triggered AS events, which were then confirmed by in vivo UV crosslinking and immunoprecipitation (CLIP) and mini-gene reporter assays. Jalview software was used to determine the preferential binding motif with relation to exon skipping (ES) events. Furthermore, coimmunoprecipitation (Co-IP) and Phospho-tag SDS-PAGE experiments were used to investigate PAK5-mediated phosphorylation regulation on SRSF11, and in vitro kinase experiments validated the interaction. RESULTS: In CRC, SRSF11 was discovered to be overexpressed and associated with a poor prognosis. And SRSF11 played a pro-metastatic role in vitro and in vivo. By screening SRSF11-regulated AS events, we identified the binding motif of SRSF11-triggered splicing-switching of HSPA12A AS, which specifically regulated HSPA12A AS by directly binding to a motif in exon 2. Mechanistically, the HSPA12A transcript with exon 2 retention increased N-cadherin expression by promoting RNA stability. Furthermore, the oncogenic kinase PAK5 phosphorylated SRSF11 at serine 287, protecting it from ubiquitination degradation. CONCLUSIONS: SRSF11 exerts pro-metastatic effects in CRC by inhibiting the AS of HSPA12A pre-RNA. Our findings point to SRSF11-regulated HSPA12A splicing as a novel relationship between SRSF11-regulated splicing and CRC metastasis and suggest a PAK5/SRSF11/HSPA12A axis as a potential therapeutic target and prognostic biomarker in CRC. © 2022 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics. DOI: 10.1002/ctm2.1113 PMCID: PMC9670187 PMID: 36394206 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no potential conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/36190128
1. mBio. 2022 Oct 26;13(5):e0180422. doi: 10.1128/mbio.01804-22. Epub 2022 Oct 3. Structural and Mechanistic Bases of Viral Resistance to HIV-1 Capsid Inhibitor Lenacapavir. Bester SM(1), Adu-Ampratwum D(2), Annamalai AS(1), Wei G(1), Briganti L(1), Murphy BC(1), Haney R(1), Fuchs JR(2), Kvaratskhelia M(1). Author information: (1)Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, Colorado, USA. (2)Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State Universitygrid.261331.4, Columbus, Ohio, USA. Lenacapavir (LEN) is a long-acting, highly potent HIV-1 capsid (CA) inhibitor. The evolution of viral variants under the genetic pressure of LEN identified Q67H, N74D, and Q67H/N74D CA substitutions as the main resistance associated mutations (RAMs). Here, we determined high-resolution structures of CA hexamers containing these RAMs in the absence and presence of LEN. Our findings reveal that the Q67H change induces a conformational switch, which adversely affects the inhibitor binding. In the unliganded protein, the His67 side chain adopts the closed conformation by projecting into the inhibitor binding pocket and thereby creating steric hindrance with respect to LEN. Upon the inhibitor binding, the His67 side chain repositions to the open conformation that closely resembles the Gln67 side chain in the WT protein. We propose that the switch from the closed conformation to the open conformation, which is needed to accommodate LEN, accounts for the reduced inhibitor potency with respect to the Q67H CA variant. The N74D CA change results in the loss of a direct hydrogen bond and in induced electrostatic repulsions between CA and LEN. The double Q67H/N74D substitutions exhibited cumulative effects of respective single amino acid changes. An examination of LEN binding kinetics to CA hexamers revealed that Q67H and N74D CA changes adversely influenced the inhibitor binding affinity (KD) by primarily affecting the dissociation rate constant (koff). We used these structural and mechanistic findings to rationally modify LEN. The resulting analog exhibited increased potency against the Q67H/N74D viral variant. Thus, our studies provide a means for the development of second-generation inhibitors with enhanced barriers to resistance. IMPORTANCE LEN is an investigational long-acting agent for future HIV-1 treatment regimens. While ongoing clinical trials have highlighted a largely beneficial profile of LEN for the treatment of HIV-1 infected people with limited therapy options, one notable shortcoming is a relatively low barrier of viral resistance to the inhibitor. Cell culture-based viral breakthrough assays identified N74D, Q67H, and N74D/Q67H capsid changes as the main resistance associated mutations (RAMs). N74D and Q67H capsid substitutions have also emerged in clinical trials in some patients who received subcutaneous LEN. Understanding the structural basis behind viral resistance to LEN is expected to aid in the rational development of improved inhibitors with enhanced barriers to resistance. Here, we report high resolution structures of the main drug resistant capsid variants, which provide mechanistic insight into the viral resistance to LEN. We used these findings to develop an improved inhibitor, which exhibited enhanced activity against the viral Q67H/N74D capsid phenotype compared with that of parental LEN. DOI: 10.1128/mbio.01804-22 PMCID: PMC9600929 PMID: 36190128 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/36202818
1. Nat Commun. 2022 Oct 6;13(1):5879. doi: 10.1038/s41467-022-33662-6. Prion-like low complexity regions enable avid virus-host interactions during HIV-1 infection. Wei G(1), Iqbal N(2), Courouble VV(3), Francis AC(4)(5), Singh PK(6)(7), Hudait A(8), Annamalai AS(1), Bester S(1), Huang SW(1)(9), Shkriabai N(1), Briganti L(1), Haney R(1), KewalRamani VN(9), Voth GA(8), Engelman AN(6)(7), Melikyan GB(5), Griffin PR(3), Asturias F(2), Kvaratskhelia M(10). Author information: (1)Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA. (2)Department of Biochemistry & Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA. (3)Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA. (4)Institute of Molecular Biophysics, Department of Biological Sciences, Florida State University, Tallahassee, FL, 32306, USA. (5)Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA. (6)Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA. (7)Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA. (8)Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Franck Institute, The University of Chicago, Chicago, IL, 60637, USA. (9)Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA. (10)Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA. [email protected]. Cellular proteins CPSF6, NUP153 and SEC24C play crucial roles in HIV-1 infection. While weak interactions of short phenylalanine-glycine (FG) containing peptides with isolated capsid hexamers have been characterized, how these cellular factors functionally engage with biologically relevant mature HIV-1 capsid lattices is unknown. Here we show that prion-like low complexity regions (LCRs) enable avid CPSF6, NUP153 and SEC24C binding to capsid lattices. Structural studies revealed that multivalent CPSF6 assembly is mediated by LCR-LCR interactions, which are templated by binding of CPSF6 FG peptides to a subset of hydrophobic capsid pockets positioned along adjoining hexamers. In infected cells, avid CPSF6 LCR-mediated binding to HIV-1 cores is essential for functional virus-host interactions. The investigational drug lenacapavir accesses unoccupied hydrophobic pockets in the complex to potently impair HIV-1 inside the nucleus without displacing the tightly bound cellular cofactor from virus cores. These results establish previously undescribed mechanisms of virus-host interactions and antiviral action. © 2022. The Author(s). DOI: 10.1038/s41467-022-33662-6 PMCID: PMC9537594 PMID: 36202818 [Indexed for MEDLINE] Conflict of interest statement: A.N.E. has received compensation from ViiV Healthcare Co. for work unrelated to this study. No other authors have potential competing interests to declare.
http://www.ncbi.nlm.nih.gov/pubmed/36272024
1. Drugs. 2022 Sep;82(14):1499-1504. doi: 10.1007/s40265-022-01786-0. Lenacapavir: First Approval. Paik J(1). Author information: (1)Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand. [email protected]. Erratum in Drugs. 2023 Jul;83(11):1061. doi: 10.1007/s40265-023-01908-2. Lenacapavir (Sunlenca®) is a long-acting capsid inhibitor of human immunodeficiency virus type 1 (HIV-1) being developed by Gilead Sciences Inc. It is available as an oral tablet and injectable solution, with the latter being a slow-release formulation to allow bi-annual subcutaneous administration. In August 2022, lenacapavir received its first approval in the EU for use in combination with other antiretroviral(s) in adults with multi-drug resistant HIV infection, for whom it is otherwise not possible to construct a suppressive anti-viral regimen. This article summarizes the milestones in the development of lenacapavir leading to this first approval for the treatment of HIV-1 infection. © 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG. DOI: 10.1007/s40265-022-01786-0 PMCID: PMC10267266 PMID: 36272024 [Indexed for MEDLINE] Conflict of interest statement: During the peer review process the manufacturer of the agent under review was offered an opportunity to comment on the article. Changes resulting from any comments received were made by the authors on the basis of scientific completeness and accuracy. Julia Paik is a salaried employee of Adis International Ltd/Springer Nature, and declares no relevant conflicts of interest. All authors contributed to the review and are responsible for the article content.
http://www.ncbi.nlm.nih.gov/pubmed/34871187
1. Curr Opin HIV AIDS. 2022 Jan 1;17(1):15-21. doi: 10.1097/COH.0000000000000713. Lenacapavir: a first-in-class HIV-1 capsid inhibitor. Dvory-Sobol H(1), Shaik N, Callebaut C, Rhee MS. Author information: (1)Gilead Sciences, Foster City, California, USA. PURPOSE OF REVIEW: This review summarizes available data for lenacapavir, an investigational first-in-class agent that disrupts functioning of HIV capsid protein across multiple steps in the viral life cycle. RECENT FINDINGS: Lenacapavir demonstrated picomolar potency in vitro with no cross resistance to existing antiretroviral classes and potent antiviral activity in persons with HIV-1. In persons with HIV-1, there was no preexisting resistance to lenacapavir regardless of treatment history. Lenacapavir can be administered orally either daily or weekly and subcutaneously up to every 6 months. In heavily treatment-experienced persons with multidrug-resistant HIV-1 and in treatment-naive persons with HIV-1, lenacapavir in combination with other antiretroviral agents led to high rates of virologic suppression and was well tolerated. SUMMARY: Ongoing studies are evaluating long-acting dosing of lenacapavir for treating HIV-1 in combination with other antiretrovirals and preventing HIV-1 as a single agent. Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved. DOI: 10.1097/COH.0000000000000713 PMID: 34871187 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36166211
1. Nefrologia (Engl Ed). 2021 Mar-Apr;41(2):123-136. doi: 10.1016/j.nefroe.2020.11.011. Epub 2021 May 11. Iron replacement therapy in the management of anaemia in non-dialysis Chronic kidney disease patients: Perspective of the Spanish Nephrology Society Anaemia Group. Cases A(1), Puchades MJ(2), de Sequera P(3), Quiroga B(4), Martin-Rodriguez L(5), Gorriz JL(6), Portolés J(5); en representación del Grupode Anemia de la S.E.N.. Author information: (1)Universitat de Barcelona, IDIBAPS, Spain. (2)Servicio de Nefrología, Hospital Clínico, INCLIVA, Universidad de Valencia, Valencia, Spain. (3)Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, Spain. (4)Servicio de Nefrología, Hospital Universitario de la Princesa, Madrid, Servicio de Nefrología, Hospital Clínico, Valencia, INCLIVA, Universidad de Valencia, Spain. (5)Servicio de Nefrología, Hospital Universitario Puerta de Hierro Majadahonda, REDInREN ISCiii 016/009/009 RETYC, Majadahonda, Madrid, Spain. (6)Servicio de Nefrología, Hospital Clínico, INCLIVA, Universidad de Valencia, Valencia, Spain. Electronic address: [email protected]. This work presents an update on the management of iron deficiency in patients with chronic kidney disease (CKD), either with or without anaemia. A review is made of the recommendations of the guidelines for the treatment of iron deficiency in CKD. It also presents new studies on iron deficiency in patients with CKD, as well as new findings about iron therapy and its impact on clinical outcomes. Anaemia is a common complication of CRF, and is associated with a decrease in the quality of life of the patients, as well as an increase in morbidity and mortality. Iron deficiency (absolute or functional) is common in non-dialysis chronic kidney disease patients, and may cause anaemia or a low response to erythropoiesis-stimulating agents. For this reason, the clinical guidelines for the treatment of the anaemia in Nephrology indicate the correction of the deficiency in the presence of anaemia. Iron replacement therapy is indicated in patients with CKD and anaemia (Hb < 12 g/dl) in accordance with the guidelines. There is no unanimity in the indication of iron replacement therapy in patients with Hb > 12 g/dl, regardless of whether they have an absolute or functional iron deficiency. Intravenous iron replacement therapy is safe, more efficient and rapid than oral therapy for achieving an increase haemoglobin lels and reducing the dose of erythropoiesis-stimulating agents. For the administration of intravenous iron in non-dialysis chronic renal failure patients a strategy of high doses and low frequency would be preferred on being more convenient for the patient, preserves better the venous capital, and is safe and cost-effective. Iron plays an essential role in energy metabolism and other body functions beyond the synthesis of haemoglobin, for which the iron deficiency, even in the absence of anaemia, could have harmful effects in patients with CKD. The correction of the iron deficiency, in the absence of anaemia is associated with functional improvement in patients with heart failure, and in muscle function or fatigue in patients without CKD. Despite the evidence of benefits in the correction of iron deficiency in patients with CKD, more studies are required to evaluate the impact of the correction of the iron deficiency in the absence of anaemia on morbidity and mortality, quality of life and physical capacity, as well as the long-term effect of oral and intravenous iron replacement therapy in this population. Copyright © 2020 Sociedad Española de Nefrología. Published by Elsevier España, S.L.U. All rights reserved. DOI: 10.1016/j.nefroe.2020.11.011 PMID: 36166211
http://www.ncbi.nlm.nih.gov/pubmed/28153964
1. Perit Dial Int. 2017 1-2;37(1):6-13. doi: 10.3747/pdi.2016.00193. Anemia in Peritoneal Dialysis Patients; Iron Repletion, Current and Future Therapies. Zeidan A(1), Bhandari S(2). Author information: (1)Department of Academic Renal Research, Hull and East Yorkshire Hospital Trust and Hull York Medical School, Kingston Upon Hull, UK. (2)Department of Academic Renal Research, Hull and East Yorkshire Hospital Trust and Hull York Medical School, Kingston Upon Hull, UK [email protected]. Iron deficiency, both functional and absolute, is common in patients with chronic kidney disease (CKD), especially those requiring dialysis. Guidelines advocate treatment of iron-deficiency anemia in patients with CKD and those on peritoneal dialysis (PD). Oral iron is often insufficient and slow to improve hemoglobin concentrations because of high hepcidin levels causing impaired absorption and mobilization, while intravenous (IV) supplementation replenishes and maintains iron stores more effectively and is now standard practice (Kidney Disease Improving Global Outcomes [KDIGO] 2012 guidelines). However, there still remain concerns about the effects of labile iron and possible increased risk of infections for this group of patients.To date, the majority of published studies have focused on hemodialysis (HD) patients; very limited data are available regarding patients on PD. This review summarizes the rationale for iron therapy, methods of treatment, potential adverse effects, and long-term concerns in PD patients. In addition we highlight some interesting potential future therapies under study. Copyright © 2017 International Society for Peritoneal Dialysis. DOI: 10.3747/pdi.2016.00193 PMID: 28153964 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29533917
1. Wiad Lek. 2017;70(6 pt 2):1215-1218. [Iron supplementation in chronic kidney disease]. [Article in Polish] Graczyk M(1), Dylewska M(1). Author information: (1)Katedra i Klinika Nefrologii, Dializoterapii i Chorób Wewnętrznych, Warszawski Uniwersytet Medyczny, Warszawa, Polska. Treatment with iron preparations remains one of the main directions in the treatment of anemia in patients with chronic kidney disease. Intravenous agents, although effective, may have serious adverse effects, while oral iron supplementation may be ineffective due to malabsorption and gastrointestinal side effects. The solution may be modern drugs such as ferric pyrophosphate added to dialysis fluid or liposomal iron without gastrointestinal adverse effects. PMID: 29533917 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33516607
1. Nefrologia (Engl Ed). 2021 Mar-Apr;41(2):123-136. doi: 10.1016/j.nefro.2020.11.003. Epub 2021 Jan 28. Iron replacement therapy in the management of anaemia in non-dialysis chronic renal failure patients: Perspective of the Spanish Nephrology Society Anaemia Group. [Article in English, Spanish] Cases A(1), Puchades MJ(2), de Sequera P(3), Quiroga B(4), Martin-Rodriguez L(5), Gorriz JL(6), Portolés J(5); en representación del Grupo de Anemia de la S.E.N.. Author information: (1)Universitat de Barcelona, IDIBAPS, España. (2)Servicio de Nefrología, Hospital Clínico, INCLIVA, Universidad de Valencia, Valencia, España. (3)Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, España. (4)Servicio de Nefrología, Hospital Universitario de la Princesa, Madrid, Servicio de Nefrología, Hospital Clínico, Valencia, INCLIVA, Universidad de Valencia, España. (5)Servicio de Nefrología, Hospital Universitario Puerta de Hierro Majadahonda, REDInREN ISCiii 016/009/009 RETYC, Majadahonda, Madrid, España. (6)Servicio de Nefrología, Hospital Clínico, INCLIVA, Universidad de Valencia, Valencia, España. Electronic address: [email protected]. Comment in Nefrologia (Engl Ed). 2022 Nov-Dec;42(6):736-737. doi: 10.1016/j.nefroe.2021.04.016. This work presents an update on the management of iron deficiency in patients with chronic renal failure (CRF), either with or without anaemia. A review is made of the recommendations of the guidelines for the treatment of iron deficiency in CRF. It also presents new studies on iron deficiency in patients with CRF, as well as new findings about iron deficiency and its impact on clinical outcomes. Anaemia is a common complication of CRF, and is associated with a decrease in the quality of life of the patients, as well as an increase in morbidity and mortality. Iron deficiency (absolute or functional) is common in non-dialysis chronic renal failure patients, and may cause anaemia or a low response to erythropoiesis-stimulating agents. For this reason, the clinical guidelines for the treatment of the anaemia in Nephrology advise the correction of the deficiency in the presence of anaemia. Iron replacement therapy is indicated in patients with CRF and anaemia (Hb < 12 g/dL) in accordance with the guidelines. There is no unanimity in the indication of iron replacement therapy in patients with Hb>12 g/dL, regardless of whether they have an absolute or functional iron deficiency. Intravenous iron replacement therapy is safe, more efficient and rapid than oral therapy for achieving an increase haemoglobin levels and reducing the dose of erythropoiesis-stimulating agents. For the administration of intravenous iron in non-dialysis chronic renal failure patients a strategy of high doses and low frequency would be preferred on being more convenient for the patient, better conserving of the venous tree, and on being safe and cost-effective. Iron plays an essential role in energy metabolism and other body functions beyond the synthesis of haemoglobin synthesis, for which the iron deficiency, even in the absence of anaemia, could have a harmful effect in patients with CRF. The correction of the iron deficiency, in the absence of anaemia is associated with functional improvement in patients with heart failure, and in muscle function or fatigue in patients without CRF. Despite the evidence of benefits in the correction of iron deficiency in patients with CRF, more studies are required to evaluate the impact of the correction of the iron deficiency in the absence of anaemia on morbidity and mortality, quality of life and physical capacity, as well as the long-term effect of oral and intravenous iron replacement therapy in this population. Copyright © 2020 Sociedad Española de Nefrología. Published by Elsevier España, S.L.U. All rights reserved. DOI: 10.1016/j.nefro.2020.11.003 PMID: 33516607
http://www.ncbi.nlm.nih.gov/pubmed/30970355
1. Acta Haematol. 2019;142(1):44-50. doi: 10.1159/000496492. Epub 2019 Apr 10. Iron Deficiency Anemia in Chronic Kidney Disease. Gafter-Gvili A(1)(2)(3), Schechter A(4), Rozen-Zvi B(5)(6). Author information: (1)Department of Medicine A, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel, [email protected]. (2)Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel, [email protected]. (3)Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel, [email protected]. (4)Department of Medicine A, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel. (5)Nephrology and Hypertension Unit, Rabin Medical Center, Petah Tikva, Israel. (6)Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. Iron deficiency anemia is a common complication of chronic kidney disease (CKD). CKD patients suffer from both absolute and functional iron deficiency. Absolute iron deficiency is defined by severely reduced or absent iron stores, while functional iron deficiency is defined by adequate iron stores but insufficient iron availability for incorporation into erythroid precursors. This is due to increased levels of hepcidin. Anemia in CKD is associated with an increased risk of morbidity and mortality. The association between anemia and mortality may be related to the severity of anemia. All CKD patients should be screened for anemia during the initial evaluation for CKD. Criteria used to define iron deficiency are different among CKD compared to normal renal function. Among CKD patients, absolute iron deficiency is defined when the transferrin saturation (TSAT) is ≤20% and the serum ferritin concentration is ≤100 ng/mL among predialysis and peritoneal dialysis patients or ≤200 ng/mL among hemodialysis patients. Functional iron deficiency, also known as iron-restricted erythropoiesis, is characterized by TSAT ≤20% and elevated ferritin levels. Iron supplementation is recommended for all CKD patients with anemia. There is general agreement according to guidelines that intravenous (i.v.) iron supplementation is the preferred method for CKD patients on dialysis (CKD stage 5D) and either i.v. or oral iron is recommended for patients with CKD ND (CKD stages 3-5). In this review we discuss the evidence base for these recommendations. © 2019 S. Karger AG, Basel. DOI: 10.1159/000496492 PMID: 30970355 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36165109
1. Nefrologia (Engl Ed). 2021 Jul-Aug;41(4):403-411. doi: 10.1016/j.nefroe.2021.10.007. Intravenous iron in heart failure and chronic kidney disease. Carrilho P(1). Author information: (1)Hospital Professor Doutor Fernando Fonseca EPE, Amadora, Portugal. Electronic address: [email protected]. Intravenous iron therapy is increasingly being used worldwide to treat anemia in chronic kidney disease and more recently iron deficiency in heart failure. Promising results were obtained in randomized clinical trials in the latter, showing symptomatic and functional capacity improvement with intravenous iron therapy. Meanwhile, confirmation of clinical benefit in hard-endpoints such as mortality and hospitalization is expected in large clinical trials that are already taking place. In chronic kidney disease, concern about iron overload is being substituted by claims of direct cardiovascular benefit of iron supplementation, as suggested by preliminary studies in heart failure. We discuss the pitfalls of present studies and gaps in knowledge, stressing the known differences between iron metabolism in heart and renal failure. Systemic and cellular iron handling and the role of hepcidin are reviewed, as well as the role of iron in atherosclerosis, especially in view of its relevance to patients undergoing dialysis. We summarize the evidence available concerning iron overload, availability and toxicity in CKD, that should be taken into account before embracing aggressive intravenous iron supplementation. Copyright © 2020 Sociedad Española de Nefrología. Published by Elsevier España, S.L.U. All rights reserved. DOI: 10.1016/j.nefroe.2021.10.007 PMID: 36165109 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31477258
1. Adv Chronic Kidney Dis. 2019 Jul;26(4):272-291. doi: 10.1053/j.ackd.2019.05.002. Novel Oral Iron Therapies for Iron Deficiency Anemia in Chronic Kidney Disease. Pergola PE(1), Fishbane S(2), Ganz T(3). Author information: (1)Renal Associates PA, San Antonio, TX. Electronic address: [email protected]. (2)Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY. (3)David Geffen School of Medicine, UCLA, Los Angeles, CA. Iron deficiency anemia (IDA) is a frequent complication of chronic kidney disease (CKD) and is associated with adverse outcomes in these patients. Patients with CKD and IDA remain largely undertreated. Conventional oral iron agents are insufficiently effective due to poor absorption and cause gastrointestinal side effects; thus, novel oral iron preparations are needed. This article covers current treatment guidelines for patients with anemia and CKD and clinical trial data for iron-repletion agents currently in use, as well as for novel oral iron therapies in development. Ferric citrate, a novel oral iron-repletion agent approved for patients with non-dialysis-dependent CKD and IDA, demonstrated improvements in hemoglobin levels and iron parameters, with good tolerability in patients with non-dialysis-dependent CKD. When used as a phosphate binder, ferric citrate also improves hemoglobin and iron parameters in dialysis-dependent CKD, but additional trials are needed to evaluate its efficacy as an iron-repletion agent in this setting. Other novel oral iron preparations in development for IDA in patients with CKD include ferric maltol, which is approved in Europe and the United States for IDA in adult patients, and sucrosomial iron, which has been evaluated in IDA associated with CKD and several other clinical settings. Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved. DOI: 10.1053/j.ackd.2019.05.002 PMID: 31477258 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28403561
1. Hemodial Int. 2017 Jun;21 Suppl 1:S78-S82. doi: 10.1111/hdi.12561. Epub 2017 Apr 12. Iron deficiency anemia in chronic kidney disease: Uncertainties and cautions. Agarwal R(1). Author information: (1)Department of Medicine, Indiana University School of Medicine and Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA. Anemia in chronic kidney disease is common and iron deficiency is an important cause. To repair iron-deficiency anemia, replacement of iron is needed. Iron can be replaced either by the oral route or by the intravenous route. In a meta-analysis, 5 of the 6 trials were short-term, 1 to 3 months, and compared to oral iron, the mean increase in hemoglobin with intravenous iron was only 0.31 g/dL. However, one of the studies included in this meta-analysis was 6 months long and had a mean decline in hemoglobin of 0.52 g/dL associated with intravenous iron administration. Given the short duration of most of the clinical trials comparing oral with intravenous administration of iron the long-term safety of these modes of administration of supplemental iron could not be assessed. Replacement of iron by the oral route is associated with mostly minor complications such as black stools, constipation, and abdominal discomfort. In contrast, intravenous administration of iron may lead to severe adverse events such as anaphylaxis and, as a more recent randomized trial has suggested, delayed complications such as infections and cardiovascular disease. Delayed complications of repeated intravenous iron use are difficult to recognize at an individual level therefore inpatients who have had recent cardiovascular events or are infected, intravenous iron should probably be avoided. Balancing safety and efficacy would require clinical judgment because 1 size may not fit all till we have better data to support the liberal use of parenteral iron. © Published 2017. This article is a U.S. Government work and is in the public domain in the USA. DOI: 10.1111/hdi.12561 PMID: 28403561 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28412770
1. Pediatr Nephrol. 2018 Feb;33(2):227-238. doi: 10.1007/s00467-017-3663-y. Epub 2017 Apr 15. Anemia in chronic kidney disease. Atkinson MA(1), Warady BA(2). Author information: (1)Division of Pediatric Nephrology, Johns Hopkins University School of Medicine, 200 N. Wolfe St, Baltimore, MD, 21287, USA. [email protected]. (2)Division of Pediatric Nephrology, Children's Mercy Kansas City, Kansas City, MO, USA. Anemia is common and associated with adverse outcomes in children with chronic kidney disease (CKD). Many factors contribute to declining hemoglobin as CKD progresses, but impaired production of erythropoietin by failing kidneys is a central cause. Hepcidin-mediated iron restriction also contributes to anemia by downregulating both intestinal iron absorption and release of stored iron for erythropoiesis. The core components of anemia management remain erythropoiesis-stimulating agents (ESA) and iron supplementation, but despite these therapies, a substantial number of children remain anemic. Although escalating ESA dose to target higher hemoglobin has been associated with adverse outcomes in adults, no trials have investigated this association in children, and maintaining hemoglobin levels in a narrow range with conservative ESA dosing is challenging. Judicious use of iron supplementation can enhance the response to ESAs, but the iron storage markers most commonly used in clinical practice have limitations in distinguishing which patients will benefit most from additional iron. Several novel anemia therapies, including hypoxia-inducible factor stabilizers, prolyl hydroxylase inhibitors, and dialysate-delivered iron supplements, have been developed and may offer options for alternative anemia management. However, the safety and efficacy of these agents in children with CKD has yet to be assessed. DOI: 10.1007/s00467-017-3663-y PMID: 28412770 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34514189
1. Kidney Int Rep. 2021 Jun 5;6(9):2261-2269. doi: 10.1016/j.ekir.2021.05.020. eCollection 2021 Sep. Treatment of Iron Deficiency Anemia in CKD and End-Stage Kidney Disease. Gutiérrez OM(1). Author information: (1)Division of Nephrology, Department of Medicine and Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA. Iron deficiency is common in individuals with chronic kidney disease and plays a major role in the development of anemia. Oral and intravenous iron agents are both available to replete iron in patients with chronic kidney disease diagnosed with iron deficiency. The choice of which agent to use is most often dictated by goals of therapy, tolerability, convenience, and response to prior therapy. Diminished absorption of iron in the gastrointestinal tract and a high incidence of gastrointestinal adverse effects can reduce the efficacy of oral iron agents, necessitating the use of i.v. iron formulations to treat iron deficiency anemia, particularly in patients requiring kidney replacement therapy. Newer oral agents may help to overcome these limitations and help treat iron deficiency in those not requiring kidney replacement therapy. Recent studies have provided new evidence that more aggressive repletion of iron in patients with chronic kidney disease requiring kidney replacement therapy may provide benefits with respect to anemia management and hard clinical outcomes such as cardiovascular disease and survival. © 2021 International Society of Nephrology. Published by Elsevier Inc. DOI: 10.1016/j.ekir.2021.05.020 PMCID: PMC8418942 PMID: 34514189
http://www.ncbi.nlm.nih.gov/pubmed/27236129
1. Semin Nephrol. 2016 Mar;36(2):94-8. doi: 10.1016/j.semnephrol.2016.02.002. Diagnosis of Iron-Deficiency Anemia in Chronic Kidney Disease. Bahrainwala J(1), Berns JS(2). Author information: (1)Renal, Electrolyte and Hypertension Division, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA. Electronic address: [email protected]. (2)Renal, Electrolyte and Hypertension Division, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA. Anemia is a common and clinically important consequence of chronic kidney disease (CKD). It is most commonly a result of decreased erythropoietin production by the kidneys and/or iron deficiency. Deciding on the appropriate treatment for anemia associated with CKD with iron replacement and erythropoietic-stimulating agents requires an ability to accurately diagnose iron-deficiency anemia. However, the diagnosis of iron-deficiency anemia in CKD patients is complicated by the relatively poor predictive ability of easily obtained routine serum iron indices (eg, ferritin and transferrin saturation) and more invasive gold standard measures of iron deficiency (eg, bone marrow iron stores) or erythropoietic response to supplemental iron. In this review, we discuss the diagnostic utility of currently used serum iron indices and emerging alternative markers of iron stores. Copyright © 2016 Elsevier Inc. All rights reserved. DOI: 10.1016/j.semnephrol.2016.02.002 PMID: 27236129 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35058395
1. Wiad Lek. 2021;74(12):3230-3233. NOVEL IRON BIOMARKERS IN CHRONIC KIDNEY DISEASE. Zapora-Kurel A(1), Malyszko J(2). Author information: (1)HYPERTENSION AND INTERNAL MEDICINE, MEDICAL UNIVERSITY OF BIALYSTOK, BIALYSTOK, POLAND. (2)NEPHROLOGY, DIALYSIS AND INTERNAL MEDICINE, WARSAW MEDICAL UNIVERSITY, WARSAW, POLAND. CKD is one of the fastest growing causes of death in the world and in 2040, it is estimated that it will be in the top five causes of death. In order to slow down this process, it is necessary to improve prevention, inhibit development and treat complications including anemia. Anemia is one of the common complication of chronic kidney disease (CKD), which is a significant clinical problem. It is most often the result of decreased renal production of erythropoietin and / or iron deficiency. Iron deficiency anemia is one of the most common problems in CKD that increases mortality. In order to successfully treat anemia in CKD with erythropoiesis-stimulating agentsand (ESA) and iron substitution, it is necessary to determine iron iron level. The diagnosis of iron deficiency anemia in patients with CKD is complicated due to the relatively low predictive ability of routine serum iron markers (e.g., ferritin and transferrin saturation) and more invasive measurements such as bone marrow iron stores. In the review novel biomarkers of iron metabolism are discussed such as hypoxia-inducible factor, erythroferon, growth differentiation factor 15 etc. with their possible clinical relevance. PMID: 35058395 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22303745
1. Rev Med Suisse. 2012 Jan 11;8(323):70-3. [Management of renal anemia in patients with chronic kidney disease: the role of the general practitioner]. [Article in French] Ruedin P(1), Dickenmann M, Martin PY, Wüthrich RP. Author information: (1)[email protected] The prevalence of chronic kidney disease (CKD) is high and diabetic nephropathy is a leading cause of CKD. One of the most common complications of CKD is anemia, the frequency and severity of which increase as kidney failure progresses. Renal anemia is primarily caused by reduced renal erythropoietin production. It can also be associated with iron deficiency caused by reduced iron absorption, occult blood loss and impaired iron mobilization. This work provides an overview of the management of renal anemia with focus on intravenous iron therapy, which is more effective than oral iron administration in CKD due to reduced iron absorption. PMID: 22303745 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/18824288
1. Am J Kidney Dis. 2008 Nov;52(5):907-15. doi: 10.1053/j.ajkd.2008.08.001. Epub 2008 Sep 27. Safety of ferumoxytol in patients with anemia and CKD. Singh A(1), Patel T, Hertel J, Bernardo M, Kausz A, Brenner L. Author information: (1)Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. Comment in Am J Kidney Dis. 2008 Nov;52(5):826-9. doi: 10.1053/j.ajkd.2008.09.006. BACKGROUND: Iron deficiency anemia is a common complication in patients with chronic kidney disease (CKD). Currently available intravenous (IV) iron replacement therapies have either inconvenient regimens of administration or adverse event profiles that limit their utility in the outpatient setting. Ferumoxytol is a novel, semisynthetic, carbohydrate-coated, superparamagnetic iron oxide nanoparticle that is administered IV as an injection. The main objective of this study was to assess the safety of ferumoxytol for the treatment of patients with CKD stages 1 to 5 and 5D. STUDY DESIGN: Phase 3, randomized, double-blind, placebo-controlled, crossover, multicenter study of a single 510-mg dose of ferumoxytol versus saline as placebo. SETTING & PARTICIPANTS: 750 patients with CKD stages 1 to 5 and 5D. INTERVENTION: An IV injection of either 17 mL of ferumoxytol or saline placebo over 17 seconds on day 0 and the alternate agent on day 7. OUTCOMES & MEASUREMENTS: Descriptive comparison of adverse events, laboratory tests, and vital signs. RESULTS: Of 750 randomly assigned patients with CKD, 60% were not on dialysis therapy. 713 patients received ferumoxytol, and 711 received placebo. There were 420 adverse events reported; 242 in 152 patients (21.3%) with ferumoxytol and 178 in 119 patients (16.7%) with placebo. The incidence of related adverse events was 5.2% with ferumoxytol and 4.5% with placebo. The most common related adverse events after each treatment included symptoms related to the injection/infusion site, dizziness, pruritus, headache, fatigue, and nausea. Serious adverse events occurred in 21 patients (2.9%) after ferumoxytol and 13 patients (1.8%) after placebo. Serious related adverse events were observed in 1 patient (0.1%) after each treatment. There was no meaningful decrease in blood pressure after administration of ferumoxytol or placebo. LIMITATIONS: Follow-up was 7 days after each study treatment. CONCLUSIONS: Ferumoxytol is well tolerated and has a safety profile similar to placebo in anemic patients with CKD stages 1 to 5 and 5D. DOI: 10.1053/j.ajkd.2008.08.001 PMID: 18824288 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20126670
1. Drugs Today (Barc). 2009 Nov;45(11):779-86. doi: 10.1358/dot.2009.45.11.1420459. Ferumoxytol for the treatment of anemia in chronic kidney disease. Rosner MH(1), Bolton WK. Author information: (1)Division of Nephrology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA. Iron deficiency anemia is a common occurrence in patients with chronic kidney disease and many patients do not respond well to supplementation with oral iron. There are now five intravenous iron preparations available for use in the chronic kidney disease patient, with ferumoxytol being the most recently approved agent. As opposed to previously available intravenous irons, ferumoxytol has the advantage of not needing a test dose, allowing a large dose of iron (510 mg) to be given in a short period of time by bolus injection, and no reported cases of anaphylaxis. Ferumoxytol has advantages for use in the outpatient setting to treat iron deficiency, in patients with chronic kidney disease not yet on dialysis and in patients on peritoneal dialysis. The use of ferumoxytol in the hemodialysis population where thrice weekly intravenous access is the norm is less clear. Cost-effectiveness studies and post-approval studies on ferumoxytol as well as changes in the cost structure of dialysis reimbursement will likely have a large impact on the use of this new agent. Copyright 2009 Prous Science, S.A.U. or its licensors. All rights reserved. DOI: 10.1358/dot.2009.45.11.1420459 PMID: 20126670 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/21684231
1. Nephrol Ther. 2012 Feb;8(1):41-6. doi: 10.1016/j.nephro.2011.04.005. Epub 2011 Jun 17. [Treatment of iron deficiency in predialysis state by low molecular weight iron dextran high doses intravenously]. [Article in French] Fievet P(1), Coppin M, Brazier F, Lefèvre M, Stephan R, Demontis R. Author information: (1)Service de néphrologie hémodialyse, centre hospitalier Laennec, boulevard Laennec, 60100 Creil, France. [email protected] Anemia is a common complication of chronic kidney disease (CKD) in predialysis stage. Iron deficiency is more common than in normal patients and plays a key role in the genesis of anemia. Its correction avoids the use of erythropoiesis stimulating agents (ESA) or reduces their dosage. Treatment with oral iron is often poorly tolerated and ineffective, necessitating the use of intravenous iron. New forms of injectable iron allow the use of high doses and correct iron deficiency in a single administration with consequent preservation of venous capital and lower costs. We studied the effectiveness of iron dextran of low molecular weight (LMWID) in high doses to correct iron deficiency and treat anemia in predialysis CKD patients. Twenty-nine doses of 500 to 1600 mg were administered to 25 patients followed for CKD (GFR between 60 and 10 ml/min per 1.73 m(2)), selected on biological criteria of iron deficiency defined by a ratio of transferrin saturation (TSAT) <20% and/or serum ferritin of less than 100 μg/L. Patients received treatment by ESA in 16 cases out of 29. One month after treatment, hemoglobin (Hb) increased significantly (11.4±1.6 vs 10.4±1.4 g/dL, P=0.0003) along with a significant increase in TSAT (21.3±7.3 vs 13.3±3.8%, P=0.000003) and serum ferritin (286±253 vs 91±60 μg/L, P=0.00005). Six patients had a serum ferritin greater than 500 μg/L after treatment, which may put them at risk of iron overload. Their serum ferritin was higher than the rest of the population before treatment, while the TSAT was no different, reflecting a functional deficiency. Their hemoglobin did not increase after treatment in contrast to the rest of the population suggesting the unavailability of iron for erythropoiesis with accumulation in the reticuloendothelial system. Renal function did not change significantly and there were no cases of acute renal failure. No immediate side effect was observed. Three patients presented delayed reactions to such self-limiting myalgia and arthralgia. No venous inflammatory reaction was noted. The administration of high doses of LMWID is effective in treating anemia of CKD in the predialysis stage with a satisfactory tolerance, without affecting kidney function and helps preserve the venous capital. It should be reserved for patients whose serum ferritin is less than or equal to 150 μg/L. Copyright © 2011 Association Société de néphrologie. Published by Elsevier SAS. All rights reserved. DOI: 10.1016/j.nephro.2011.04.005 PMID: 21684231 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17106764
1. Int Urol Nephrol. 2006;38(3-4):719-23. doi: 10.1007/s11255-006-0035-0. Iron deficiency in patients with chronic kidney disease: potential role for intravenous iron therapy independent of erythropoietin. Post JB(1), Wilkes BM, Michelis MF. Author information: (1)Division of Nephrology, Lenox Hill Hospital, 100 East 77th Street, New York, NY 10021, USA. [email protected] The prevalence of iron deficiency and its contribution to the anemia of end stage renal disease has been extensively studied, but much less is known about the role of iron deficiency in the pathogenesis of the anemia of chronic kidney disease in predialysis patients. All new hemodialysis patients entering a single hemodialysis unit between July 1999 and April 2002 were included in the study. The admission laboratory tests and the Health Care Financing Administration (HCFA) 2728 form were examined to determine the prevalence of erythropoietin use, anemia (Hb<11 g/dl), and iron deficiency (ferritin<100 ng/ml and transferrin saturation %<20%). In a second part of the study, the effect of intravenous iron gluconate replacement in patients with stage III & IV chronic kidney disease was examined. Anemia was present in 68% of all patients starting hemodialysis. Iron deficiency was a common feature occurring in 29% of patients taking erythropoietin (49% of all patients) and 26% of patients without erythropoietin (51% of all patients). Following the administration of intravenous iron gluconate to four patients, there was a significant rise in hemoglobin levels from 10.6+/-0.19 to 11.7+/-g/dl (p=0.02). CONCLUSION: Iron deficiency is common in predialysis patients. Replenishing iron stores in anemic patients with chronic kidney disease significantly increases hemoglobin levels and should be considered as an integral part of the therapy for treating anemia in the predialysis population. DOI: 10.1007/s11255-006-0035-0 PMID: 17106764 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29481308
1. FASEB J. 2018 Jul;32(7):3752-3764. doi: 10.1096/fj.201700667R. Epub 2018 Feb 26. Inhibition of fibroblast growth factor 23 (FGF23) signaling rescues renal anemia. Agoro R(1), Montagna A(1), Goetz R(2), Aligbe O(1), Singh G(1), Coe LM(1), Mohammadi M(2), Rivella S(3), Sitara D(1)(4). Author information: (1)Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA. (2)Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, USA. (3)Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; and. (4)Department of Medicine, New York University School of Medicine, New York, New York, USA. Severe anemia and iron deficiency are common complications in chronic kidney disease. The cause of renal anemia is multifactorial and includes decreased erythropoietin (Epo) production, iron deficiency, and inflammation, and it is currently treated with injections of synthetic Epo. However, the use of recombinant Epo has several adverse effects. We previously reported that high fibroblast growth factor 23 (FGF23) levels in mice are associated with decreased red blood cell production, whereas genetic inactivation of Fgf23 results in expansion of the erythroid lineage. The present study is the first to show that high FGF23 levels in a mouse model of renal failure contribute to renal anemia, and inhibiting FGF23 signaling stimulates erythropoiesis and abolishes anemia and iron deficiency. Moreover, we show that inhibition of FGF23 signaling significantly decreases erythroid cell apoptosis and influences the commitment of hematopoietic stem cells toward the erythroid linage. Furthermore, we show that blocking FGF23 signaling attenuates inflammation, resulting in increased serum iron and ferritin levels. Our data clearly demonstrate that elevated FGF23 is a causative factor in the development of renal anemia and iron deficiency, and importantly, blocking FGF23 signaling represents a novel approach to stimulate erythropoiesis and possibly improve survival for millions of chronic kidney disease patients worldwide.-Agoro, R., Montagna, A., Goetz, R., Aligbe, O., Singh, G., Coe, L. M., Mohammadi, M., Rivella, S., Sitara, D. Inhibition of fibroblast growth factor 23 (FGF23) signaling rescues renal anemia. DOI: 10.1096/fj.201700667R PMCID: PMC5998980 PMID: 29481308 [Indexed for MEDLINE] Conflict of interest statement: The authors thank M. N. T. Wada (University of Sao Paulo, Sao Paulo, Brazil and New York University School of Dentistry, New York, USA) and R. C. Bernstein (New York University College of Dentistry) for technical assistance. The authors are grateful to W. R. Abrams (New York University College of Dentistry) for invaluable assistance with the technical visualization of the data, and M. Gregory and P. Lopez (New York University School of Medicine) for assistance with flow cytometry and flow data analyses. This work was supported in part by funds from the American Heart Association (12SDG12080152) and the U.S. Department of Defense (W81XWH-16-1-0598; to D.S.), and the U.S. National Institutes of Health, National Institute of Dental and Craniofacial Research (DE 13686; to M.M.). The authors declare no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/20630409
1. Heart Fail Clin. 2010 Jul;6(3):347-57. doi: 10.1016/j.hfc.2010.02.001. Anemia in chronic kidney disease: new advances. Patel TV(1), Singh AK. Author information: (1)Renal Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA. Anemia resulting from iron and erythropoietin deficiencies is a common complication of advanced chronic kidney disease (CKD). This article covers major advances in our understanding of anemia in patients with CKD, including newly discovered regulatory molecules, such as hepcidin, to innovative intravenous iron therapies. The use of erythropoiesis-stimulating agents (ESA) in the treatment of anemia has undergone seismic shift in the past 3 years as a result of adverse outcomes associated with targeting higher hemoglobin levels with these agents. Potential mechanisms for adverse outcomes, such as higher mortality, are discussed. Despite the disappointing experience with ESAs, there is a tremendous interest in other novel agents to treat anemia in CKD. Lastly, while awaiting updated guidelines, the authors outline their recommendations on how to best manage patients who are anemic and have CKD. Copyright 2010. Published by Elsevier Inc. DOI: 10.1016/j.hfc.2010.02.001 PMID: 20630409 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29336855
1. Am J Kidney Dis. 2018 Mar;71(3):423-435. doi: 10.1053/j.ajkd.2017.09.026. Epub 2018 Jan 11. Update on Anemia in ESRD and Earlier Stages of CKD: Core Curriculum 2018. Fishbane S(1), Spinowitz B(2). Author information: (1)Division of Nephrology, Department of Medicine, Hofstra Northwell School of Medicine, Great Neck, NY. Electronic address: [email protected]. (2)Division of Nephrology, New York-Presbyterian/Queens, Flushing, NY. Anemia is a frequent complication during the later stages of chronic kidney disease. When present, it may cause symptoms such as fatigue and shortness of breath. The pathogenesis of anemia in chronic kidney disease is complex, but a central feature is a relative deficit of erythropoietin. New information has elucidated the critical role of the hypoxia-sensing system in mediating erythropoietin synthesis and release. Iron deficiency is a second important factor in the anemia of chronic kidney disease. New insights into the dynamics of iron metabolism have clarified the role of chronic inflammation and hepcidin as key mediators of impaired iron utilization. In this article, we review the epidemiology, pathobiology, clinical evaluation, and treatment of anemia in chronic kidney disease. Copyright © 2017 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved. DOI: 10.1053/j.ajkd.2017.09.026 PMID: 29336855 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28372549
1. BMC Nephrol. 2017 Apr 3;18(1):117. doi: 10.1186/s12882-017-0523-8. The Ferumoxytol for Anemia of CKD Trial (FACT)-a randomized controlled trial of repeated doses of ferumoxytol or iron sucrose in patients on hemodialysis: background and rationale. Macdougall IC(1), Dahl NV(2), Bernard K(2), Li Z(2), Batycky A(2), Strauss WE(3). Author information: (1)Department of Renal Medicine, King's College Hospital, Denmark Hill, London, UK. (2)AMAG Pharmaceuticals, Inc., 1100 Winter Street, Waltham, MA, 02451, USA. (3)AMAG Pharmaceuticals, Inc., 1100 Winter Street, Waltham, MA, 02451, USA. [email protected]. Erratum in BMC Nephrol. 2018 Apr 26;19(1):97. doi: 10.1186/s12882-018-0899-0. BACKGROUND: Iron deficiency anemia (IDA) is a common manifestation of chronic kidney disease (CKD), affecting most patients on hemodialysis and imposing a substantial clinical burden. Treatment with iron supplementation increases hemoglobin levels and can reduce the severity of anemia in patients with CKD. While correcting anemia in these patients is an important therapeutic goal, there is a lack of long-term trials directly comparing intravenous iron therapies in patients with CKD receiving hemodialysis. METHODS/DESIGN: The Ferumoxytol for Anemia of CKD Trial (FACT) is a 13-month, open-label, randomized, multicenter, international, prospective study with 2 substudies. Entry criteria for the main study include adults with IDA (defined as hemoglobin <11.5 g/dL [<115.0 g/L] and a transferrin saturation <30%), serum ferritin <800 ng/mL (<1798 pmol/L), and receiving hemodialysis for ≥3 months. Patients are randomized to receive ferumoxytol (1.02 g over 2 doses) or iron sucrose (1.0 g over 10 doses) during the initial 5-week treatment period. Those with persistent/recurrent IDA over the 11-month observation period will receive additional 5-week treatment periods, as appropriate. The primary efficacy endpoint of the main study is the mean change in hemoglobin from Baseline to Week 5 for each treatment period. The secondary efficacy endpoints include the mean change in transferrin saturation from Baseline to Week 5 and the proportion of patients with a hemoglobin increase of ≥1.0 g/dL at any time from Baseline to Week 5. Safety will be assessed through an examination of the adverse event profile over the course of the study. An "oxidative stress" substudy in approximately 100 patients will assess the effects of treatment on biomarkers of oxidative stress/inflammation during the initial 5-week treatment period, and a magnetic resonance imaging substudy in approximately 70 patients will assess the potential for iron deposition in target tissues over 24 months. DISCUSSION: FACT fulfills the need for a long-term comparative trial in patients with IDA and CKD receiving hemodialysis. The efficacy and safety results will provide useful information for guiding therapy in this population. Two hundred ninety-six patients have been enrolled, and completion of the main study is expected soon. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01227616 (registered October 22, 2010); EudraCT number: 2010-022133-28. DOI: 10.1186/s12882-017-0523-8 PMCID: PMC5379516 PMID: 28372549 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/19776721
1. Kidney Int. 2009 Dec;76(11):1137-41. doi: 10.1038/ki.2009.357. Epub 2009 Sep 23. Iron-refractory iron deficiency anemia: new molecular mechanisms. Cui Y(1), Wu Q, Zhou Y. Author information: (1)Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, Suzhou, China. Iron deficiency anemia is a common complication in end-stage renal disease (ESRD) and impairs the therapeutic efficacy of recombinant erythropoietin. Oral or parental iron supplements usually are effective in treating iron deficiency anemia. Some patients, however, respond poorly to iron supplements and are diagnosed as having iron-refractory iron deficiency anemia. The condition exacerbates ESRD but its underlying mechanism was unclear. Hepcidin is a central player in iron homeostasis. It downregulates the iron exporter ferroportin, thereby inhibiting iron absorption, release, and recycling. In ESRD, plasma hepcidin levels are elevated, which contributes to iron deficiency in patients. Matriptase-2, a liver transmembrane serine protease, has been found to have a major role in controlling hepcidin gene expression. In mice, defects in the Tmprss6 gene encoding matriptase-2 result in high hepcidin expression and cause severe microcytic anemia. Similarly, mutations in the human TMPRSS6 gene have been identified in patients with iron-refractory iron deficiency. Thus, matriptase-2 is critical for iron homeostasis and may have an important role in ESRD. DOI: 10.1038/ki.2009.357 PMCID: PMC2869468 PMID: 19776721 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25468387
1. Am J Kidney Dis. 2015 May;65(5):728-36. doi: 10.1053/j.ajkd.2014.10.014. Epub 2014 Nov 4. A 12-week, double-blind, placebo-controlled trial of ferric citrate for the treatment of iron deficiency anemia and reduction of serum phosphate in patients with CKD Stages 3-5. Block GA(1), Fishbane S(2), Rodriguez M(3), Smits G(4), Shemesh S(5), Pergola PE(6), Wolf M(7), Chertow GM(8). Author information: (1)Denver Nephrologists PC, Denver, CO. Electronic address: [email protected]. (2)Hofstra-North Shore LIJ School of Medicine, Great Neck, NY. (3)Nephrology Service, IMIBIC, Hospital Universitario, Cordoba, Spain. (4)Denver Nephrologists PC, Denver, CO. (5)Keryx Biopharmaceuticals Inc, New York, NY. (6)Renal Associates PA, San Antonio, TX. (7)Department of Medicine, Institute for Public Health and Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL. (8)Stanford University School of Medicine, Palo Alto, CA. BACKGROUND: Iron deficiency anemia and serum phosphate levels > 4.0mg/dL are relatively common in chronic kidney disease stages 3 to 5 and are associated with higher risks of progressive loss of kidney function, cardiovascular events, and mortality. STUDY DESIGN: Double-blind, placebo-controlled, randomized trial. SETTING & PARTICIPANTS: 149 patients with estimated glomerular filtration rates < 60 mL/min/1.73 m(2), iron deficiency anemia (hemoglobin, 9.0-12.0 g/dL; transferrin saturation [TSAT]≤ 30%, serum ferritin ≤ 300 ng/mL), and serum phosphate levels ≥ 4.0 to 6.0mg/dL. Use of intravenous iron or erythropoiesis-stimulating agents was prohibited. INTERVENTION: Randomization to treatment for 12 weeks with ferric citrate coordination complex (ferric citrate) or placebo. OUTCOMES & MEASUREMENTS: Coprimary end points were change in TSAT and serum phosphate level from baseline to end of study. Secondary outcomes included change from baseline to end of treatment in values for ferritin, hemoglobin, intact fibroblast growth factor 23 (FGF-23), urinary phosphate excretion, and estimated glomerular filtration rate. RESULTS: Ferric citrate treatment increased mean TSAT from 22% ± 7% (SD) to 32% ± 14% and reduced serum phosphate levels from 4.5 ± 0.6 to 3.9 ± 0.6 mg/dL, while placebo exerted no effect on TSAT (21% ± 8% to 20% ± 8%) and less effect on serum phosphate level (4.7 ± 0.6 to 4.4 ± 0.8 mg/dL; between-group P<0.001 for each). Ferric citrate increased hemoglobin levels (from 10.5 ± 0.8 to 11.0 ± 1.0 g/dL; P<0.001 vs placebo), reduced urinary phosphate excretion 39% (P<0.001 vs placebo), and reduced serum intact FGF-23 levels from a median of 159 (IQR, 102-289) to 105 (IQR, 65-187) pg/mL (P=0.02 vs placebo). The incidence and severity of adverse effects were similar between treatment arms. LIMITATIONS: The study is limited by relatively small sample size and short duration and by having biochemical rather than clinical outcomes. CONCLUSIONS: Short-term use of ferric citrate repletes iron stores, increases hemoglobin levels, and reduces levels of serum phosphate, urinary phosphate excretion, and FGF-23 in patients with chronic kidney disease stages 3 to 5. Copyright © 2015 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved. DOI: 10.1053/j.ajkd.2014.10.014 PMID: 25468387 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32556307
1. Blood. 2020 Aug 13;136(7):783-789. doi: 10.1182/blood.2019004330. How I treat renal anemia. Fishbane S(1), Coyne DW(2). Author information: (1)Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY; and. (2)Division of Nephrology, School of Medicine, Washington University, St. Louis, MO. 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. © 2020 by The American Society of Hematology. DOI: 10.1182/blood.2019004330 PMID: 32556307 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26342303
1. Blood Rev. 2016 Jan;30(1):65-72. doi: 10.1016/j.blre.2015.07.006. Epub 2015 Aug 18. Iron therapy in chronic kidney disease: Recent changes, benefits and risks. Ribeiro S(1), Belo L(2), Reis F(3), Santos-Silva A(4). Author information: (1)Research Unit on Applied Molecular Biosciences (UCIBIO), REQUIMTE, Department of Biological Sciences, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal. Electronic address: [email protected]. (2)Research Unit on Applied Molecular Biosciences (UCIBIO), REQUIMTE, Department of Biological Sciences, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal. Electronic address: [email protected]. (3)Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Unidade 1, Polo 3, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit, University of Coimbra, Polo 1, First floor, Rua Larga, 3004-504 Coimbra, Portugal. Electronic address: [email protected]. (4)Research Unit on Applied Molecular Biosciences (UCIBIO), REQUIMTE, Department of Biological Sciences, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal. Electronic address: [email protected]. Anemia is a common complication in patients with chronic kidney disease (CKD), mainly due to inadequate renal production of erythropoietin. In hemodialysis (HD) patients this condition may be aggravated by iron deficiency (absolute or functional). The correction of this anemia is usually achieved by treatment with erythropoiesis stimulating agents (ESAs) and iron (oral or intravenous). Studies questioning the safety of ESAs (especially at higher doses) changed the pattern of anemia treatment in CKD patients. According to the new guidelines, when transferrin saturation is lower than 30% and ferritin lower than 500 ng/mL, a trial with iron should be started, to avoid therapy with ESAs or at least to reduce the doses needed to treat the anemia. Recent reports showed increasing ferritin levels, towards values above 800 ng/mL, in CKD patients treated according to the guidelines. In this review we focus on the risks of the increased iron use to treat CKD anemia, namely, iron overload and toxicity, increased risk of infections, as well as mortality. DOI: 10.1016/j.blre.2015.07.006 PMID: 26342303 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/19833421
1. Blood Rev. 2010 Jan;24(1):39-47. doi: 10.1016/j.blre.2009.09.001. Epub 2009 Oct 14. Anemia in renal disease: diagnosis and management. Lankhorst CE(1), Wish JB. Author information: (1)Division of Nephrology, University Hospitals Case Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, USA. [email protected] Chronic kidney disease (CKD) is a widespread health problem in the world and anemia is a common complication. Anemia conveys significant risk for cardiovascular disease, faster progression of renal failure and decreased quality of life. Patients with CKD can have anemia for many reasons, including but not invariably their renal insufficiency. These patients require a thorough evaluation to identify and correct causes of anemia other than erythropoietin deficiency. The mainstay of treatment of anemia secondary to CKD has become erythropoiesis-stimulating agents (ESAs). The use of ESAs does carry risks and these agents need to be used judiciously. Iron deficiency often co-exists in this population and must be evaluated and treated. Correction of iron deficiency can improve anemia and reduce ESA requirements. Partial, but not complete, correction of anemia is associated with improved outcomes in patients with CKD. Copyright 2009 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.blre.2009.09.001 PMID: 19833421 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16470356
1. Internist (Berl). 2006 Mar;47(3):233-4, 236-8, 240-1. doi: 10.1007/s00108-006-1576-0. [Renal anemia - an important secondary disease in renal insufficiency]. [Article in German] Mayer C(1), Achenbach H, Stumvoll M, Fiedler G. Author information: (1)Abteilung für Endokrinologie, Diabetologie und Nephrologie, Medizinische Klinik und Poliklinik III, Universität Leipzig, Leipzig. [email protected] Anemia is as a frequent complication in patients with chronic kidney disease, which gains in importance in the treatment of patients with renal disease. The main cause of renal anemia is the inadequately low production of endogenous erythropoietin. Often the patients develop an additional absolute or functional iron deficiency, which complicates the diagnostic and therapeutic procedures. Substitution of recombinant human erythropoietin (r-HuEPO) is the most effective therapy. The goal is a stable haemoglobin level >11 g/dl. An often additional existing iron deficiency should be balanced adequately according to the guidelines. With consequent and early treatment morbidity, mortality, and quality of life can be effectively improved. DOI: 10.1007/s00108-006-1576-0 PMID: 16470356 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16949463
1. Semin Nephrol. 2006 Jul;26(4):261-8. doi: 10.1016/j.semnephrol.2006.06.001. Pathogenesis of renal anemia. Nangaku M(1), Eckardt KU. Author information: (1)Division of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo, Japan. [email protected] Anemia is a common complication of chronic kidney disease. Although mechanisms involved in the pathogenesis of renal anemia include chronic inflammation, iron deficiency, and shortened half-life of erythrocytes, the primary cause is deficiency of erythropoietin (EPO). Serum EPO levels in patients with chronic kidney disease are usually within the normal range and thus fail to show an appropriate increase with decreasing hemoglobin levels, as found in nonrenal anemias. Studies elucidating the regulation of EPO expression led to the identification of the hypoxia inducible factor-hypoxia responsive element system. However, despite much progress in understanding the molecular mechanisms through which cells can sense oxygen availability and translate this information into altered gene expression, the reason why EPO production is inappropriately low in diseased kidneys remains incompletely understood. Both alterations in the function of EPO-producing cells and perturbations of the oxygen-sensing mechanism in the kidney may contribute. As with other anemias, the consequences of renal anemia are a moderate decrease in tissue oxygen tensions and counterregulatory mechanisms that maintain total oxygen consumption, including a persistent increase in cardiac output. DOI: 10.1016/j.semnephrol.2006.06.001 PMID: 16949463 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17804903
1. Am J Nephrol. 2007;27(6):565-71. doi: 10.1159/000107927. Epub 2007 Sep 5. Nonhematological benefits of iron. Agarwal R(1). Author information: (1)Division of Nephrology, Department of Medicine, Indiana University School of Medicine, and Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA. [email protected] Iron deficiency anemia is common in people with chronic kidney disease (CKD) and its importance in supporting erythropoiesis is unquestioned especially in those patients treated with erythropoietin. Clinical symptomatology such as fatigability, cold intolerance, failure to concentrate and poor effort intolerance is often attributed to anemia or uremia. That iron deficiency, per se, can cause these symptoms is poorly recognized. Clinical and animal studies that support the benefits of iron supplementation, independent of increasing hemoglobin, such as those on immune function, physical performance, thermoregulation, cognition, and restless leg syndrome and aluminum absorption is the subject of this narrative review. (c) 2007 S. Karger AG, Basel. DOI: 10.1159/000107927 PMID: 17804903 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28682026
1. G Ital Nefrol. 2017 Mar;34(Suppl 69):20-35. [Clinical management of anemia in patients with CKD]. [Article in Italian] Rivera RF(1), Alibrandi MTS(2), Di Lullo L(3), Fioccari F(4). Author information: (1)U.O. Nefrologia e Dialisi, Ospedale San Gerardo, ASST Monza, Italy. (2)U.O. Nefrologia, Dialisi e Ipertensione, IRCCS Ospedale San Raffaele, Milan, Italy. (3)U.O.C Nefrologia e Dialisi, Ospedale L. Parodi Delfino, Colleferro, Rome, Italy. (4)U.O.C. Nefrologia e Dialisi, Ospedale S. Paolo, Civitavecchia, Rome, Italy. Anemia is a frequent complication in chronic kidney disease (CKD), and it is often accompanied by various clinical symptoms. The primary cause of anemia in CKD patients is the reduction in the erythropoietin production, which results in a decrease of signaling molecule that stimulates red blood cell production. Other possible causes of anemia in CKD include iron deficiency, inflammation, and the accumulation of uremic toxin. This chapter focuses the discussion on the strategy of the management of anemia in patients with CKD. Erythropoiesis-stimulating agents (ESAs) and adjuvant iron therapy represent the primary treatment for anemia in chronic kidney disease. The introduction of ESAs into clinical practice was a success goal, mediating an increase in hemoglobin concentrations without the risk for recurrent blood transfusions and improving quality of life substantially. Copyright by Società Italiana di Nefrologia SIN, Rome, Italy. PMID: 28682026 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/19325171
1. Clin Med Res. 2008 Dec;6(3-4):93-102. doi: 10.3121/cmr.2008.811. Safety issues with intravenous iron products in the management of anemia in chronic kidney disease. Hayat A(1). Author information: (1)SUNY Downstate Medical Center, 710 Parkside Avenue, Brooklyn, NY 11226, USA. [email protected] Anemia is a very common clinical problem in patients with chronic kidney disease (CKD) and is associated with increased morbidity and mortality in these patients. Erythropoietin is a hormone synthesized that is deficient in the majority of patients with advanced kidney disease, thereby predisposing these patients to anemia. The other cause of anemia is deficiency of iron. Iron deficiency anemia is common in people with CKD and its importance in supporting erythropoiesis is unquestioned, especially in those patients treated with erythropoietin. Intravenous iron is frequently used to treat anemia in CKD patients and is very efficacious in increasing hemoglobin but at the same time there are some safety issues associated with it. The objective of this review is to assess the frequency of adverse drug events associated with four different iron formulations: two iron dextran products known as high and low molecular weight iron dextran, iron sucrose, and sodium ferric gluconate complex. Several electronic databases were searched. In general, with the exception of high molecular weight iron dextran, serious or life-threatening adverse events appeared rare. Iron sucrose has the least reported adverse events and high molecular weight iron dextran has the highest number of reported adverse events. Low molecular weight iron dextran and ferric gluconate fall in between these two for number of adverse drug events. DOI: 10.3121/cmr.2008.811 PMCID: PMC2670525 PMID: 19325171 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17533016
1. Am J Kidney Dis. 2007 Jun;49(6):736-43. doi: 10.1053/j.ajkd.2007.03.007. Iron management in nondialysis-dependent CKD. Fishbane S(1). Author information: (1)SUNY at Stony Brook School of Medicine, USA. [email protected] Iron deficiency has been studied extensively in patients with chronic kidney disease on hemodialysis therapy. However, few studies looked at iron treatment in the nondialysis chronic kidney disease population. Limited data suggest that iron deficiency is common in patients with chronic kidney disease with anemia; this lack of iron can hinder the effectiveness of erythropoiesis. The diagnosis of iron deficiency should involve clinical judgment, with an emphasis on clinical characteristics of the patient because of the limited amount of literature examining the interpretation of iron testing results. When iron deficiency is diagnosed in nondialysis patients with chronic kidney disease, a search must be initiated for any sources of blood loss. After addressing any blood loss, the preferred route of iron treatment must be determined. To date, no clear advantage was shown with intravenous versus oral administration in nondialysis patients, as shown in the hemodialysis setting. Thus, oral iron therapy may be a more reasonable option unless oral therapy previously failed. Additional research is needed to support evidence-based guidelines for the treatment of iron deficiency in the nondialysis chronic kidney disease population because this population differs from hemodialysis patients in the decreased extent of blood loss. DOI: 10.1053/j.ajkd.2007.03.007 PMID: 17533016 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22935483
1. J Am Soc Nephrol. 2012 Oct;23(10):1631-4. doi: 10.1681/ASN.2011111078. Epub 2012 Aug 30. Mechanisms of anemia in CKD. Babitt JL(1), Lin HY. Author information: (1)Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA. [email protected] Anemia is a common feature of CKD associated with poor outcomes. The current management of patients with anemia in CKD is controversial, with recent clinical trials demonstrating increased morbidity and mortality related to erythropoiesis stimulating agents. Here, we examine recent insights into the molecular mechanisms underlying anemia of CKD. These insights hold promise for the development of new diagnostic tests and therapies that directly target the pathophysiologic processes underlying this form of anemia. DOI: 10.1681/ASN.2011111078 PMCID: PMC3458456 PMID: 22935483 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23608591
1. Heart Rhythm. 2013 Aug;10(8):1220-8. doi: 10.1016/j.hrthm.2013.04.014. Epub 2013 Apr 19. Modulation of KCNQ1 alternative splicing regulates cardiac IKs and action potential repolarization. Lee HC(1), Rudy Y, Po-Yuan P, Sheu SH, Chang JG, Cui J. Author information: (1)Department of Biomedical Engineering, Cardiac Bioelectricity and Arrhythmia Center, Washington University in St. Louis, St Louis, Missouri 63130-4899, USA. Comment in Heart Rhythm. 2013 Aug;10(8):1229-30. doi: 10.1016/j.hrthm.2013.06.017. Heart Rhythm. 2013 Nov;10(11):e82-3. doi: 10.1016/j.hrthm.2013.09.056. Heart Rhythm. 2013 Nov;10(11):e83. doi: 10.1016/j.hrthm.2013.09.055. BACKGROUND: Slow delayed-rectifier potassium current (IKs) channels, made of the pore-forming KCNQ1 and auxiliary KCNE1 subunits, play a key role in determining action potential duration (APD) in cardiac myocytes. The consequences of drug-induced KCNQ1 splice alteration remain unknown. OBJECTIVE: To study the modulation of KCNQ1 alternative splicing by amiloride and the consequent changes in IKs and action potentials (APs) in ventricular myocytes. METHODS: Canine endocardial, midmyocardial, and epicardial ventricular myocytes were isolated. Levels of KCNQ1a and KCNQ1b as well as a series of splicing factors were quantified by using the reverse transcriptase-polymerase chain reaction and Western blot. The effect of amiloride-induced changes in the KCNQ1b/total KCNQ1 ratio on AP was measured by using whole-cell patch clamp with and without isoproterenol. RESULTS: With 50 μmol/L of amiloride for 6 hours, KCNQ1a at transcriptional and translational levels increased in midmyocardial myocytes but decreased in endo- and epicardial myocytes. Likewise, changes in splicing factors in midmyocardial were opposite to that in endo- and epicardial myocytes. In midmyocardial myocytes amiloride shortened APD and decreased isoproterenol-induced early afterdepolarizations significantly. The same amiloride-induced effects were demonstrated by using human ventricular myocyte model for AP simulations under beta-adrenergic stimulation. Moreover, amiloride reduced the transmural dispersion of repolarization in pseudo-electrocardiogram. CONCLUSIONS: Amiloride regulates IKs and APs with transmural differences and reduces arrhythmogenicity through the modulation of KCNQ1 splicing. We suggested that the modulation of KCNQ1 splicing may help prevent arrhythmia. Copyright © 2013 Heart Rhythm Society. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.hrthm.2013.04.014 PMCID: PMC3771516 PMID: 23608591 [Indexed for MEDLINE] Conflict of interest statement: CONFLICTS OF INTEREST: none
http://www.ncbi.nlm.nih.gov/pubmed/35268286
1. J Clin Med. 2022 Feb 23;11(5):1200. doi: 10.3390/jcm11051200. Catamenial Pneumothorax as the First Expression of Thoracic Endometriosis Syndrome and Pelvic Endometriosis. Ciriaco P(1), Muriana P(1), Carretta A(1), Ottolina J(2), Candiani M(2), Negri G(1). Author information: (1)Department of Thoracic Surgery, Scientific Institute and University Vita-Salute San Raffaele, Hospital San Raffaele Milano, 20132 Milan, Italy. (2)Department of Obstetrics and Gynecology, Scientific Institute and University Vita-Salute San Raffaele, Hospital San Raffaele Milano, 20132 Milan, Italy. OBJECTIVE: The menstrual-related catamenial pneumothorax (CP) can be the first expression of thoracic endometriosis syndrome (TES), which is the presence of endometriotic lesions in the lungs and pleura, and pelvic endometriosis (PE). This study aims to analyze our experience with this specific correlation describing our multidisciplinary approach to CP. METHODS: Hospital records of 32 women, operated for CP at our Department from January 2001 to December 2021 were reviewed. Surgical treatment consisted of videothoracoscopy and laparoscopy when indicated. RESULTS: TES and PE were diagnosed in 13 (40.6%) and 12 (37.5%) women, respectively. The association of TES and PE was present in 11 cases (34%). Fifteen patients (46.9%) underwent laparoscopy, of which 11 concurrently with videothoracoscopy. Most of the patients affected had stage III-IV endometriosis (40.6%). All patients received hormonal therapy after surgery. Five patients with PE conceived spontaneously resulting in six live births. The mean follow-up was 117 ± 71 months (range 8-244). Pneumothorax recurrence occurred in six patients (18.8%). At present, all women are asymptomatic, with no sign of pneumothorax recurrence. CONCLUSIONS: CP might be the first expression of TES and/or PE. A multidisciplinary approach is advocated for optimal management of the disease. DOI: 10.3390/jcm11051200 PMCID: PMC8911039 PMID: 35268286 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35286587
1. Gen Thorac Cardiovasc Surg. 2022 Sep;70(9):818-824. doi: 10.1007/s11748-022-01802-w. Epub 2022 Mar 14. The importance of diaphragmatic surgery, chemical pleurodesis and postoperative hormonal therapy in preventing recurrence in catamenial pneumothorax: a retrospective cohort study. Campisi A(#)(1)(2), Ciarrocchi AP(#)(3), Grani G(3), Sanna S(3), Congiu S(3), Mazzarra S(3), Argnani D(3), Salvi M(3), Stella F(3). Author information: (1)Thoracic Surgery Unit, Department of Thoracic Diseases, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, 34 Carlo Forlanini Street, 47121, Forlì, Italy. [email protected]. (2)Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huai Hai Road, Shanghai, 200030, China. [email protected]. (3)Thoracic Surgery Unit, Department of Thoracic Diseases, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, 34 Carlo Forlanini Street, 47121, Forlì, Italy. (#)Contributed equally BACKGROUND: Catamenial pneumothorax (CP) is defined as a recurrent, spontaneous pneumothorax occurring within a day before or 72 h after the onset of menstruation. Most first episodes go undiagnosed and treated as primary spontaneous pneumothorax, and only after recurrence is the clinical suspicion of CP raised. No gold-standard management approach exists, especially in terms of managing diaphragmatic involvement. METHODS: This study is a single-centre cohort retrospective study of 24 female patients who underwent surgery for pneumothorax due to diaphragmatic endometriosis between January 2008 and December 2016. Two groups were compared: a group that underwent pleurodesis alone (8 patients) and a group that underwent diaphragmatic surgery and pleurodesis (16 patients). RESULTS: There were differences in BMI and smoking habits between the two groups. The right diaphragm was involved more often (6vs15, p = 0.190). VATS was the preferred surgical approach and only one conversion occurred in the diaphragmatic surgery group (p = 0.470). Diaphragmatic abnormalities were present in all the patients, brown/violet spots (100%) in the pleurodesis group and perforations (100%) in the diaphragmatic surgery group (p < 0.001). There were no differences in days of chest tube removal and length of stay. The recurrence rate was 100% in the pleurodesis alone group while it was only 12.5% in the diaphragmatic surgery group (< 0.001). CONCLUSIONS: In our experience, diaphragmatic surgery and pleurodesis followed by hormonal therapy was an effective approach in preventing recurrence in patients with catamenial pneumothorax and diaphragmatic involvement. © 2022. The Author(s), under exclusive licence to The Japanese Association for Thoracic Surgery. DOI: 10.1007/s11748-022-01802-w PMID: 35286587 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35774053
1. Radiol Case Rep. 2022 Jun 25;17(9):3119-3125. doi: 10.1016/j.radcr.2022.06.012. eCollection 2022 Sep. Catamenial pneumothorax: a rare manifestation of endometriosis. Lameira P(1), Abecasis M(1), Palma S(1), Leitão J(1). Author information: (1)Department of Radiology, Centro Hospitalar Universitário Lisboa Norte, Serviço de Imagiologia Geral, Av. Prof. Egas Moniz MB, 1649-028 Lisboa, Portugal. Endometriosis is a common gynecological disease that primarily affects premenopausal women. It is mainly found in the pelvis but may be found at several extrapelvic locations. Thoracic endometriosis is a rare extrapelvic location of endometriosis and the leading cause of catamenial pneumothorax. We describe the case of a 35-year-old woman with a background of pelvic pain presenting to the emergency department with chest pain and dyspnea. The chest X-ray in the emergency department showed a large right-sided pneumothorax. Further imaging studies during patient evaluation revealed extensive fibrotic changes in the pelvis and well-defined solid nodules with high signal on T2 and T1-weighted images on MRI in abdominal and thoracic locations, rendering the diagnosis of a catamenial pneumothorax in a patient with pelvic, abdominal and thoracic endometriosis. © 2022 The Authors. Published by Elsevier Inc. on behalf of University of Washington. DOI: 10.1016/j.radcr.2022.06.012 PMCID: PMC9237952 PMID: 35774053
http://www.ncbi.nlm.nih.gov/pubmed/36179536
1. Eur J Obstet Gynecol Reprod Biol. 2022 Nov;278:141-147. doi: 10.1016/j.ejogrb.2022.09.019. Epub 2022 Sep 21. Is hormonal manipulation after surgical treatment of catamenial pneumothorax effective in reducing the rate of recurrence? A systematic review and meta-analysis. Elsayed HH(1), Hassaballa AS(2), Mostafa MH(3), El Ghanam M(2), Ahmed MH(2), Gumaa M(4), Moharram AA(5). Author information: (1)Thoracic Surgery Department, Ain Shams University, Cairo, Egypt. Electronic address: [email protected]. (2)Cardiothoracic Surgery Department, Ain Shams University, Cairo, Egypt. (3)Obstetrics and Gynecology Department, Ain Shams University, Cairo, Egypt. (4)TRUST Research Centre, Cairo, Egypt. (5)Department of Anaethesia, Intensive Care and Pain Management, Ain Shams University, Cairo, Egypt. OBJECTIVES: Catamenial pneumothorax CP is a rare form of spontaneous pneumothorax in females forming part of thoracic endometriosis syndrome. Studies have suggested possible benefit from postoperative hormonal administration. As this treatment is inconsistent, we aimed at performing the first meta-analysis to study the efficacy of adding hormonal treatment after surgery to reduce the chances of recurrent catamenial pneumothorax. METHODS: CENTRAL, MEDLINE/PubMed, Cochrane Library, and Scopus were systematically searched from inception up to December 15, 2021. Studies reporting five or more patients with end point outcome were included. The main outcome assessed was postoperative recurrence of CP after hormonal manipulation. Baseline, procedural, outcome, and validity data were systematically appraised and pooled with random-effect methods. meta- regression for the effect of patient age and follow up period were tested. Publication bias was examined. This trial was registered with PROSPERO under registration number CRD42022325377. RESULTS: Our electronic search retrieved 644 citations, 48 of which were selected for full-text review. Eleven studies with a combined population of 111 patients fulfilled the inclusion criteria. All patients reached an endpoint of follow up for postoperative recurrence of catamenial pneumothorax after receiving hormonal treatment. Overall study validity was acceptable, with a median score of 6 on the Newcastle Ottawa scale NOS appraising the quality of observational studies. CP is almost always a right-side disease (107/111 = 96.3 %). The risk of postoperative recurrence with hormonal treatment was 17.3 % (8.9 - 25.8 %) with moderate non-significant heterogeneity (I2 = 40.85 %; P = 0.076). The cumulative risk of recurrence for all patients not receiving postoperative hormonal therapy included in our study was 54.2 % (19/35 patients). Meta regression showed age to be a significant predictor of postoperative recurrence (p = 0.03). As the age increases one year, the risk of recurrence decreases by 6 % (0.2 - 3 %). Publication bias was detected by visualizing the funnel plot of standard error, Egger's test with p < 0.01 and Begg & Mazumdar test with p < 0.01. CONCLUSION: The study included the largest number of CP patients with outcome findings of postoperative recurrence with hormonal treatment despite the small number of studies, non-randomised fashion and publication bias. Our findings recommend the use of hormonal manipulation after thoracic surgical intervention for catamenial pneumothorax unless evident contraindications. Younger patients are at a higher risk of recurrence after surgery. Copyright © 2022 Elsevier B.V. All rights reserved. DOI: 10.1016/j.ejogrb.2022.09.019 PMID: 36179536 [Indexed for MEDLINE] Conflict of interest statement: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
http://www.ncbi.nlm.nih.gov/pubmed/35924495
1. Rev Med Liege. 2022 Jul;77(7-8):421-425. [A catamenial pneumothorax, an unknown entity]. [Article in French; Abstract available in French from the publisher] Nguyen LD(1), Guérisse F(2). Author information: (1)Service des Urgences, CHU Tivoli, La Louvière, Belgique. (2)Service des Urgences, CHU de Charleroi, Belgique. A 31-year-old patient is admitted to the emergency room because of an acute right thoracic pain associated with a dyspnea. The patient reports the stopping of Decapeptyl®, a treatment taken in regards to an endometriosis, but interrupted to get pregnant. An x-ray highlights a pneumothorax of 15 mm at the right apical level. It is a second episode for this patient. Catamenial pneumothorax is one of the most frequent manifestation in terms of a thoracic endometriosis syndrome (TES). It concerns a rare pathology, unrecognized and underdiagnosed. The diagnosis should be invoked on all patients having the childbearing age who are presenting themselves at the emergencies with a right thoracic pain. The medical care is multidisciplinary, the association of a hormonal therapy and then a surgical treatment being the best therapeutical approach. This case report describes the recurrence of a catamenial pneumotorax induced by the stopping of the endometriosis treatment and reviews the physiopathology, the diagnosis and its multidisciplinary management. Publisher: Une patiente de 31 ans est admise aux urgences pour douleur thoracique droite apparue brutalement et associée à une dyspnée. La patiente rapporte l’arrêt du Decapeptyl®, traitement pris dans le cadre d’une endométriose, mais interrompu pour un désir de grossesse. Une radiographie mettra en évidence un pneumothorax de 15 mm au niveau apical droit. Il s’agit du deuxième épisode chez cette patiente. Le pneumothorax cataménial (PC) est l’une des manifestations les plus fréquentes dans le cadre d’un syndrome d’endométriose thoracique (SET). Il s’agit d’une pathologie rare, méconnue et sous-diagnostiquée. Il est à évoquer chez toutes patientes en âge de procréer se présentant aux urgences avec une douleur thoracique droite. La prise en charge est multidisciplinaire, l’association d’un traitement hormonal, puis chirurgical, semble être la meilleure approche thérapeutique. Cet article rapporte la récidive d’un PC, récidive induite par l’arrêt du traitement de l’endométriose, et revoit la physiopathologie, le diagnostic et la prise en charge de celui-ci. PMID: 35924495 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34375738
1. J Minim Invasive Gynecol. 2022 Jan;29(1):41-55. doi: 10.1016/j.jmig.2021.08.005. Epub 2021 Aug 8. Thoracic Endometriosis: A Review Comparing 480 Patients Based on Catamenial and Noncatamenial Symptoms. Topbas Selcuki NF(1), Yilmaz S(2), Kaya C(3), Usta T(4), Kale A(5), Oral E(6). Author information: (1)Department of Obstetrics and Gynecology, University of Health Sciences Turkey, Istanbul Sisli Hamidiye Etfal Training and Research Hospital (Dr. Topbas Selcuki). (2)Department of Obstetrics and Gynecology, Acibadem Altunizade Hospital (Dr. Yilmaz). (3)Department of Obstetrics and Gynecology, Acibadem Mehmet Ali Aydinlar University, Acibadem Bakirkoy Hospital (Dr. Kaya). (4)Department of Obstetrics and Gynecology, Acibadem Mehmet Ali Aydinlar University, Acibadem Altunizade Hospital (Dr. Usta). Electronic address: [email protected]. (5)Department of Obstetrics and Gynecology, University of Health Sciences Turkey, Istanbul Kartal Dr. Lutfi Kirdar City Hospital (Dr. Kale). (6)Department of Obstetrics and Gynecology, Bezmialem Vakif University (Dr. Oral), Istanbul, Turkey. OBJECTIVE: This review aimed to categorize thoracic endometriosis syndrome (TES) according to whether the presenting symptoms were catamenial and to evaluate whether such a categorization enables a better management strategy. DATA SOURCES: An electronic search was conducted using the PubMed/Medline database. METHODS OF STUDY SELECTION: The following keywords were used in combination with the Boolean operators AND OR: "thoracic endometriosis syndrome," "thoracic endometriosis," "diaphragm endometriosis," and "catamenial pneumothorax." TABULATION, INTEGRATION, AND RESULTS: The initial search yielded 445 articles. Articles in non-English languages, those whose full texts were unavailable, and those that did not present the symptomatology clearly were further excluded. After these exclusions, the review included 240 articles and 480 patients: 61 patients in the noncatamenial group and 419 patients in the catamenial group. The groups differed significantly in presenting symptoms, surgical treatment techniques, and observed localization of endometriotic loci (p <.05). CONCLUSION: This review points out the significant differences between patients with TES with catamenial and noncatamenial symptoms. Such categorization and awareness by clinicians of these differences among patients with TES can be helpful in designing a management strategy. When constructing management guidelines, these differences between patients with catamenial and noncatamenial symptoms should be taken into consideration. Copyright © 2021. Published by Elsevier Inc. DOI: 10.1016/j.jmig.2021.08.005 PMID: 34375738 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27539547
1. Angew Chem Int Ed Engl. 2016 Oct 17;55(43):13408-13421. doi: 10.1002/anie.201601091. Epub 2016 Aug 19. Targeted Covalent Inhibitors for Drug Design. Baillie TA(1). Author information: (1)Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Box 357610, Seattle, WA, 98195-7610, USA. [email protected]. In contrast to the traditional mechanism of drug action that relies on the reversible, noncovalent interaction of a ligand with its biological target, a targeted covalent inhibitor (TCI) is designed such that the initial, reversible association is followed by the formation of a covalent bond between an electrophile on the ligand and a nucleophilic center in the protein. Although this approach offers a variety of potential benefits (high potency and extended duration of action), concerns over the possible toxicological consequences of protein haptenization have hindered the development of the TCI concept. Recently, approaches to mitigate the risk of serious adverse reactions to this new class of agent have emerged, thus stimulating interest in the field and leading to authorization of the first cadre of TCIs to be marketed. The covalent inhibitor approach is rapidly gaining acceptance as a valuable tool in drug discovery, and is poised to make a major impact on the design of enzyme inhibitors and receptor modulators. © 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim. DOI: 10.1002/anie.201601091 PMID: 27539547 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23928507
1. Phytomedicine. 2013 Oct 15;20(13):1186-9. doi: 10.1016/j.phymed.2013.07.004. Epub 2013 Aug 6. Is green tea a potential trigger for autoimmune hepatitis? Gallo E(1), Maggini V, Berardi M, Pugi A, Notaro R, Talini G, Vannozzi G, Bagnoli S, Forte P, Mugelli A, Annese V, Firenzuoli F, Vannacci A. Author information: (1)University of Florence, Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), Center of Molecular Medicine (CIMMBA), Florence, Italy. A case of autoimmune liver hepatitis is reported: the onset was triggered by consumption of green tea infusion in a patient taking oral contraceptives and irbesartan. We hypothesize that our patient, carrying genetic variant of hepatic metabolism making her particularly susceptible to oxidative stress, developed an abnormal response to a mild toxic insult, afforded by a combination of agents (oral contraceptives+irbesartan+green tea) that normally would not be able to cause damage. Her particular hepatic metabolism further increased the drugs' concentration, favoring the haptenization of liver proteins, eventually leading to the development of an autoimmune hepatitis. Copyright © 2013 Elsevier GmbH. All rights reserved. DOI: 10.1016/j.phymed.2013.07.004 PMID: 23928507 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35037874
1. Medicina (B Aires). 2022;82(1):147-150. [Catamenial pneumothorax]. [Article in Spanish; Abstract available in Spanish from the publisher] Toffolo Pasquini M(1), Auvieux R(2), Tchercansky A(2), Buero A(2), Chimondeguy D(2), Mendez J(3). Author information: (1)Servicio de Cirugía General, Hospital Británico de Buenos Aires, Argentina. E-mail: [email protected]. (2)Servicio de Cirugía Torácica, Hospital Británico de Buenos Aires, Argentina. (3)Servicio de Anatomía Patológica, Hospital Británico de Buenos Aires, Argentina. Thoracic endometriosis syndrome (TES) is a rare disorder characterized by the presence of ectopic endometrial tissue in the chest cavity. The typical clinical manifestation is a spontaneous pneumothorax, which usually presents with chest pain, dyspnea, and/or cough. The diagnosis requires a high level of clinical suspicion and a complete gynecological history. Imaging studies can help with the diagnosis, although the gold standard is video-assisted thoracoscopic surgery (VATS). Surgical treatment in combination with at least 6 months of hormonal medical treatment has been shown to improve the prognosis and reduce the recurrence of this entity. We present the case of a 40-year-old patient with a history of pelvic endometriosis and multiple episodes of pneumothorax, who consulted at our institution for a new episode of spontaneous pneumothorax. A VATS was performed where nodules in the parietal pleura and diaphragmatic orifices were identified. In the postoperative period, she continued with hormonal treatment. At 6 months of follow-up, she reported improvement in pain and did not present new episodes of pneumothorax. Publisher: El síndrome de endometriosis torácica (TES) es un trastorno poco común caracterizado por la presencia de tejido endometrial ectópico en la cavidad torácica. La manifestación clínica típica es un neumotórax espontáneo, que generalmente se presenta con dolor torácico, disnea y/o tos. El diagnóstico requiere un alto nivel de sospecha clínica junto con una historia ginecológica completa. Los estudios de imágenes pueden ayudar con el diagnóstico, pero el gold standard es la cirugía toracoscópica videoasistida (VATS). Se ha demostrado que el tratamiento quirúrgico en combinación con al menos 6 meses de tratamiento médico hormonal mejora el pronóstico y reduce la recurrencia de esta entidad. Presentamos el caso de una paciente de 40 años con antecedentes de endometriosis pélvica y múltiples episodios de neumotórax, que consultó en nuestra institución por un nuevo episodio de neumotórax espontáneo. Se realizó una VATS donde se identificaron nódulos en la pleura parietal y orificios diafragmáticos. En el postoperatorio continuó con tratamiento hormonal. A los 6 meses de seguimiento refirió mejoría del dolor y no presentó nuevos episodios de neumotórax. PMID: 35037874 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22613852
1. Chem Immunol Allergy. 2012;97:32-46. doi: 10.1159/000335614. Epub 2012 May 3. Etiology and pathogenesis of adverse drug reactions. Hausmann O(1), Schnyder B, Pichler WJ. Author information: (1)Department of Rheumatology and Clinical Immunology/Allergology, Inselspital, University of Bern, Bern, Switzerland. In clinical routine, adverse drug reactions (ADR) are common, and they should be included in the differential diagnosis in all patients undergoing drug treatment. Only part of those ADR are immune-mediated hypersensitivity reactions and thus true drug allergies. Far more common are non-immune-mediated ADR, e.g. due to the pharmacological properties of the drug or to the individual predisposition of the patient (enzymopathies, cytokine dysbalance, mast cell hyperreactivity). In true drug allergiesT cell- and immunoglobulin E (lgE)-mediated reactions dominate the clinical presentation. T cell-mediated ADR usually have a delayed appearance and include skin eruptions in most cases. Nevertheless, it should not be forgotten that they may involve systemic T cell activation and thus take a severe, sometimes lethal turn. Clinical danger signs are involvement of mucosal surfaces, blistering within the exanthematous skin areas and systemic symptoms, e.g. fever or malaise. Drug presentation via antigen-presenting cells to T cells can either involve the classical pathway of haptenization of endogenous proteins or be directly mediated via noncovalent binding to immune receptors (MHC molecules or T cell receptors), the so-called p-i concept. Flare-up reactions during the acute phase of T cell-mediated ADR should not be mistaken for true drug allergies, as they only occur in the setting of a highly activated T cell pool. IgE-mediated ADR are less frequent and involve mast cells and/or basophils as peripheral effector cells. Recent data suggest that certain patients with drug allergy have a preexistent sensitization although they have never been exposed to the culprit drug, probably due to cross-reactivity. Thus, allergic drug reactions on first encounter are possible. In general, the extent of cross-reactivity is higher in IgE-compared to T cell-mediated ADR. Based on a specific ethnic background and only for severe T cell-mediated ADR to certain drugs, a strong HLA association has been established recently. Copyright © 2012 S. Karger AG, Basel. DOI: 10.1159/000335614 PMID: 22613852 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35118122
1. Skin Appendage Disord. 2022 Jan;8(1):1-7. doi: 10.1159/000518191. Epub 2021 Sep 1. Trichotillomania: What Do We Know So Far? Melo DF(1), Lima CDS(2)(3), Piraccini BM(4)(5), Tosti A(6). Author information: (1)Dermatology Department, University of State of Rio de Janeiro (UERJ), Rio de Janeiro, Brazil. (2)Dermatology Department, University of State of Pará (UEPA), Belém, Brazil. (3)Dermatology Department, University Center of Pará (CESUPA), Belém, Brazil. (4)Department of Experimental, Diagnostic and Specialty Medicine (DIMES) Alma Mater Studiorum University of Bologna, Bologna, Italy. (5)IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy. (6)Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, Florida, USA. Trichotillomania is defined as an obsessive-compulsive or related disorder in which patients recurrently pull out hair from any region of their body. The disease affects mainly female patients, who often deny the habit, and it usually presents with a bizarre pattern nonscarring patchy alopecia with short hair and a negative pull test. Trichoscopy can reveal the abnormalities resulting from the stretching and fracture of hair shafts, and biopsy can be necessary if the patient or parents have difficulties in accepting the self-inflicted nature of a trichotillomania diagnosis. Trichotillomania requires a comprehensive treatment plan and interdisciplinary approach. Physicians should always have a nonjudgmental, empathic, and inviting attitude toward the patient. Behavioral therapy has been used with success in the treatment of trichotillomania, but not all patients are willing or able to comply with this treatment strategy. Pharmacotherapy can be necessary, especially in adolescents and adult patients. Options include tricyclic antidepressants, selective serotonin reuptake inhibitors, and glutamate-modulating agents. Glutamate-modulating agents such as N-acetylcysteine are a good first-line option due to significant benefits and low risk of side effects. Physicians must emphasize that the role of psychiatry-dermatology liaison is extremely necessary with concurrent support services for the patient and parents, in case of pediatric patients. In pediatric cases, parents should be advised and thoroughly educated that negative feedback and punishment for hair pulling are not going to produce positive results. Social support is a significant pillar to successful habit reversal training; therefore, physicians must convey the importance of familial support to achieving remission. This is a review article that aims to discuss the literature on trichotillomania, addressing etiology, historical aspects, clinical and trichoscopic features, main variants, differential diagnosis, diagnostic clues, and psychological and pharmacological management. Copyright © 2021 by S. Karger AG, Basel. DOI: 10.1159/000518191 PMCID: PMC8787581 PMID: 35118122 Conflict of interest statement: Dr. Tosti reports being a consultant − DS Laboratories, Monat Global, Almirall, Tirthy Madison, Eli Lilly, Bristol Myers Squibb, and P&G.
http://www.ncbi.nlm.nih.gov/pubmed/30560020
1. Intractable Rare Dis Res. 2018 Nov;7(4):271-274. doi: 10.5582/irdr.2018.01094. Prune belly syndrome: Approaches to its diagnosis and management. Achour R(1), Bennour W(2), Ksibi I(2), Cheour M(2), Hamila T(1), Hmid RB(1), Kacem S(2). Author information: (1)Emergency Department of Gynecology and Obstetrics, Maternity and Neonatology Center, Faculty of Medicine, Tunis-El Manar University, Tunis, Tunisia. (2)Neonatology Department, Maternity and Neonatology Center, Faculty of Medicine, Tunis-El Manar University, Tunis, Tunisia. Prune Belly syndrome (PBS) or Eagle-Barrett syndrome is an anatomo-radiological syndrome consisting of a complex and rare malformation characterized by the following triad of symptoms: deficiency of the abdominal muscles, malformations of the urinary tract, and bilateral cryptorchidism. The exact etiology is unknown, though PBS predominantly occurs in males. The clinical manifestations can vary widely, from stillbirth to renal and major respiratory dysplasia to almost normal children. The current study included a total of 3 patients. The findings included clinical characteristics, diagnostics, therapy, and clinical outcomes. All patients were diagnosed with congenital aplasia of the abdominal wall and a variety of urogenital malformations. Cryptorchidism and a mega-bladder were observed in 2 patients and distinctive renal malformations, such as renal dysplasia, were observed in 1 patient. Treatment varies but usually includes surgical management of symptoms. One patient required urgent urinary surgery; a vesicotomy was urgently performed due to anuria. These aspects explain the great diversity of opinions on the approach to this syndrome, but the severity of renal dysplasia is the main prognostic factor. Two newborns died a few days later due to severe renal failure. Despite these concerns, many patients with PBS report being in physical and mental health and having a good quality of life. DOI: 10.5582/irdr.2018.01094 PMCID: PMC6290839 PMID: 30560020
http://www.ncbi.nlm.nih.gov/pubmed/35526214
1. Spec Care Dentist. 2023 Jan;43(1):67-72. doi: 10.1111/scd.12728. Epub 2022 May 8. Dental treatment of patients with prune belly syndrome. Quilici G(1), Tolarova MM(1), Quilici M(2), Quilici DL(3). Author information: (1)University of the Pacific Arthur A. Dugoni School of Dentistry, San Francisco, California, USA. (2)Oakland, California, USA. (3)Department of Veterans Affairs, California, McClellan, USA. BACKGROUND: Prune belly syndrome (PBS), also known as Eagle-Barrett syndrome (EGBRS), is a rare congenital disease characterized by deficiency or absence of abdominal wall muscles, urological abnormalities, and bilateral cryptorchidism. TYPES OF STUDIES REVIEWED: A review of literature was done using four search engines (PubMed, Google Scholar, Scopus, Science Direct) and keywords (individually and in combinations): prune belly syndrome, PBS, Eagle-Barrett syndrome, dental manifestation, clinical manifestation, and psychological aspects. The search was run with no language restrictions and covered the 1965-2021 time period. RESULTS: The search yielded a large number of articles. The vast majority were dealing with a variety of treatments. PBS is a multisystem disease with a variable spectrum ranging from mild cases to infant mortality. Comorbidities of PBS (63% gastrointestinal, 65% orthopedic, and 49% cardiopulmonary) present challenges for treatment. PBS affects quality of life of patients and caregivers. We selected and summarized published information that is relevant to oral health and dental care. CONCLUSIONS AND PRACTICAL IMPLICATIONS: Providing information to dental practitioners will improve their understanding of PBS. It will help them to better treat patients with PBS and it will encourage more dental providers to welcome patients with PBS into their dental clinics. © 2022 The Authors. Special Care in Dentistry Published by Special Care Dentistry Association and Wiley Periodicals LLC. DOI: 10.1111/scd.12728 PMCID: PMC10083899 PMID: 35526214 [Indexed for MEDLINE] Conflict of interest statement: The authors declares no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35429431
1. Can J Urol. 2022 Apr;29(2):11116-11118. Robotic treatment of ureteropelvic junction obstruction in Eagle-Barrett Syndrome. Faber LS(1), Riley JM(2). Author information: (1)Department of Surgery (Division of Urology), University of New Mexico, Albuquerque, New Mexico, USA. (2)Department of Urology at University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. Eagle-Barrett Syndrome (EBS) is a rare congenital condition characterized by the triad of absent or defective abdominal wall muscles, urinary tract abnormalities, and bilateral cryptorchidism. Ureteropelvic junction obstruction (UPJO) is seldom reported in these patients, despite it being a common cause of childhood obstructive uropathy. We present the case of a patient with EBS who was subsequently identified as having symptomatic UPJO that was successfully treated with robotic pyeloplasty. PMID: 35429431 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32774275
1. Case Rep Oncol. 2020 Jul 2;13(2):774-782. doi: 10.1159/000507921. eCollection 2020 May-Aug. Mantle Cell Lymphoma Presenting as a Subcutaneous Mass of the Right Leg. Fajardo DA(1), France J(2), Targonska BI(3)(4)(5), Kahlon HB(6), Coppes MJ(4)(5). Author information: (1)University of Nevada Reno School of Medicine, Reno, Nevada, USA. (2)Sierra Pathology Associates, Reno, Nevada, USA. (3)Reno Radiological Associates, Reno, Nevada, USA. (4)Department of Pediatrics, University of Nevada Reno School of Medicine, Reno, Nevada, USA. (5)Renown Children's Hospital, Reno, Nevada, USA. (6)Family Medicine, Renown Medical Group, Reno, Nevada, USA. Mantle cell lymphoma (MCL) is a relatively rare B-cell non-Hodgkin lymphoma, typically presenting with extensive lymphadenopathy, bone marrow involvement, and splenomegaly. Extranodal sites can also be involved. We discuss a 73-year-old man whose MCL presented with a 6-month history of a subdermal mass of the right upper thigh and no systemic symptoms. Copyright © 2020 by S. Karger AG, Basel. DOI: 10.1159/000507921 PMCID: PMC7383204 PMID: 32774275 Conflict of interest statement: The authors declare to have no conflict of interest. Financial support (all based on employment) is provided under “Funding Sources.”