originals - Revista Nefrologia
Transcription
originals - Revista Nefrologia
Included in ISI-WOK, MEDLINE, EMBASE, IME, IBECS, SCIELO V o l u m e 3 2 - N u m b e r 1 - 2 01 2 WORLD KIDNEY DAY 2012: THE GLOBAL ROLE OF KIDNEY TRANSPLANTATION EARLY DIAGNOSIS OF ATHEROSCLEROSIS BY CAROTID ULTRASOUND IN CHRONIC KIDNEY DISEASE LONGITUDINAL OBSERVATIONAL STUDIES AND CAUSALITY RENAL SUPPORTIVE CARE AND PALLIATIVE CARE HAEMODIALYSIS USING HIGH CUT-OFF DIALYSERS FOR TREATING ACUTE RENAL FAILURE IN MULTIPLE MYELOMA EARLY BIOMARKERS OF ACUTE KIDNEY FAILURE AFTER HEART ANGIOGRAPHY OR HEART SURGERY SURVIVAL ON HAEMODIALYSIS AND POVERTY LEVEL DDAVP INTRANASAL SOLUTION FOR TREATING HYPOTENSION DURING HAEMODIALYSIS SESSIONS URIC ACID AS A MARKER OF ALL-CAUSE MORTALITY IN THE ELDERLY Sociedad Española de Nefrología Official Publication of the Spanish Society of Nephrology Full version in English and Spanish at www.revistanefrologia.com Nefrología Journal Editor-in-Chief: Carlos Quereda Rodríguez-Navarro Executive editor: Roberto Alcázar Arroyo Deputy editors: Andrés Purroy Unanua, Mariano L. Rodríguez Portillo, Ángel Luis Martín de Francisco, Fernando García López, Víctor Lorenzo Sellares Honorary editors: Luis Hernando Avendaño, David Kerr, Rafael Matesanz Acedos SUBJECT EDITORS (editors of thematic areas) Experimental Nephrology A. Ortiz* J. Egido de los Ríos S. Lamas J.M. López Novoa D. Rodríguez Puyol J.M. Cruzado Clinical Nephrology M. Praga* J. Ara J. Ballarín G. Fernández Juárez F. Rivera A. Segarra Diabetic Nephropathy F. de Álvaro* J.L. Górriz A. Martínez Castelao J.F. Navarro J.A. Sánchez Tornero R. Romero Hereditary Nephropathies R. Torra* X. Lens J.C. Rodríguez Pérez M. Navarro E. Coto V. García Nieto Chronic Kidney Disease A.L. Martín de Francisco* A. Otero E. González Parra I. Martínez J. Portolés Pérez CRF-Ca/P Metabolism E. Fernández* J. Cannata Andía R. Pérez García M. Rodríguez J.V. Torregrosa Arterial Hypertension R. Marín* J.M. Alcázar L. Orte R. Santamaría A. Rodríguez Jornet Nephropathy and Cardiovascular Risk J. Díez* A. Cases J. Luño Quality in Nephrology F. Álvarez-Ude* M.D. Arenas E. Parra Moncasi P. Rebollo F. Ortega Acute Renal Failure F. Liaño* F.J. Gainza J. Lavilla E. Poch Peritoneal Dialysis R. Selgas* M. Pérez Fontán C. Remón M.E. Rivera Gorrin G. del Peso Haemodialysis A. Martín Malo* P. Aljama F. Maduell J.A. Herrero J.M. López Gómez J.L. Teruel Renal Transplantation J. Pascual* M. Arias J.M. Campistol J.M. Grinyó M.A. Gentil A. Torres Paediatric Nephrology I. Zamora* N. Gallego A.M. Sánchez Moreno R. Vilalta Nephropathology J. Blanco* I.M. García E. Vázquez Martul A. Barat Cascante Evidence-Based Nephrology Vicente Barrio* (Director of Supplements), Fernando García López (Methodology assessment), Editors: María Auxiliadora Bajo, José Conde, Joan M. Díaz, Mar Espino, Domingo Hernández, Ana Fernández, Milagros Fernández, Fabián Ortiz, Ana Tato. Continued Training (journal NefroPlus) Andrés Purroy*, R. Marín, J.M. Tabernero, F. Rivera, A. Martín Malo. * Coordinators of thematic area EDITORIAL BOARD A. Alonso J. Arrieta F.J. Borrego D. del Castillo P. Gallar M.A. Frutos D. Jarillo A. Mazuecos A. Oliet L. Pallardo J.J. Plaza D. Sánchez Guisande J. Teixidó J. Alsina P. Barceló J. Bustamente A. Darnell P. García Cosmes M.T. González L. Jiménez del Cerro J. Lloveras B. Miranda J. Olivares V. Pérez Bañasco L. Revert A. Serra F.A. Valdés F. Anaya G. Barril F. Caravaca C. de Felipe S. García de Vinuesa A. Gonzalo R. Lauzurica J.F. Macías E. Martín Escobar J.M. Morales R. Peces J.M. Tabernero A. Vallo G. de Arriba C. Bernis A. Barrientos A. Caralps P. Errasti F. García Martín M. González Molina I. Lampreabe B. Maceira J. Mora J. Ortuño S. Pérez García J.L. Rodicio L. Sánchez Sicilia A. Vigil J. Aranzábal E. Fernández Giráldez F.J. Gómez Campderá P. Gómez Fernández E. Huarte E. López de Novales R. Marcén J. Montenegro A. Palma L. Piera J. Rodríguez Soriano A. Tejedor INTERNATIONAL COMMITEE BOARD E. Burdmann (Brazil) B. Canaud (France) J. Chapman (Australia) R. Coppo (Italy) R. Correa-Rotter (Mexico) F. Cosío (USA) G. Eknoyan (USA) A. Felsenfeld (USA) J.M. Fernández Cean (Uruguay) J. Frazao (Portugal) M. Ketteler (Germany) Levin, Adeera (Canada) Li, Philip K.T. (Hong Kong, China) L. Macdougall (United Kingdon) P. Massari (Argentina) S. Mezzano (Chile) B. Rodríguez Iturbe (Venezuela) C. Ronco (Italy) J. Silver (Israel) P. Stevinkel (Sweden) A. Wiecek (Poland) C. Zoccali (Italy) COUNCIL OF THE SPANISH SOCIETY OF NEPHROLOGY SUBSCRIPTIONS, ADVERTISING AND PUBLISHING Information and subscriptions: S.E.N. Secretary: [email protected] Tel: 902 929 210 Queries regarding of manuscripts: [email protected] Avda. dels Vents 9-13, Esc. B, 2.º 1.ª Edificio Blurbis 08917 Badalona Tel. 902 02 09 07 - Fax. 93 395 09 95 Rambla del Celler 117-119, 08190 Sant Cugat del Vallès. Barcelona Tel. 93 589 62 64 - Fax. 93 589 50 77 Distribuido por: E.U.R.O.M.E.D.I.C.E., Ediciones Médicas, S.L. © Copyright 2012. Grupo Editorial Nefrología. All rights reserved. • ISSN: 2013-2514 © Sociedad Española de Nefrología 2012. All international rights reserved. No part of this publication may be reproduced, stored in a retrieval system or transmitted in any form or by any means, mechanical, electronic, photocopying, recording or otherwise, without the prior written permission of the publisher. Nefrología is distributed exclusively among medical professionals. President: Dr. Alberto Martínez Castelao Vice-president: Dr. Isabel Martínez Secretary: Dr. José Luis Górriz Director of Nefrología Publishing Group: Dr. Carlos Quereda Rodríguez Chairperson of the Dialysis and Transplantation Registry: Dr. Ramón Saracho Treasurer: Dr. María Dolores del Pino Chairpersons of Education and Research: Ordinary members: Dr. Gema Fernández Fresnedo Dr. Juan Francisco Navarro Dr. Elvira Fernández Giráldez Dr. Julio Pascual Dr. José María Portolés Web Page of Nefrología: E-mail Editor-in-Chief: Dr. Josep Maria Cruzado Chairperson for selection of the SEN Congress presentations: Dr. Rosa Sánchez Hernández Links: www.revistanefrologia.com [email protected] [email protected] contents Included in ISI-WOK, MEDLINE, EMBASE, IME, IBECS, SCIELO Included in ISI-WOK, MEDLINE, EMBASE, IME, IBECS, SCIELO Volume 32 - Number 1 - 2012 V o l u m e 3 2 - N u m b e r 1 - 2 01 2 EDITORIALS 1 • The global role of kidney transplantation Guillermo García-García, Paul Harden, Jeremy Chapman, for the World Kidney Day Steering Committee 2012 WORLD KIDNEY DAY 2012: THE GLOBAL ROLE OF KIDNEY TRANSPLANTATION EARLY DIAGNOSIS OF ATHEROSCLEROSIS BY CAROTID ULTRASOUND IN CHRONIC KIDNEY DISEASE LONGITUDINAL OBSERVATIONAL STUDIES AND CAUSALITY RENAL SUPPORTIVE CARE AND PALLIATIVE CARE 7 • Carotid ultrasound for the early diagnosis of atherosclerosis in chronic kidney disease HAEMODIALYSIS USING HIGH CUT-OFF DIALYSERS FOR TREATING ACUTE RENAL FAILURE IN MULTIPLE MYELOMA EARLY BIOMARKERS OF ACUTE KIDNEY FAILURE AFTER HEART ANGIOGRAPHY OR HEART SURGERY SURVIVAL ON HAEMODIALYSIS AND POVERTY LEVEL DDAVP INTRANASAL SOLUTION FOR TREATING HYPOTENSION DURING HAEMODIALYSIS SESSIONS Àngels Betriu-Bars, Elvira Fernández-Giráldez URIC ACID AS A MARKER OF ALL-CAUSE MORTALITY IN THE ELDERLY Sociedad Española de Nefrología EDITORIAL COMMENTS 12 • Longitudinal observational studies and causality Alfonso Muriel, Domingo Hernández-Marrero, Víctor Abraira 15 • The value of serum free light chain assay in patients with monoclonal gammopathies and renal failure M. Luisa Campos, Nuno M. Barbosa de Carvalho, Guillermo Martín-Reyes Official Publication of the Spanish Society of Nephrology Full version in English and Spanish at www.revistanefrologia.com IMAGES ON COVER Granulomatous interstitial nephritis due to sarcoidosis. C. Galeano and A. Saiz. Nephrology and Anatomical Pathology Departments Ramón y Cajal University Hospital. Madrid, Spain. SHORT REVIEW 20 • Renal supportive care and renal palliative care: revision and proposal in kidney replacement therapy Juan P. Leiva-Santos, Rosa Sánchez-Hernández, Helena García-Llana, M. José Fernández-Reyes, Manuel Heras-Benito, Álvaro Molina-Ordas, Astrid Rodríguez, Fernando Álvarez-Ude SPECIAL ARTICLE 28 • Start-up of a clinical sample processing, storage and management platform: organisation and development of the REDinREN Biobank Laura Calleros, María A. Cortés, Alicia Luengo, Inés Mora, Brenda Guijarro, Paloma Martín, Alberto Ortiz-Arduán, Rafael Selgas, Diego Rodríguez-Puyol, Manuel Rodríguez-Puyol ORIGINALS 35 • Haemodialysis using high cut-off dialysers for treating acute renal failure in multiple myeloma Guillermo Martín-Reyes, Remedios Toledo-Rojas, Álvaro Torres-Rueda, Eugenia Sola-Moyano, Lourdes Blanca-Martos, Laura Fuentes-Sánchez, M. Dolores Martínez-Esteban, M. José Díez-de los Ríos, Alicia Bailén-García, Miguel González-Molina, Isabel García-González 44 • Early biomarkers of acute kidney failure after heart angiography or heart surgery in patients with acute coronary syndrome or acute heart failure Isidro Torregrosa, Carmina Montoliu, Amparo Urios, Nisrin Elmlili, Isabel Juan, M. Jesús Puchades, Miguel Á. Solís, Rafael Sanjuán, M. Luisa Blasco, Carmen Ramos, Patricia Tomás, José Ribes, Arturo Carratalá, Alfonso Miguel 53 • The calculation of baseline serum creatinine overestimates the diagnosis of acute kidney injury in patients undergoing cardiac surgery A.M. Candela-Toha, M. Recio-Vázquez, A. Delgado-Montero, J.M. del Rey, A. Muriel, F. Liaño, T. Tenorio 59 • Early detection of chronic kidney disease: Collaboration of Belgrade nephrologists and primary care physicians Ljubica Djukanović, Visnja Lezaić, Nada Dimković, Gordana Perunicić Peković, Danica Bukvić, Sanja Bajcetić, Jelena Pavlović, Ana Bontić, Nadezda Zec, Danijela Momcilović, Marina Stojanović Stanojević 67 • Serum uric acid as a marker of all-cause mortality in an elderly patients cohort Manuel Heras, María J. Fernández-Reyes, Rosa Sánchez, Álvaro Molina, Astrid Rodríguez, Fernando Álvarez-Ude 73 • Bone mineral density and bone metabolism in hemodialysis patients. Correlation with PTH, 25OHD3 and leptin A. Polymeris, K. Doumouchtsis, E. Grapsa 79 • The lack of income is associated with reduced survival in chronic haemodialysis Sergio Marinovich, Carlos Lavorato, Guillermo Rosa-Diez, Liliana Bisigniano, Víctor Fernández, Daniela Hansen-Krogh contents Included in ISI-WOK, MEDLINE, EMBASE, IME, IBECS, SCIELO Volume 89 32 - Number 1 - 2012 • Effect of intranasal DDAVP in prevention of hypotension during hemodialysis Seyed S. Beladi-Mousavi, Marzieh Beladi-Mousavi, Fatemeh Hayati, Mehdi Talebzadeh 94 • Echocardiographic impact of hydration status in dialysis patients Isabel Juan-García, María J. Puchades, Rafael Sanjuán, Isidro Torregrosa, Miguel Á. Solís, Miguel González, Marisa Blasco, Antonio Martínez, Alfonso Miguel SHORT ORIGINALS 103 • Vascular accesses in haemodialysis: a challenge to be met Gloria Antón-Pérez, Patricia Pérez-Borges, Francisco Alonso-Almán, Nicanor Vega-Díaz 108 • Changes in body composition parameters in patients on haemodialysis and peritoneal dialysis M. Cristina Di-Gioia, Paloma Gallar, Isabel Rodríguez, Nuria Laso, Ramiro Callejas, Olimpia Ortega, Juan C. Herrero, Ana Vigil LETTERS TO THE EDITOR A) Comments on published articles 114 • The role of interleukin 6 in the pathogenesis of hyponatremia associated with Guillain-Barré syndrome Se Jin Park, Ki Soo Pai, Ji Hong Kim, Jae II Shin 114 • Aciclovir and valaciclovir neurotoxicity in patients with renal failure Gloria Ruiz-Roso, Antonio Gomis, Milagros Fernández-Lucas, Martha Díaz-Domínguez, José L. Teruel-Briones, Carlos Quereda 115 • Estimating glomerular filtration rate in order to adjust drug doses: confusion abounds Javier Peral-Aguirregoitia, Unax Lertxundi-Etxebarria, Ramon Saracho-Rotaeche, Sira Iturrizaga-Correcher, M. José Martínez-Bengoechea B) Brief papers on research and clinical experiments 118 • Is peripheral and/or catheter blood necessary for performing haemoculture in haemodialysis patients whose central venous catheter presents bacteraemia? Juan F. Betancor-Jiménez, Francisco Alonso-Almán, Yanet Parodis-López, Beatriz Quintana-Viñau, Sonia González-Martínez, Cristina García-Laverick, Patricia Pérez-Borges, José C. Rodríguez-Pérez 120 • Serial ultrasound of the vascular access Juan A. Martín-Navarro, M. José Gutiérrez-Sánchez, Vladimir Petkov-Stoyanov 122 • Economic impact of estimating renal function in patients with systemic lupus erythematosus Marco U. Martínez-Martínez, Carlos Abud-Mendoza 123 • Chronic kidney disease in the elderly: the impact of patients’ sex Manuel Heras, Pedro García-Cosmes, M. José Fernández-Reyes, M. Teresa Guerrero, Rosa Sánchez C) Brief case reports 124 • Adrenal myelolipoma associated with primary hyperaldosteronism Vanesa Camarero-Temiño, Verónica Mercado-Ortiz, Badawi Hijazi-Prieto, Pedro Abaigar-Luquin 125 • Reactive haemophagocytic syndrome associated with parvovirus B19 in a kidney-pancreas transplant patient Maico Tavera, Jorgelina Petroni, Luis León, Elena Minue, Domingo Casadei 127 • Emphysematous cystitis resolved with medical treatment. A case report and literature review Héctor Parra-Riffo, Juan Lemus-Peñaloza, Paula Maira 127 • Rhabdomyolysis and acute renal failure following hard physical activity in a patient treated with rosuvastatin Diana Martínez-López, Ricardo Enríquez, Ana E. Sirvent, M. Dolores Redondo-Pachón, Isabel Millán, Francisco Amorós 129 • Intestinal pseudo-obstruction secondary to persistent constipation due to lanthanum carbonate Vanesa Camarero-Temiño, Verónica Mercado-Valdivia, Badawi Hijazi-Prieto, Pedro Abaigar-Luquin 130 • Acute renal failure due to gabapentin. A case report and literature Eduardo Torregrosa-de Juan, Pau Olagüe-Díaz, Pilar Royo-Maicas, Enrique Fernández-Nájera, Rafael García-Maset 131 • Haemorrhagic fever with renal failure syndrome: a case report Rodrigo Avellaneda-Campos http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society editorials The global role of kidney transplantation Guillermo García-García1, Paul Harden2, Jeremy Chapman3, for the World Kidney Day Steering Committee 2012* *World Kidney Day, International Society of Nephrology. Brussels (Belgium) 1 Nephrology Department. Hospital Civil de Guadalajara, University of Guadalajara Health Sciences Center (CUCS). Guadalajara, Jalisco (Mexico) 2 Oxford Kidney Unit and Oxford Transplant Centre. Churchill Hospital. Oxford (United Kingdom) 3 Centre for Transplant and Renal Research. Westmead Millennium Institute, Sydney University, Westmead Hospital. Sidney (Australia) Nefrologia 2012;32(1):1-6 doi:10.3265/Nefrologia.pre2012.Jan.11333 INTRODUCTION K idney transplantation is acknowledged as a major advance of modern medicine which provides high-quality life years to patients with irreversible kidney failure (end-stage renal disease, ESRD) worldwide. What was an experimental, risky and very limited treatment option fifty years ago, is now routine clinical practice in more than 80 countries. What was once limited to a few individuals in a small number of leading academic centers in high income economies, is now transforming lives as a routine procedure in most high- and middle-income countries – but can do much more. The largest numbers of transplants are performed in the USA, China, Brazil and India, while the greatest population access to transplantation is in Austria, USA, Croatia, Norway, Portugal and Spain. There are still many limitations in access to transplantation across the globe. World Kidney Day on March 8th 2012 will bring focus to the tremendous life-changing potential of kidney transplantation as a challenge to politicians, corporations, charitable organizations and healthcare professionals. This commentary raises awareness of the progressive success of organ transplantation, highlight concerns about restricted community access and human organ trafficking and commercialism, while also exploring the real potential for transforming kidney transplantation into the routine treatment option for ESRDacross the world. Correspondence: World Kidney Day Steering Committee 2012. World Kidney Day, International Society of Nephrology, Rue des Fabriques 1, 1000 Brussels, Belgium. [email protected] OUTCOMES OF KIDNEY TRANSPLANTATION The first successful organ transplantation is widely acknowledged to be a kidney transplant between identical twins performed in Boston on 23rd Dec 1954 which heralded the start of a new era for patients with ESRD1. In the development years between 1965 and 1980, patient survival progressively improved towards 90% and graft survival rose from less than 50% at one year to at least 60% after a first deceased donor kidney transplant, based on immunosuppression with azathioprine and prednisolone. The introduction of ciclosporin in the mid 1980s was a major advance, leading to one year survival rates of more than 90% and graft survival of 80%2. In the last 20 years, better understanding of the benefits of combined immunosuppressant drugs coupled with improved organ matching and preservation, as well as chemoprophylaxis of opportunistic infections, have all contributed to a progressive improvement in clinical outcomes. Unsensitised recipients of first deceased donor kidney transplants and living donor recipients can now expect 1-year patient and transplant survival to be at least 95% and 90% respectively. 1 New developments have led several groups to report excellent results even from carefully selected ABO Blood group incompatible transplants in recipients with low titre ABO-antibodies 3. Even for those with high titres of donor specific HLAantibodies, who were previously ‘untransplantable, better de-sensitisation protocols 4 and paired kidney exchange programs 5 now afford real opportunities for successful transplantation. Group members: World Kidney Day (WKD) is a joint initiative of the International Society of Nephrology and the International Federations of Kidney Foundations. WKD Steering Committee members: Abraham G, Beerkens P, Chapman JR, Couser W, Erk T, Feehally J, Garcia GG, Li PKT, Riella M, Segantini L, Shay P. 1 editorials Guillermo García-García et al. The global role of kidney transplantation Ethnic minorities and disadvantaged populations continue to suffer worse outcomes; Aboriginal Canadians, for example, have lower 10-year patient (50% vs 75%) and graft (26% vs 47%) survival compared with white patients.6 African American kidney transplant recipients have shorter graft survival compared to Asian, Hispanic, and White populations in the United States of America.7 In New Zealand, Maori and Pacific Island recipients of deceased donor transplants have a 50% 8-year graft survival compared to 14 years for non-indigenous recipients, in part due to differences inmortality.8 By contrast, despite a resource poor environment, Rizvi et alreport 1 and 5-year survival rates of 92% and 85%, respectively, among 2,249 living related kidney transplants in Pakistan,9 whilst in Mexico, 90% and 80% one-year survival for living and deceased donor kidney transplants, was reported among 1,356 transplants performed at a single centre.10 But, while it is possible to achieve such excellent long-term results, most patients and their families in resource poor environments are not able to afford the high cost immunosuppressants and antiviral medications needed to reduce the risk of graft loss and mortality.11 THE PLACE OF KIDNEY TRANSPLANTATION IN TREATMENT FOR ESRD Kidney transplantation improves long-term survival compared to maintenance dialysis. In 46,164 patients on the transplant waiting list in the USA between 1991-1997, mortality was 68% lower for transplant recipients than for those remaining on the transplant waiting list after >3 yrs follow-up.12 The transplanted 20-39 year old patients of both sexes were predicted to live 17 years longer than those remaining on the transplant waiting list, an effect that was even more marked in diabetics. The number of people known to have ESRD worldwide is growing rapidly, as a result of improved diagnostic capabilities and also the global epidemic of type 2 diabetes and other causes of chronic kidney disease (CKD). Dialysis costs are expensive even for developed countries, but prohibitive for many emerging economies. The majority of patients commencing dialysis for ESRD in low-income countries die or stop treatment within the first 3 months of initiating dialysis due to cost restraints.13 The cost of maintenance hemodialysis varies considerably by country and healthcare system. In Pakistan maintenance hemodialysisis reported to be US$1680 per year, which is beyond the reach of most of the population without humanitarian financial aid.14 Despite exemplars, both provision of hemodialysis facilities and uptake of peritoneal dialysis remain very limited in middle and low-income countries. Whilst the costs of transplantation exceed those of maintenance dialysis in the first year post-transplantation (eg. in Pakistan US$5245 vs US$1680 in the first year), the costs are much reduced compared to dialysis in subsequent 2 years, especially with the advent of inexpensive generic immunosuppression.15 Transplantation thus expands access and reduces overall costs for successful treatment of ESRD. Pre-emptive transplantation is an attractive option for both patients and payers with both reduced costs and improved graft survival.16 Pre-emptive transplantation is associated with a 25% reduction in transplant failure and 16% reduction in mortality compared to recipients receiving a transplant after starting dialysis.17 Transplantation of the kidney, when properly applied, is thus the treatment of choice for patients with ESRD because of lower costs and better outcomes. GLOBAL DISPARITIES IN ACCESS TO KIDNEY TRANSPLANTATION Substantial disparities in access to transplantation across the world are demonstrated in Figure 1 (derived from the World Health Organisation/OrganisationMondiale de la Sante (WHO/OMS) Global Observatory on Donation and Transplantation.18) which demonstrates the relationship between transplant rate and Human Development Index (HDI). There is a reduced transplant rate in low and middle HDI countries, and a large spread of transplant rates even amongst the richer nations. Transplant rates of more than 30 per million population (pmp) in 2010 were restricted to Western Europe, USA, and Australia, with a slightly broader spread of countries achieving between 20 and 30 pmp. There are also within-country disparities in transplant rates among minorities and other disadvantaged populations. In Canada, all minority groups have significantly lower transplant rates; compared to whites, rates in Aboriginal and African Canadians, Indo Asians, and East Asians were 46%, 34%, and 31% lower respectively.19 In the US, transplantation rates are significantly lower among AfricanAmericans, women, and the poor, compared to Caucasians, men and the more affluent populations.20 The situation is similar in Australia where Aboriginal Australians fare worse than non-indigenous Australians (12% vs 45%) and in New Zealand where Maori/Pacific Islanders are disadvantaged (14% vs 53%).21 In Mexico, the transplant rate among uninsured patients is 7 pmp compared with 72 pmp among those with health insurance.22 Multiple immunologic and non-immunologic factors contribute to social, cultural, and economic disparities in transplant outcomes, including biological, immune, genetic, metabolic, and pharmacological factors as well as associated co-morbidities, time on dialysis, donor and organ characteristics, patient socio-economic status, medication adherence, access to care, and public health policies.23 Developing countries often have especially poor transplant Nefrologia 2012;32(1):1-6 Guillermo García-García et al. The global role of kidney transplantation deceased organ donors, and most developed countries are trying to emulate their success. A return to ‘donation after cardiac death’ instead of the now standard ‘donation after brain death’, has enhanced the deceased organ donation numbers in several countries, with 2.8 DCD donors pmp in the US and 1.1 pmp in Australia now emanating from this source. Protocols for rapid cooling and urgent retrieval of kidneys after cardiac death, and in some circumstances other organs, have developed over the past five years to reduce the duration and consequences of warm ischaemia.25 Another strategy for increasing the rate of transplantation has been to extend the acceptance criteria for deceased organ donors. Such ‘extended criteria’ donors require additional consideration and specific consent by the recipient. There is risk in accepting an ‘extended criteria’ kidney since the transplants are less successful in the long term, but also a risk to waiting longer on dialysis for a standard criteria donor. rates not only because of these multiple interacting factors, but also because of inferior infrastructure and an insufficient trained workforce. Deceased donation rates may also be impacted by lack of a legal framework governing brain death and by religious, cultural and social constraints. When these factors are all compounded by patient anxieties about the success of transplantation, physician bias, commercial incentives favoring dialysis and geographical remoteness, poor access to transplantation is almost inevitable for most of the world’s population. IMPROVING ACCESS TO TRANSPLANTATION Both living donation and deceased donor donation are now recognized by the WHO as critical to the capacity of nations to develop self-sufficiency for organ transplantation.24 No country in the world, however, generates sufficient organs from these sources to meet the needs of their citizens. Austria, USA, Croatia, Norway, Portugal and Spain stand out as countries with high rates of AFR 60.0 AMR editorials A number of strategies have been designed and implemented to reduce disparities among disadvantaged populations. The EMR Croatia EUR SEAR Norway Portugal Spain 50.0 Deceased and Living Donor Kidney Transplants Per Million Population WPR Australia 40.0 Turkey Iran 30.0 C. Rica 20.0 Rep. of Corea Syria Singapore Mauritius Japan 10.0 India South Africa Thailand Algeria 0.0 0.5 0.6 0.7 0.8 0.9 1.0 Human Development Index Grouped by WHO Regions (AFR = Africa, AMR = Americas, EMR = Eastern Mediterranean, EUR = Europe, SEAR = South Eastern Asia, WPR = Western Pacific) Figure 1. Number of deceased and living donor kidney transplants in World Health Organisation Member States in 2010, correlated with human development index. Nefrologia 2012;32(1):1-6 3 editorials Guillermo García-García et al. The global role of kidney transplantation Transplantation Society has established the Global Alliance for Transplantation in an effort to reduce worldwide disparities in transplantation. The program includes collecting global information, expandingf education about transplantation, and developing guidelines for organ donation and transplantation. The International Society of Nephrology (ISN) Global Outreach program has catalysed the development of kidney transplant programs across a large number of countries with targeted fellowship training and creation of long term institutional links between developed and developing transplant centers through its Sister Center Program. This has led to the establishment of successful kidney transplantation in countries such as Armenia, Ghana and Nigeria where none existed before and expansion of existing programs in Belarus, Lithuania and Tunisia. A model of collaboration for dialysis and transplantation between government and the community in the resource poor world has been successfully established in Pakistan with government assistance for infrastructure, utilities, equipment, and up to 50% of the operating budget, while the community, including affluent individuals, corporations and the public, donate the remainder. 14 In 2001, in Central America, a specialized unit of pediatric nephrology and urology was opened in Nicaragua with funds provided initially by the Associazione per il Bambino Nefropatico, a kidney foundation based in Milan, Italy supplemented by a consortium of private and public organizations, including the International Pediatric Nephrology Association and the Nicaraguan Ministry of Health. Subsequently the Nicaraguan government and a local kidney foundation recognized the success of the program and accepted gradual transfer of the costs of treatment, including the provision of immunosuppressive medications for renal transplantation. A similar successful partnership between government and private sector has recently been reported in India. 26 ETHICAL CHALLENGES AND THE LEGAL ENVIRONMENT The impact of the global organ donor shortage and the dramatic disparities demonstrated by the WHO data, are experienced in many different ways requiring varied responses. But one common factor is the relative wealth of the nation and the individual. The poor receive the fewest transplants and the rich are most often transplanted either in their own country or through finding an organ through illegal purchase from the poor or an executed prisoner. Trafficking in human organs and commercialisation of the beneficial act of organ donation were unusual and extremely hazardous in the 1980’s, became frequent but still very hazardous in the 1990’s, then becoming a gruesomely burgeoning trade from the turn of the century. The WHO has estimated that up to 10% of all organ transplants were of commercial origin by 2005.27 The first WHO Guiding Principles in this field were agreed in 1991 and made clear by the decision of national governments to ban commercialization of organ donation and transplantation.28 This principle was reaffirmed unanimously by theWorld Health Assembly in 2010 when the updated WHO Guiding Principles for human organ and tissue donation and transplantation, were endorsed.29 Almost all countries with transplantation programs and even some without active programs have carried that ban on commercialism through to their own legislation, making it illegal to buy or sell organs. Sadly this has not prevented continuation of the trade illegally in countries such as China and Pakistan, nor has it prevented new entrants to this lucrative trade from taking advantage of their own or other nations’ impoverished and vulnerable populations to provide kidneys and even livers for the desperate wealthy in need of transplantation. There are tremendous opportunities to correct disparities in kidney disease and transplantation worldwide, but it is important to recognize that funding of ESRD treatment should be associated with funding for early detection and prevention of the progressive kidney diseases that lead to ESRD. Comprehensive programs should include community screening and prevention of CKD, especially in high-risk populations, as well as dialysis and transplantation for ESRD. Iran, alone, claims to have resolved national self sufficiency for kidney transplantation through a scheme of part government, part patient-funded sale of kidneys by vendors. The resultant slow development of deceased organ donation in Iran restricting liver, heart and lung transplant programs, as well as the disparity of socioeconomic status between donors and recipients, both testify to the universality of the problems that arise from organ transplant commercialisation. The restriction of transplantation to Iranian nationals only under this program has however largely ensured that this national experiment has not flowed on to create commercial organ trafficking across Iranian national borders. An integrated approach to the expansion of transplantation requires training programs for nephrologists, transplant surgeons, nursing staff, and donor coordinators; nationally funded organ procurement organizations providing transparent and equitable retrieval and allocation; and the establishment of national ESRD registries. The Transplantation Society and the ISN have taken a joint stand against the despoiling of transplantation therapy and victimization of the poor and vulnerable by doctors and other providers operating in these illegal programs. In 2008, more than 150 representatives from across the world from different disciplines of health care, national policy 4 Nefrologia 2012;32(1):1-6 Guillermo García-García et al. The global role of kidney transplantation development, law and ethics came together in Istanbul to discuss and define professional principles and standards for organ transplantation. The resultant Declaration of Istanbul30 has now been endorsed by more than 110 professional and governmental organizations and implemented by many of these organizations with a goal to eliminate transplant tourism and enhance the ethical practice of transplantation globally.31 SUMMARY There remain major challenges to providing optimal treatment for ESRD worldwide and a need, particularly in low income economies, to mandate more focus on community screening and implementation of simple measures to minimize progression of CKD. The recent designation of renal disease as an important noncommunicable disease at the UN High Level Meeting on NCDs is one step in this direction.32 But early detection and prevention programs will never prevent ESRD in everyone with CKD, and kidney transplantation is an essential, viable, cost-effective and life-saving therapy which should be equally available to all people in need. It may be the only tenable long-term treatment option for ESRD in low-income countries since it is both cheaper and provides a better outcome for patients than other treatment for ESRD. However, the success of transplantation has not been delivered evenly across the world, and substantial disparities still exist in access to transplantation, we remain troubled by commercialization of living donor transplantation and exploitation of vulnerable populations for profit. There are solutions available. These include demonstrably successful models of kidney transplant programs in many developing countries; growing availability of less expensive generic immunosuppressive agents; improved clinical training opportunities; governmental and professional guidelines legislating prohibition of commercialization and defining professional standards of ethical practice; and a framework for each nation to develop self sufficiency in organ transplantation through focus on both living donation and especially nationally managed deceased organ donation programs. The ISN and TTS have pledged to work together in coordinated joint global outreach programs to help establish and grow appropriate kidney transplant programs in low and middle income countries utilizing their considerable joint expertise. World Kidney Day 2012 provides a focus to help spread this message to governments, all health authorities and communities across the world. Conflicts of interest Authors declare no potential Conflicts of interest. Nefrologia 2012;32(1):1-6 editorials REFERENCES 1. Murray JE. Ronald Lee Herrick Memorial: June 15, 1931-December 27, 2010. Am J Transplant 2011;11(3):419. 2. Clayton P, Excell L, Campbell S, McDonald S, Chadban S. Transplantation. In: McDonald S, Excell L, Livingston B (eds.). ANZDATA Registry Report 2010. Australia and New Zealand Dialysis and Transplant Registry. Adelaide, South Australia; p. 8.1-8.31. Available at http://www.anzdata.org.au/anzdata/AnzdataReport/33rdReport/Ch 08.pdf. [Accessed nov 29, 2011]. 3. Shimmura H, Tanabe K, Ishida H, Tokumoto T, Ishikawa N, Miyamoto N, et al. Lack of correlation between results of ABO-incompatible living kidney transplantation and anti-ABO blood type antibody titers under our current immunosuppression.Transplantation 2005;80(7):985-8. 4. Peng A, Vo A, Jordan SC. Transplantation of the highly human leukocyte antigen–sensitized patient: long-term outcomes and future directions. Transplantation Reviews 2006;20:46-156 5. Warren DS, Montgomery RA. Incompatiblekidney transplantation: lessons from a decade of desensitization and paired kidney exchange. Immunol Res 2010;47(1-3):257-64. 6. Weber CLC, Rush DN, Jeffery JR, Cheang M, Karpinski ME. Kidney transplantation outcomes in Canadian aboriginals. Am J Transplant 2006;6:1882-9. 7. Gordon EJ, Ladner DP, Caicedo JC, Franklin J. Disparities in kidney transplant outcomes: A review. Semin Nephrol 2010;30(1):81-9. 8. Collins JF. Kidney disease in Maori and Pacific people in New Zealand. Clin Nephrol 2010;74:S61-S65. 9. Rizvi SA, Naqvi SAA, Zafar MN, Hussain Z, Hashmi A, Hussain M, et al. Living related renal transplants with lifelong follow-up. A model for the developing world. Clin Nephrol 2010;74 Suppl 1:S142-9. 10. Monteón FJ, Gómez B, Valdespino C, Chavez S, Sandoval M, Flores A, et al. The kidney transplant experience at Hospital de Especialidades, Centro Médico Nacional de Occidente, IMSS, Guadalajara México. In: Cecka JM, Terasaki P (eds.). Clinical Transplants 2003. Los Angeles: UCLA Inmunogenetics Center; 2003. p. 165-74. 11. Jha V. Current status of end-stage disease care in South Asia. Ethn Dis 2009 Spring;19(1 Suppl 1):S1-27-32. 12. Wolfe RA, Ashby VB, Milford EL, Ojo AO, Ettenger RE, Agodoa LY, et al.Comparison of mortality in all patients on dialysis, patients on dialysis awaiting transplantation and recipients of a first cadaveric transplant. N Engl J Med 1999;341:1725-30. 13. Sakhuja V, Sud K. End-stage renal disease in India and Pakistan: Burden of disease and management issues. Kidney Int Suppl 2003;(83):S115-8. 14. Rizvi SAH, Naqvi SAA, Zafar MN et al. A Renal Transplantation Model for developing countries. Am J Transplant 2011:11:2302-7. 15. Sud K, et al. Indian J of Nephrology 1999;9:83-91. 16. Meier-Kriesche HU, Kaplan B. Waiting time on dialysis as the strongest modifiable risk factor for renal transplant outcomes. Transplantation 2002;74(10):1377-81. 17. Kasiske BL, Snyder JJ, Matas MD, Ellison MD, Gill JS, Kausz ATl.Preemptive kidney transplantation: The advantage and the advantaged. J Am Soc Nephrol 2002;13:1358-6. 18. World Health Organization. Global Knowledge Base on Transplantation. Available at http://www.who.int/transplantation/knowledgebase/en/. [Accessed nov 29, 2011]. 5 editorials Guillermo García-García et al. The global role of kidney transplantation 19. Yeates K. Health Disparities in renal disease in Canada. Semin Nephrol 2010;30:12-8. 20. Alexander GC, Sehgal AR. Barriers to cadaveric renal transplantation among blacks, women, and the poor. JAMA 1998;280:114852. 21. McDonald S. Incidence and treatment of ESRD among indigenous peoples of Australasia. Clin Nephrol 2010;74:suppl 1:S28-31. 22. Garcia-Garcia G, Renoirte-Lopez K, Marquez-Magaña I. Disparities in renal care in Jalisco, Mexico. Semin Nephrol 2010;30:3-7. 23. Gordon EJ, Ladner DP, Caicedo JC, Franklin J. Disparities in kidney transplant outcomes: A review. Semin Nephrol 2010;30:81-9. 24. 3rd Global WHO Consultation March 2010. Organ Donation and Transplantation: Striving to Achieve Self-Sufficiency. Transplantation 2011;91 11S:S27-S114. 25. Bernat JJ, D’Alesandro AM, Port FK, Bleck TP, Heard SO, Medina J, et al. Report of a National conference on donation after cardiac death. Am J Transplant 2006;6:281-91. 26. Abraham G, John GT, Sunil S, Fernando EM, Reddy YNV. Evolution of renal transplantation in India over the last four decades. NDT Plus 2010;3:203-7. 27. Shimazono Y. The state of the international organ trade: a provisio- 28. 29. 30. 31. 32. nal picturebasedonintegration of availableinformation. Bull World Health Organ 2007;85(12):955-62. World Health Organization. Forty-fourth World Health Assembly, resolution and decisions. Geneva, Switzerland: World Health Organization; 1991 (WHA 44/1991/REC/1). Annex 6. World Health Organization. Sixty-third World Health Assembly. Geneva, Switzerland: World Health Organization; 2010. Available at: http://www.who.int/transplantation/Guiding_PrinciplesTransplantation_WHA63.22en.pdf. [Accessed nov 29, 2011]. Participants in the International Summit onTransplantTourism and Organ Trafficking Convened by the Transplantation Society and International Society of Nephrology in Istanbul, Turkey, April 30-May 2, 2008. The Declaration of Istanbul on organ trafficking and transplanttourism. Transplantation 2008;86(8):1013-8. Delmonico FL, Domínguez-Gil B, Matesanz R, Noel L. A call for government accountability to achieve nationalself-sufficiency in organdonation and transplantation. Lancet 2011;378(9800):1414-8. United Nations General Assembly. Political declaration of the High-level Meeting of the General Assembly on the Prevention and Control of Non-communicable Diseases A/66/L. 1, September 16, 2011. Sent for review: 9 Jan. 2012 | Accepted: 9 Jan 2012. 6 Nefrologia 2012;32(1):1-6 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society editorials Carotid ultrasound for the early diagnosis of atherosclerosis in chronic kidney disease Àngels Betriu-Bars1, Elvira Fernández-Giráldez2 Unitat de detecció i tractament de les malalties aterotrombòtiques (UDETMA) del Servicio de Nefrología. Hospital Universitari Arnau de Vilanova. Institut de recerca biomèdica de Lleida. Universidad de LLeida. Spain 2 Servicio de Nefrología. Unitat de detecció i tractament de les malalties aterotrombòtiques (UDETMA) del Servicio de Nefrología. Hospital Universitari Arnau de Vilanova. Institut de recerca biomèdica de Lleida. Universidad de LLeida. Spain 1 Nefrologia 2012;32(1):7-11 doi:10.3265/Nefrologia.pre2011.Dec.11258 A therothrombotic disease (ATD) is a progressive disorder and its most common clinical manifestations, acute myocardial infarction and stroke, are responsible for the highest morbidity and mortality rates in the Western world. Sudden death due to infarction with no prior symptoms occurs in 50% of men and 64% of women.1 One of the main tools to control the incidence of vascular disease is prevention. Formulas are available to estimate cardiovascular risk (Framingham risk score, etc.)2 They are based on epidemiological studies that determine the risk of suffering a cardiovascular event (fatal or non-fatal) within 5 to 10 years.3 While the available system to calculate cardiovascular risk is easily generalisable, and universal, it does present some difficulties. The main drawback is its low sensitivity for detecting individuals at a high risk to suffer a cardiovascular event. Large epidemiological studies show that 62% of subjects with a prior history of myocardial infarction present none or only one of the cardiovascular risk factors,4 which, undoubtedly, prevents them from being properly identified at a time to take effective preventive actions. In recent years, there have been significant technological advances in medical imaging techniques, particularly in the field of vascular disease. Below, we describe the advantages, disadvantages and uses of imaging techniques in the general population and in chronic kidney disease (CKD) patients. Correspondence: Àngels Betriu Bars Servicio de Nefrología. Unitat de detecció i tractament de les malalties aterotrombòtiques. Rovira Roure, 80. 25198 Lleida. Spain. [email protected] [email protected] GENERAL POPULATION Various methods have been used for the diagnosis and prevention of cardiovascular disease (CVD). Studies on the calcification of coronary arteries using helicoidal or multidetector computed tomography (CT) accurately measure coronary calcium content (coronary calcium score). However, this method is not useful in patients at a low cardiovascular risk according to traditional estimation methods. 5 Although coronary CT has a high specificity to diagnose coronary atherosclerosis, and some studies suggest that it directly correlates to mortality over time,6 this is a costly technique that requires advanced technology, and therefore, it cannot be used for the early detection of ATD in the general population. More significantly, coronary calcium scores measure the presence of calcium deposition in the coronary arteries, and therefore, it fails to diagnose early stages of vascular disease, as ATD is a progressive vascular disorder, and calcification only occurs in its advanced stages. Coronary arteriography has recently been complemented with the use of intravascular ultrasound7 techniques that offer a more precise diagnosis of coronary atherosclerosis. However, this method is painful and expensive, and therefore cannot be considered a good option for the early detection of ATD in the general population. Carotid ultrasound has several advantages compared to the previously described methods, but the use of this technique for early diagnosis of atherosclerosis is not generalised. Ultrasound of supra-aortic trunks is used in patients with symptomatic atherosclerosis, particularly those who have suffered a stroke. As these patients have already suffered an event due to arterial disease, we can only provide a diagnosis or confirmation of a suspected diagnosis and establish the surgical indication, rather than implementing preventive therapeutic interventions. 7 editorials Applying this technique in early diagnosis of ATD requires rigorous training of the technician and the specialist reading of the properly acquired images in order to obtain accurate parameters for the early diagnosis of the disease. Carotid intima-media thickness (cIMT) measured by carotid ultrasound was described by Pignoli et al in 1984 and validated by the same authors in 1986. They conclusively established a strong correlation between the thickness of the arterial wall, measured by ultrasound, with the histological findings in these patients.8 Since then, carotid ultrasound has been known as a technique with potential applications in the field of atherosclerosis, but 20 years later, it is not part of the routine clinical practice to diagnose vascular disease, mainly due to the high variability in the resulting parameters, highly dependent on the acquisition technique employed. The recommendations of the XIII European Stroke Conference in 2004 (Manheim IMT Consensus) 9 pconstitute a definite step forward in generalising the proper technique for clinical practice. Carotid artery ultrasound, which measures cIMT and detects atheromatous plaques (Figure 1 A and B), is a strong indicator of overall vascular health. cIMT is a well recognized and accepted marker to predict cardiovascular disease.10 In clinical trials, the cIMT is used alongside traditional risk factors. These trials highlight the usefulness and importance of using non-invasive techniques to evaluate the effect of risk factors on the vascular wall. Through the years, clinical trials have provided evidence corroborating the importance of measuring cIMT to prevent cardiovascular events (the thicker the cIMT, the higher the risk of myocardial infarction and cerebral infarction).11 cIMT was accepted as a surrogate marker of atherosclerosis by the US Food and Drug Administration (FDA) and the European Agency for the Evaluation of Medicinal Products. At present, cIMT is the noninvasive technique most frequently used in clinical trials to determine atherosclerosis. Clinical guidelines and medical societies recommend it to obtain a more precise evaluation of cardiovascular health in selected populations,12 and in cardiovascular disease prevention programmes.13 Upon adjustment for age and sex, an increase of 0.1 mm in cIMT, indicates a 10%-15% increase in the risk of myocardial infarction, and a 13%-18% increase in the risk of stroke.11 Davidsson14 and coworkers have recently shown, in a multivariate analysis of 391 men in Sweden, odd ratios for cardiovascular events of 2.09 (95% confidence interval (CI) 1.05-4.16; p= 0.03) in subjects with atheromatous plaques in the carotid artery. Nambi et al15 published the latest results from the ARIC study, demonstrating a correlation between an increase in cIMT , or the presence of atheromatous 8 Àngels Betriu-Bars et al. Early diagnosis of atherosclerosis plaques, and the incidence of cardiovascular events. They reported the predictive role of carotid ultrasound (cIMT and plaques) in a large epidemiological study of 13,145 healthy individuals, who presented 1,812 adverse cardiovascular events over a 15-year follow up. The area under the curve estimating the sensitivity and accuracy of their logistic regression analysis increased significantly when cIMT and the presence of atheromatous plaque were added to the list of conventional cardiovascular risk factors. CAROTID DISEASE ULTRASOUND IN CHRONIC KIDNEY Current tables to estimate cardiovascular risk have not been designed for CKD patients, whose main cardiovascular risk factors are different from those in the general population. CKD patients experience the so-called “reverse epidemiology”. In studies in haemodialysis patients, no association was found between serum cholesterol levels or systolic blood pressure with coronary artery disease, cerebrovascular disease or peripheral vascular disease.16,17 Furthermore, there are other “non-traditional” and “emerging” factors that play a role in the pathogenesis of atherosclerosis in CKD, such as anaemia, chronic inflammation, oxidative stress, and abnormalities in mineral metabolism that are not taken into account when using traditional predictors of cardiovascular risk. This evidence, along with the high incidence rate of CVD among CKD patients,18 supports the use of new, more sensitive tools for the early detection and prevention of cardiovascular disease.19 Guidelines from the European Society of Cardiology recommend a close monitoring of traditional cardiovascular risk factors in patients with chronic kidney disease.20 , Furthermore, the National Kidney Foundation (NKF) recommends the use of carotid ultrasound for a more precise assessment of cardiovascular health in kidney disease.21 Because cardiovascular disease is the main cause of morbidity and mortality in CKD,22 early diagnosis of vascular disease using ultrasound techniques is becoming increasingly widespread in this patient population. We lack conclusive evidence on how changes in cIMT relate to cardiovascular risk factors in CKD, but it has been reported that cIMT increases as glomerular filtration rate decreases.23 Our group has confirmed the high prevalence and severity of atherosclerosis in patients with different stages of kidney disease (n=409). 24 The diagnostic methods used were carotid ultrasound and the ankle-brachial index (ABI). Both tests have been validated as predictors of adverse cardiovascular events. 25 We have developed a Nefrologia 2012;32(1):7-11 Àngels Betriu-Bars et al. Early diagnosis of atherosclerosis A editorials B (A) View of the artery wall for measuring carotid intima-media thickness (cIMT) or the distance between the tunica adventitia and the intima layer of the arterial wall, at the level of the common carotid artery. Arrows delimit the cIMT; (B) Plaque along the upper wall of the common carotid artery (plaque defined as cIMT >1.5mm). Figure 1. Ultrasound image of carotid arteries classification of vascular disease based on the results of ABI and carotid ultrasound, using for the cIMT reference intervals adjusted for age and sex. We found that a high percentage of kidney patients had atheromatous vascular disease, and this percentage was much higher than in ageand sex-matched control subjects with a normal kidney function (n=851). Relevant findings from this study were the higher prevalence of ATD in patients on dialysis compared to those at earlier CKD stages, and the lack of correlation between vascular disease and LDL cholesterol levels. The presence of atheromatous plaque was higher in dialysis patients (78.3%) 26 compared to pre-dialysis patients (55.6%) 27 or the normal control group (43.2%). Variables with a positive association with atherosclerosis included age, dialysis treatment, and serum levels of plasma C-reactive protein. Being a female was a protective factor also in CKD. Carotid ultrasound also offers a critical additional information, the assessment of vascular calcification. Vascular calcification is a strong predictor of morbidity and mortality in the general population and in CKD. In CKD patients, vascular calcification is more prevalent, appears at an earlier age, and is more severe.28 The degree of arterial calcification can be measured using CT to obtain the Agatston score, which is the only quantitative method currently available, and therefore it is the gold standard, mostly used in clinical research. Semi-quantitative methods have been proven useful, as those described by Adragao and Kauppila. They employ simple radiology, are more adapted to Nefrologia 2012;32(1):7-11 clinical practice, and have proved to have a good predictive power of cardiovascular events in CKD. Carotid ultrasound is also the only available technique to localise the site of calcium deposition within the arterial wall. Our group has completed an ultrasound study examining the location of calcium in carotid and femoral artery wall. The brachial artery served as a negative control since this area is free of atherosclerosis.29 We observed that calcification was localized exclusively in the intimal layer, following two distinct patterns: 1) Calcification of the atheromatous plaque (Figure 2 A) and 2). Linear calcification of the intimal layer, anatomically unrelated to the atheromatous plaque (Figure 2 B). Multivariate analysis associated this previously unrecognized intimal calcium location with age, serum levels of phosphate and C-reactive protein, and with the presence of atheromatosis. These results underscore the contribution of inflammation and atheromatosis to the calcification of elastic arteries. Although serum phosphate levels remain a not fully resolved issue, we must start focussing on preventing atheromatosis and decreasing the pro-inflammatory status of CKD patients as much as possible. CONCLUSIONS Carotid ultrasound is a tool with a number of advantages compared to other methods. First, it is easy to use and does not require complex equipment. It can be performed in 9 Àngels Betriu-Bars et al. Early diagnosis of atherosclerosis editorials A B Arrows indicate the calcified areas: (A) linear calcification in the intima; (B) calcified or type 5 plaque (with posterior acoustic shadow). Clin J Am Soc Nephrol 2010;6(2):303-10. Figure 2. Calcification patterns depicted by ultrasound. outpatient clinics or at the bedside; and is inexpensive and non invasive. Secondly, it provides doctors with valuable information for treatment, including the atheromatous burden, the severity of the calcification, and the location of calcium deposition within the vascular wall. Thirdly, it detects ATD at early stages, prior to the appearance of atheromatous plaque. Therefore, it constitutes an ideal technique to obtain evidence-based recommendations for ATD prevention. At present, we lack studies addressing the prognostic value of either the calcification of atheromatous plaques (type V plaque according to the American Heart Association) or the new linear profile of intima calcification in kidney disease patients. The Spanish study NEFRONA (www. nefrona.es) has been designed to answer important questions on the pathogenesis and progression of ATD at all CKD stages compared to ageand sex- matched controls with normal renal function through measurements of the distinct atherosclerosis and calcification patterns between groups using arterial ultrasound. Conflicts of Interest Authors declare potential conflicts of interest Honoraria as speaker: Shire, Abbott, Genzyme and Amgen 10 Acknowledgements: Grants: FIS (Carlos III Health Institute). REFERENCES 1. Naghavi M, Falk E, Hecht HS, Jamieson MJ, Kaul S, Berman D, et al.; for the SHAPE Task Force. From vulnerable plaque to vulnerable patient –Part III: executive summary of the screening for heart attack prevention and education (SHAPE) Task Force Report. Am J Cardiol 2006;98(2A):2H-15H. 2. Dawber TR, Meadors GF, Moore FE. Epidemiological Approaches to Heart Disease: The Framingham Study. American Journal of Public Health 1951;41:279-86. 3. Graham I, Ater D, Borch-Johnsen K, Boysen G, Burell G, Cifkova R, et al. European Guidelines on cardiovascular disease prevention. Fourth Joint European Societes Task Force on Cardiovasculr Disease Presentation in Clinical Practice. European J Cardiovasc Prevent and Rehabil 2007;14(S2):S1-S113. 4. Khot U. Prevalence of Conventional Risk Factors in Patients with Coronary Heart Disease. JAMA 2003;290:898-904. 5. Shaw LJ, Raggi P, Schisterman E, Berman DS, Callister TQ. Prognostic value of cardiac risk factors and coronary artery calcium screening for all cause mortality. Radiology 2003;228:826-33. 6. Budoff MJ, Shaw LJ, Liu ST, Weinstein SR, Moler TP, Tseng PH, et al. Long-term prognosis associated with coronary calcification. Observations from a registry of 25,253 patients. J Am Coll Cardiol 2007;49:1860-70. 7. Von Birgelen C, Hartmann M, Mintz GS, Van Houwelingen G, Deppermann N, Schmeermund A, et al. Relationship between Nefrologia 2012;32(1):7-11 Àngels Betriu-Bars et al. Early diagnosis of atherosclerosis 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. cardiovascular risk as predicted by established risk scores versus plaque progression as measured by serial intravascular ultrasound in left main coronary arteries. Circulation 2004;110:1579-85. Pignoli P, Tremoli E, Poli A, Oreste P, Paoletti R. Intimal plus medial thickness of the arterial wall: a direct measurement with ultrasound imaging. Circulation 1986;74(6):1399-406. Toboul PJ, Hennerici MG, Meairs S, Adams H, Amarenco P, Bornstein N, et al. Manheim carotid intima-media thickness consensus (2004-2006). An Update of behalf of the advisory board of the 3er and 4th watching the risk symposium 13th and 15th European Stroke conferences, Mannheim, Germany, 2004 and Brussels, Belgium 2006. Cerebrovasc Dis 2007;23:75-80. Stein JH, Korcarz CE, Hurst RT, Lonn E, Kendall CB, Mohler ER, et al.; American Society of Echocardiography Carotid Intima-Media Thickness Task Force. Use of carotid ultrasound to identify subclinical vascular disease and evaluate cardiovascular disease risk: a consensus statement from the American Society of Echocardiography Carotid Intima-Media Thickness Task Force. Endorsed by the Society for Vascular Medicine. J Am Soc Echocardiogr 2008;21:93-111. Lorenz MW, Markus HS, Bots ML, Rosvall M, Sitzer M. Prediction of clinical cardiovascular events with carotid intima-media thickness: a systematic review and meta-analysis. Circulation 2007;115:459-67. Bots ML, Dijk JM, Grobbee OA, Diederick E. Carotid intima-media thickness, arterial stiffness and risk of cardiovascular disease current evidence. J Hypertens 2002;20:2317-25. Verhamme P, Kerkhof F, Buysschaert I, Rietzschel E, de Groot E. Carotid intima-media thickness: more than a research toal? Review Article. Acta Cardiol 2010;65(1):59-66. Davidsson L, Fagerberg B, Bergström G, Schmidt C. Ultrasoundassessed plaque occurrence in the carotid and femoral arteries are independent predictors of cardiovascular events in middle-aged men during 10 years of follow-up. Atherosclerosis 2010;209:469-73. Nambi V, Chambless L, Folsom AR, He M, Hu Y, Mosley T, et al. Carotid intima-media thickness and presence or absence of plaque improves prediction of coronary heart disease risk: the ARIC (Atherosclerosis Risk In Communities) study. J Am Coll Cardiol 2010;55:1600-7. Iseki K, Yamazato M, Tozawa M, Takishita S. Hypocholesterolemia is a significant predictor of death in a cohort of chronic hemodialysis patients. Kidney Int 2002;61:1887-93. Cheung AK, Sarnak MJ, Yan G, Dwyer JT, Heyka RJ, Rocco MV, et al. Atherosclerotic cardiovascular disease risks in chronic hemodialysis patients. Kidney Int 2000;58:353-62. editorials 18. Go AS, Chertow GM, Fan D, McCullock CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med 2004;351:1296-1305. 19. Tuttle KR, Short RA. Longitudinal relationships among coronary artery calcification, serum phosphorus, and kidney function. Clin J Am Soc Nephrol 2009;12:1968-73. 20. European Guidelines on Cardiovascular Disease Prevention in clinical practice: executive summary. Eur J Cardiovasc Prev Rehabil 2007;14 Suppl 2:E1-40. 21. KDIGO clinical practice guideline for the diagnosis, evaluation, prevention and treatment of chronic kidney disease-mineral and bone disorder (CKD-MBD). Kidney Int Suppl 2009;(113):S1-130. 22. Go AS, Chertow GM, Fan D, McCullock CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med 2004;351:1296-305. 23. Desbien AM, Chonchol M, Gnahn H, Sander D. Kidney function and progression of carotid intima-media thickness in a community study. Am J Kidney Dis 2008;51:584-93. 24. Coll B, Betriu A, Martínez-Alonso M, Borràs M, Craver L, Amoedo ML, et al. Cardiovascular risk factors underestimate atherosclerotic burden in chronic kidney disease: usefulness of non-invasive tests in cardiovascular assessment. Nephrol Dial Transplant 2010;25:3017-25. 25. Ix JH, Katz R, de Boer IH, Kestenbaum IR, Allison MA, Siscovick DS, et al. Association of chronic kidney disease with the spectrum of ankle brachial index the CHS (Cardiovascular Health Study). J Am Coll Cardiol 2009;54:1176-84. 26. Krasniak A, Drozdz M, Pasowicz M, Chmiel G, Michalek M, Szumilak D, et al. Factors involved in vascular calcification and atherosclerosis in maintenance hemodialysis patients. Nephrol Dial Transplant 2007;22:515-21. 27. Leskinen Y, Lehtimäki T, Loimaala A, Lautamatti V, Kallio T, Huhtala H, et al. Carotid atherosclerosis in chronic renal failure-the central role of increased plaque burden. Atherosclerosis 2003;171:295-302. 28. Nolan RL, Morton AR, Pickett W. Prevalence and associations of coronary artery calcification in patients with stages 3 to 5 CKD without cardiovascular disease. Am J Kidney Dis 2008;52:849-58. 29. Coll B, Betriu A, Martínez-Alonso M, Amoedo ML, Arcidiacono MV, Borrás M, et al. Large Artery Calcification on Dialysis Patients Is Located in the Intima and Related to Atherosclerosis. Clin J Am Soc Nephrol 2010;6(2):303-10. 30. Junyent M, Martínez M, Borrás M, Betriu A, Coll B, Craver L, et al. Utilidad de las técnicas de imagen y biomarcadores en la predicción del riesgo cardiovascular en pacientes con enfermedad renal crónica en España: Proyecto NEFRONA. Nefrologia 2010;30(1):119-26. Send to review: 28 Nov. 2011 | Accepted: 6 Dec. 2011 Nefrologia 2012;32(1):7-11 11 editorial comments http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society See original article in Nefrologia Sup Ext 2011;2(7):7-13 Longitudinal observational studies and causality Alfonso Muriel1, Domingo Hernández-Marrero2, Víctor Abraira1 1 2 Unidad de Bioestadística Clínica. Hospital Universitario Ramón y Cajal, IRYCIS, CIBERESP. Madrid. Spain Servicio de Nefrología. Hospital Universitario Carlos Haya. Málaga. Spain Nefrologia 2012;32(1):12-4 doi:10.3265/Nefrologia.pre2011.Nov.11167 R esults from randomised clinical trials (CT) and CT meta-analyses provide the best scientific evidence for evaluating the effect of a treatment according to the hierarchical rankings for clinical research methods. Observational studies (OS) hold a lower position in the evidence rankings, and are considered to have less probative force for the following reasons: a) they may overestimate the effects of the treatment due to patient heterogeneity; b) they may contain biases that are inherent to their design and nature; and c) on some occasions, the interpretation of results may be confusing. However, the controlled conditions found in a CT mean that their results may not be directly applicable to patients in clinical practice. An OS, on the other hand, measures effectiveness that cannot feasibly be obtained in any other way. In clinical research, and in the field of nephrology in particular, the analysis of large patient registers or clinical databases provides information whose usefulness should not be underestimated,1,2 and which may complement CT results. Using this strategy, we can perform medical research that is closer to daily clinical practice, and focuses on the normal conditions experienced by the patients themselves.3 Generally speaking, OS, and cohort studies in particular, are useful for assessing the effect of a treatment that was not assigned at random. Here, treatment is established based on common medical practice or on the patient’s individual characteristics. Establishing a link between a treatment and a certain result may depend on a number of biases, including confounding by indication, since in this type of study, treatment is not assigned at random and may be related to health results. One obvious example is when the indication for the treatment in some situations is determined based on clinical guidelines or consensus for a specific disease. A number of statistical models have been created to assess the effect of a treatment and to check for potentially Correspondence: Alfonso Muriel García Unidad de Bioestadística Clínica, Hospital Universitario Ramón y Cajal, IRYCIS, CIBERESP, Ctra. Colmenar, km 9.100, 28034 Madrid. Spain. [email protected] [email protected] 12 confounding co-variables. For example, regression models can offer measures to associate a treatment with a result, but they cannot establish causality measures due to the lack of interchangeability among patients. Causal relationships can be established in a CT, since the patients are interchangeable and assigned to different groups at random. Interchangeability cannot be assumed in an OS, since the assigned group depends on the patient’s conditions, expressed as co-variables that change over time, and on the treatment the patient receives according to the co-variables that were analysed. It may therefore occur that timedependent variables are affected by the treatment itself. This means that both the co-variables and the treatment may change the patient’s prognosis. Marginal structural models (MSM) were proposed in the late 1990s by members of the Harvard School of Public Health3,4 to evaluate causal relationships and avoid biases in longitudinal studies. MSM are an alternative to classical regression models when there is a time-dependent confounder that is associated with the event in question, but it is also related to the treatment being evaluated. These models are called “structural” because they study causality, rather than simple association. Causal inference is performed by means of comparing, theoretically, the results of treating all patients with the results of treating none of the patients. Marginal structural models use a weighted form of the propensity score, called IPTW (inverse probability of treatment weight), in such a way as to simulate a population in which treated and untreated patients do not differ in any of their co-variables, therefore allowing us to assume interchangeability for treated and untreated patients. MSM are used in clinical research in order to resolve questions of causality. As noted by Hernan, the scenario is typically represented by observational studies that are analysed like CT.5 This type of model can be implemented by using common statistical software applications. The journal NBE published an article entitled “Modelos estructurales marginales: una herramienta útil que proporciona evidencia a los estudios observacionales” (marginal structural models: a tool for studying causal relationships in observational studies)6 which provides Alfonso Muriel et al. Observational studies and causality practical information about this type of statistical tool. This article also provides an in-depth account of the conceptual bases of an MSM, the assumptions and conditions it must contain, and the way to implement it by using specifically designed statistical software. In many medical specialties, including nephrology, patients are monitored over long periods of time as part of their care requirements, and they receive a number of treatments that are added according to their condition at a specific time. An MSM is ideal in this type of situation for avoiding confounding by indication biases. To cite an example, MSM were applied in OS of HIV-positive patients receiving antiretroviral drugs7-10; in patients taking aspirin due for cardiovascular disease11; and in patients who received corticosteroids for asthma12 or rheumatoid arthritis.13 In nephrology, MSM have also been used to determine the causal relationship between multiple treatments or exposures and the final outcome of a disease. In this respect, multiple OS have shown that patients on high doses of erythropoietin (EPO) could have higher mortality rates, although it was also suspected that patients requiring higher doses of EPO could have more co-morbidities. Likewise, the efficacy of restricting phosphorus in a diet or using a certain dialysis technique (haemodialysis vs peritoneal dialysis) to decrease mortality in kidney patients are also topics of considerable debate. Although a preliminary analysis shows that very high doses of EPO are associated with higher mortality, this effect disappeared with the implementation of a more complete MSM. 14 Use of MSM also revealed that neither restricting dietary phosphate in these patients nor the type editorial comments of dialysis (haemodialysis or peritoneal dialysis) had a decisive influence on mortality in this patient group. 15,16 Lastly, the use of MSM in a multi-centre OS from the Netherlands showed that loss of residual renal function was associated with higher mortality rate, 17 which supports the possibility of implementing kidney replacement therapy that is personalised according to the urinary volume of the patient at the start of chronic dialysis. In the field of kidney transplantation, the main purpose of MSM is to estimate the impact of metabolic changes on mortality and to evaluate the efficacy of immunosuppressive and cardioprotective drugs on both patient and kidney graft survival. In fact, the use of Cox regression models and MSM, adjusted for confounders, showed that high glycaemia and more intense insulin treatment was associated with higher mortality. 18 At the same time, with the help of MSM, it was shown that the use of mycophenolate mofetil was a more effective aid to kidney graft survival than azathioprine. 19 Lastly, a recent OS performed by our group by MSM found that the use of renin-angiotensin system blockers in kidney transplant patients is associated with a lower risk of mortality, but does not guard against losing the graft. 20 These findings confirm the usefulness of implementing an MSM in cohort OS of renal patients, but it remains unclear whether carrying out randomised CT on these patients will support these results. As we await new evidence, use of MSM in cohort OS employing time-dependent variables provides results that are valid and complementary to CT results for evaluating the clinical efficacy of specific treatments. KEY CONCEPTS 1. OS results may be comparable to those from clinical trials in terms of therapeutic efficacy, given adequate, rigorous OS design and data analysis methods. 2. The presence of time-dependent clinical confounders can result in overestimating or underestimating the effect of treatment when conventional regression models are used, due to confounding by indication. 4. The choice of confounders for implementing MSM must be done from a clinical standpoint, using the appropriate statistical support. Faulty choice of these variables can result in a bias that changes the estimator variance. 5. When using an OS to assess the efficacy of a treatment, employing MSM can increase the level of evidence, making it applicable and extendable to daily clinical practice. 3. MSM can prevent confounding by indication biases, but their assumptions and conditions must be verified. Nefrologia 2012;32(1):12-4 13 editorial comments Acknowledgements The authors would like to thank Ramón y Cajal University Hospital’s Biostatistics Unit (IRYCIS, Madrid, Spain; CIBER Epidemiology and Public Health CIBERESP) and the kidney transplant teams at University Hospital of the Canary Islands (Tenerife) and Carlos Haya Hospital (Málaga) for their participation. Conflict of interest The authors declare they have no potential conflicts of interest related to the contents of this article. REFERENCES 1. Dreyer NA, Garner S. Registries for robust evidence. JAMA 2009;302(7):790-1. 2. Hernández D, Pascual J, Abraira V, Lorenzo V, Quereda C. Estudios observacionales y registros como fuentes de evidencia en el trasplante renal. Nefrologia 2006;26 Suppl 5:66-76. 3. Hernán MA. With great data comes great responsibility: publishing comparative effectiveness research in epidemiology. Epidemiology 2011;22(3):290-1. 4. Robins JM, Hernán MA, Brumback B. Marginal structural models and causal inference in epidemiology. Epidemiology 2000;11(5):550-60. 5. Hernán MA, Alonso A, Logan R, Grodstein F, Michels KB, Willett WC, et al. Observational studies analyzed like randomized experiments: an application to postmenopausal hormone therapy and coronary heart disease. Epidemiology 2008;19(6):766-79. 6. Muriel A, Hernández D, Abraira V. Modelos estructurales marginales: una herramienta útil que proporciona evidencia a los estudios observacionales. Nefrologia Sup Ext 2011;2(7):7-13. 7. Edmonds A, Yotebieng M, Lusiama J, Matumona Y, Kitetele F, Napravnik S, et al. The effect of highly active antiretroviral therapy on the survival of HIV-infected children in a resource-deprived setting: a cohort study. PLoS Med 2011;8(6):e1001044. 8. Petersen ML, Wang Y, van der Laan MJ, Guzman D, Riley E, Bangsberg DR. Pillbox organizers are associated with improved adherence to HIV antiretroviral therapy and viral suppression: a marginal structural model analysis. Clin Infect Dis 2007;45(7):908-15. Alfonso Muriel et al. Observational studies and causality 9. Petersen ML, Wang Y, van der Laan MJ, Rhee SY, Shafer RW, Fessel WJ. Virologic efficacy of boosted double versus boosted single protease inhibitor therapy. AIDS 2007;21(12):1547-54. 10. Hernán MA, Brumback BA, Robins JM. Estimating the causal effect of zidovudine on CD4 count with a marginal structural model for repeated measures. Stat Med 2002;21(12):1689-709. 11. Cook NR, Cole SR, Hennekens CH. Use of a marginal structural model to determine the effect of aspirin on cardiovascular mortality in the Physicians’ Health Study. Am J Epidemiol 2002;155(11):104553. 12. Kim C, Feldman HI, Joffe M, Tenhave T, Boston R, Apter AJ. Influences of earlier adherence and symptoms on current symptoms: a marginal structural models analysis. J Allergy Clin Immunol 2005;115(4):810-4. 13. Choi HK, Hernan MA, Seeger JD, Robins JM, Wolfe F. Methotrexate and mortality in patients with rheumatoid arthritis: a prospective study. Lancet 2002;359(9313):1173-7. 14. Wang O, Kilpatrick RD, Critchlow CW, Ling X, Bradbury BD, Gilbertson DT, et al. Relationship between epoetin alfa dose and mortality: findings from a marginal structural model. Clin J Am Soc Nephrol 2010;5(2):182-8. 15. Lynch KE, Lynch R, Curhan GC, Brunelli SM. Prescribed dietary phosphate restriction and survival among hemodialysis patients. Clin J Am Soc Nephrol 2011;6(3):620-9. 16. Mehrotra R, Chiu YW, Kalantar-Zadeh K, Bargman J, Vonesh E. Similar outcomes with hemodialysis and peritoneal dialysis in patients with end-stage renal disease. Arch Intern Med 2011;171(2):110-8. 17. van der Wal WM, Noordzij M, Dekker FW, Boeschoten EW, Krediet RT, Korevaar JC, et al. Full loss of residual renal function causes higher mortality in dialysis patients; findings from a marginal structural model. Nephrol Dial Transplant 2011;26(9):2978-83. 18. Wiesbauer F, Heinze G, Regele H, Horl WH, Schernthaner GH, Schwarz C, et al. Glucose control is associated with patient survival in diabetic patients after renal transplantation. Transplantation 2010;89(5):612-9. 19. Kainz A, Heinze G, Korbely R, Schwarz C, Oberbauer R. Mycophenolate mofetil use is associated with prolonged graft survival after kidney transplantation. Transplantation 2009;88(9):1095-100. 20. Hernández D, Muriel A, Abraira V, Pérez G, Porrini E, Marrero D, et al. Renin-angiotensin system blockade and kidney transplantation: longitudinal cohort study. Nephrol Dial Transplant. 2011 May 28.doi:10.193/ndt/gfr276. [Epub ahead of print]. Sent for Review: 2 Nov. 2011 | Accepted: 30 Nov. 2011 14 Nefrologia 2012;32(1):12-4 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society editorial comments See original article on page 35 The value of serum free light chain assay in patients with monoclonal gammopathies and renal failure M. Luisa Campos1, Nuno M. Barbosa de Carvalho1, Guillermo Martín-Reyes2 1 2 The Binding Site Spain. Barcelona. Spain Servicio de Nefrología. Hospital Regional Universitario Carlos Haya. Málaga. Spain Nefrologia 2012;32(1):15-9 doi:10.3265/Nefrologia.pre2011.Nov.11098 M onoclonal gammopathies (MG) are blood dyscrasias that result from a neoplastic process in plasma cell (PC) clones, and are usually associated with an elevated production of a monoclonal protein (MP). This paraprotein can be an intact immunoglobulin or a light chain (LC). The immunoglobulins are composed of an identical pair of heavy chains joined to two identical kappa (κ) or lambda (λ) LC. The heavy and light chains are produced separately, and are later joined and secreted into the bloodstream. In order to ensure correct conformation of the immunoglobulins, PC produce an excess of LC, whose surplus is secreted along with the completed immunoglobulins. In this manner, 40% of LC are found free in the serum, not joined to heavy chains.1 These serum free light chains (sFLC) have a different conformation: κ FLC present as 25kDa monomers, whereas λFLC form 50kDa dimers. their equivalents in serum samples1,4; however, they are affected by the renal function of the patient, which is their primary disadvantage (Figure 1). Nephelometric techniques are the most precise and sensitive analytical methods available, but they do not make the distinction between monoclonal and polyclonal background. A recent addition to the last techniques has been the sFLC assay, which allows for measuring sFLC and determining monoclonality by establishing the ratio of κ vs λ sFLC.2 This assay allows the detection of κFLC and λFLC separately using polyclonal antibodies produced in sheep that specifically recognise epitopes in the constant region of LC that are hidden in intact immunoglobulins, but are exposed in sFLC.1 The sFLC assay is the most sensitive assessment technique currently available. However, no technique can provide by itself a diagnosis of the various types of MG, and so they must be used together in order to provide a high sensitivity in the diagnosis.5 DIAGNOSING MONOCLONAL GAMMOPATHIES The diagnosis of MG is based on detecting the circulating paraprotein. Serum protein electrophoresis (SPE) is the most commonly used technique for measuring monoclonal bands, but its sensitivity is limited: it is incapable of detecting MP at concentrations <400mg/l, levels that are frequently observed in cases of Bence-Jones (BJ) or light chain multiple myeloma (BJMM).2 Immunofixation electrophoresis (IFE) is a more sensitive technique for characterising the type of MP but not to quantify it, and therefore it is of limited application during the MG follow-up. Both urine electrophoresis and immunofixation are more sensitive than Correspondence: M. Luisa Campos The Binding Site Spain. Balmes, 243, 4º 3a. 08006 Barcelona. Spain. [email protected] For the first time, sFLC assay allowed for measuring normal serum levels and determining a reference range for normal subjects. The normal range of κFLC is 3.3-19.4mg/l, and the normal range for λFLC is 5.7-26.3mg/l, with a normal interval for the κ/λ sFLC of 0.26-1.65.6 Using this range in a retrospective study, Bradwell demonstrated that 100% of patients with BJMM have an altered κ/λ ratio, suggesting monoclonality.7 In the field of MG, sFLC assay has shown a greater sensitivity for detecting the monoclonal component, with special relevance in non-secretory MM, in which it is now possible to measure and monitor MP without having to recurre to invasive techniques, and in the case of AL amyloidosis, where international guidelines recommend measuring sFLC as part of the diagnostic process.8,9 Additionally, measuring sFLC at the moment of diagnosing MM and other MG takes on a role in prognosis and will provide a valuable baseline value for monitoring the patient during treatment.8,10 Recently, Katzmann performed a study 15 M. Luisa Campos et al. The value of serum FLC assay editorial comments The quantity of circulating FLC depends on the level of production by PC and the rate of metabolisation by the kidneys. The renal glomerulus works as a filter with a cut-off point of 40-60kDa, which is easily passed by low molecular weight molecules. sFLC pass the pores of the glomerulus towards the proximal tubule of the nephron, where they are reabsorbed and metabolised. The capacity of reabsorption for sFLC by the kidney is 10-30g/day, and so normal individuals that produce 0.5-1g/day reabsorb all FLC, and they do not reach the urine. As a result of this metabolisation pathway, the half-life of these molecules in serum is considerably short (2-6 hours) when compared with the half-life of intact immunoglobulins (5-21 days). This difference in half-life is one of the primary advantages of sFLC assay in monitoring MG. AL AMYLOIDOSIS LCDD ACUTE TUBULAR NECROSIS FALCONI SYNDROME Proximal tubule Distal tubule 5-10mg/day in urine sFLC filtrated: glomerulus CAST NEPHROPATHY Absorption: 10-30g/day sFLC+ Tamm-Horsfall precipitate and produce waxy casts Cast nephropathy 50 000 5.000 5000 500 Bence-Jones proteinuria 250 100 2000 Urine FLC in urine (mg/l) 50.000 FLC in serum (mg/l) Serum levels of FLC depend on their metabolisation by the kidneys, but at the same time, an excess in FLC affects kidney function. In this paradox, we find the reason for analysing serum samples instead of urine samples in patients with MG and increased sFLC. The sFLC assay has high sensitivity, and can detect dyscrasia earlier, since serum levels will increase and exceed the normal range before they can be detected in urine samples. With the progression of the pathology, serum FLC levels will saturate the metabolic capacity of the kidney, and only after this point of Bence-Jones proteinuria will FLC be detectable in the urine. However, this saturation is associated with a high risk of nephropathy, and with time kidney function will decrease and less sFLC will be filtered, thus accumulating at a greater rate in serum, with a greater half-life. As such, from the moment of renal failure, free light chain assays using urine samples no longer show the true clinical situation of the patient. 1000 Serum 10 10 0 6 12 18 Time (months) 24 30 Normal range serum Normal range urine (Adapted from: Bradwell AR, courtesy of R Johnson and J Feehally.) FLC: free light chains; sFLC: serum free light chains; LCDD: light-chain deposition disease. Figure 1. Free light chains and the kidney with 1877 patients with MG, showing that a higher sensitivity for diagnosis could be achieved for the majority of malignant MG using a protocol of serum analyses (SPE + sFLC + sIFE). Based on their results, these authors proposed replacing IFE in 24-hour urine samples with the sFLC assay.11 This simple algorithm allows for orientating the diagnosis towards cast nephropathy (Figure 2). In a similar manner, the International Myeloma Working Group recommended using a combination of SPE, sFLC, and sIFE for tracking MG, and suggested that the sFLC assay could substitute IFE in 24-hour urine samples. This last test, however, still remains necessary in the case of strong, unconfirmed suspicion of AL amyloidosis.8 the normal range. This abnormal value in the κ/λ coefficient is proportional to the progression of the renal disease, and can pose a challenge when diagnosing patients with renal failure prior to MG.12,13 In order to avoid this issue, Hutchison measured κ and λ sFLC in 688 patients with renal failure and no MG, establishing a new range of normality for patients with renal failure at 0.37-3.1.12,13 The new “renal range” has been validated, and maintains a sensitivity of 100%, improving the specificity for the assay in cases of renal failure from 93% to 99%.13,14 NEPHROTOXIC PROPERTIES OF SERUM FREE LIGHT CHAINS RENAL FAILURE AND THE NEW RANGE OF NORMALITY FOR PATIENTS WITH A PRIOR ABNORMAL RENAL FUNCTION One important consideration for clinical practice is that the renal failure itself affects sFLC levels due to the reduced filtration rate. However, the sFLC assay has shown that, in the majority of cases of renal failure not derived from MG, the sFLC coefficient remains within its normal range. Some patients with renal failure have values that fall just outside of 16 MG are frequently associated with renal disease, and up to 50% of patients with MM already have altered renal function at the time of diagnosis (10%-20% dependent on dialysis).15,16 The nephrotoxic properties of sFLC are well known, and are attributed with the increased risk of suffering renal damage in patients with MG. Their toxicity can develop at different levels: 1) glomerular: formation of amyloid fibrils, usually λsFLC, or deposits in the basal membrane associated with κsFLC; 2) proximal tubule: the Nefrologia 2012;32(1):15-9 M. Luisa Campos et al. The value of serum FLC assay editorial comments Suspected MG Acute renal failure SPE sFLC Altered Normal No MG Clonal FLC <500mg/l No cast nephropathy, alternative MG? Clonal FLC > _500mg/l Probable cast nephropathy Consider renal biopsy: collaboration with haematology/oncology departments Altered Normal Strong clinical suspicion of AL sIFE No MG MG identified and classified IFE in serum and 24-hour urine No MG MG identified and classified (Adapted from Dispenzieri, et al.8 and Cockwell, et al.21) AL: AL amyloidosis; FLC: free light chain; PC: plasma cells; SPE: serum protein electrophoresis; MG: monoclonal gammopathies. Figure 2. Proposed protocol for the diagnosis and follow-up of acute renal failure/monoclonal gammopathy exacerbated process of reabsorption stimulates the production of cytokines and, consequently, the inflammatory response and fibrosis produced can lead to acute tubular necrosis; or, even, by the incapacity of cells to degrade FLC that accumulate intracellularly, forming crystals characteristic of Fanconi syndrome17; 3) the distal tubule: once the absorption capacity of the proximal tubule is surpassed, the FLC precipitate with Tamm-Horsfall proteins, causing cast nephropathy with the formation of waxy casts that eventually block the nephron and produce renal failure (Figure 1). Cast nephropathy, or “myeloma kidney,” is the most commonly seen nephropathy in MM. This condition is irreversible as long as the tumour remains active, and is associated with a worse prognosis, making effective tools for an early diagnosis essential, so as to detect MG before the appearance of this renal lesions.18 nephrology department, 100% of which are identifiable using sFLC assays.13,22 Recent studies have shown that a rapid decrease in sFLC allows for a greater rate of renal recovery. This is due to the progression of the disease towards interstitial fibrosis, which results in irreversible renal damage.20 Once more, the delay before clinical intervention will determine the prognosis, since 80% of patients require a reduction in sFLC >60% by day 21 in order to reach renal recovery, and this renal recovery entails a significant improvement in survival.23 A rapid diagnosis and start of treatment are possible through good interaction between nephrologists, haematologists, and oncologists, leading to improved survival. Given the high sensitivity of the test, sFLC measurement allows for a rapid identification of nephrotoxic monoclonal proteins in patients with acute renal failure.23 IMPACT OF AN EARLY DIAGNOSIS MONITORING TREATMENT The amount of time elapsed before a diagnosis is established and treatment is started is very important. A prolonged time until MM is diagnosed increases the probability of complications, among which stands out renal lesions.19,20 The nephrologist plays a central role in the early diagnosis of the disease as a substantial portion of patients develop acute renal failure without known MG.21 Particularly in relation to myeloma kidney, 50% of patients will seek treatment in the Nefrologia 2012;32(1):15-9 In addition to providing a tool for the early diagnosis of MG, sFLC assay plays an important role in monitoring the elimination of sFLC in patients with MG. The strategy for treating MM is based on the application of chemotherapy to eliminate the malignant PC clone, and at the same time, direct elimination of the circulating sFLC in the bloodstream. 17 editorial comments The treatment of MM has evolved substantially in recent years due to the development of more effective and faster drugs, such as thalidomide, lenalidomide, and bortezomib, with higher response rates and better long-term results. Bortezomib is approved by the FDA, is well-tolerated by patients with renal failure, and its pharmacokinetics is not affected by the level of renal failure, so it can be administered with no need for readjusting the dose. Bortezomib with dexamethasone is the recommended treatment for patients with MM and renal failure of any level.24 Plasmapheresis has not shown to be as effective as expected for eliminating sFLC.25,26 The fact that a large part of the circulating sFLC are located in the extravascular space, along with limits to the duration and frequency of plasmapheresis cycles, may be the basis for the lack of effectiveness observed with this treatment. However, the studies performed until now have supported the relationship between a reduction in sFLC levels and renal recovery.21,26 More recently, different haemodialysis filters have been developed with a high cut-off point (HCO), which allows for the passage of sFLC without an excessive loss of albumin.16,27 The HCO 1100 membrane has demonstrated its high effectiveness that, in combination with proper chemotherapy, produces promising results for the recovery of patients with myeloma kidney.28,29 A swift and constant decrease in sFLC was associated with recovery of the kidney in 74% of patients in the study by Hutchison,28 and 50% in the study published by Martin-Reyes29 in this same issue of Nefrología. Given the short half-life of sFLC in comparison to intact immunoglobulins, measuring sFLC allows for determining levels before and after each cycle of haemodialysis almost in real time.29 In this manner, measuring sFLC is important in monitoring the treatment of these patients because it allows us to: 1) evaluate the efficient elimination of sFLC during dialysis; 2) analyse the efficiency of chemotherapy during treatment, with the option of adjusting the treatment regimen as needed; 3) determine the moment in which the patient has recovered renal function, starting to metabolise the sFLC anew; and 4) early diagnosis of recurrence. Each year, the incidence of MM increases, but we have the possibility of providing more effective treatment to these patients. This is due to the development of more effective new-generation chemotherapies, in addition to the possibility of offering a rapid decrease in circulating sFLC, thus reducing the probability of irreversible renal damage, which reduces survival.30 As such, this is an important time of great advancements and expectations in the treatment of MM and the recovery of these patients, in which the clinician still plays an important role in providing attentive care and quick action. However, no therapy provides effective results after a late diagnosis, and the major advantage that we have today is a simple protocol that allows us to identify patients with clinically relevant MG efficiently and earlier. 18 M. Luisa Campos et al. The value of serum FLC assay Conflicts of interest The authors M. Luisa Campos and Nuno M. Barbosa de Carvalho work for Binding Site Spain. REFERENCES 1. Bradwell AR. Serum Free Light Chain Analysis (plus Hevylite). 6.ª ed. Birmingham, UK: The Binding Site Group Ltd.; 2010. 2. Bradwell AR, Carr-Smith HD, Mead GP, Tang LX, Showell PJ, Drayson MT, et al. Highly sensitive, automated immunoassay for immunoglobulin free light chains in serum and urine. Clin Chem 2001;47(4):673-80. 3. Keren DF. Procedures for the evaluation of monoclonal immunoglobulins. Arch Pathol Lab Med 1999;123(2):126-32. 4. Hutchison CA, Basnayake K, Cockwell P. Serum free light chain assessment in monoclonal gammopathy and kidney disease. Nat Rev Nephrol 2009;5(11):621-8. 5. Katzmann JA. Screening panels for monoclonal gammopathies: time to change. Clin Biochem Rev 2009;30(3):105-11. 6. Katzmann JA, Clark RJ, Abraham RS, Bryant S, Lymp JF, Bradwell AR, et al. Serum reference intervals and diagnostic ranges for free kappa and free lambda immunoglobulin light chains: relative sensitivity for detection of monoclonal light chains. Clin Chem 2002;48(9):1437-44. 7. Bradwell AR, Carr-Smith HD, Mead GP, Harvey TC, Drayson MT. Serum test for assessment of patients with Bence Jones myeloma. Lancet 2003;361(9356):489-91. 8. Dispenzieri A, Kyle R, Merlini G, Miguel JS, Ludwig H, Hajek R, et al. International Myeloma Working Group guidelines for serumfree light chain analysis in multiple myeloma and related disorders. Leukemia 2009;23(2):215-24. 9. López-Corral L, García-Sanz R, San Miguel JF. [Value of serum free light chains assay in plasma cell disorders]. Med Clin (Barc) 2010;135(8):368-74. 10. Kyle RA, Durie BGM, Rajkumar SV, Landgren O, Blade J, Merlini G, et al. Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management. Leukemia 2010;24(6):1121-7. 11. Katzmann JA, Dispenzieri A, Kyle RA, Snyder MR, Plevak MF, Larson DR, et al. Elimination of the need for urine studies in the screening algorithm for monoclonal gammopathies by using serum immunofixation and free light chain assays. Mayo Clin Proc 2006;81(12):1575-8. 12. Hutchison CA, Harding S, Hewins P, Mead GP, Townsend J, Bradwell AR, et al. Quantitative assessment of serum and urinary polyclonal free light chains in patients with ch ro n i c k i d n e y d i se a se . C l i n J A m S o c Nep h ro l 2008;3(6):1684-90. 13. Hutchison CA, Plant T, Drayson M, Cockwell P, Kountouri M, Basnayake K, et al. Serum free light chain measurement aids the diagnosis of myeloma in patients with severe renal failure. BMC Nephrol 2008;9:11. Nefrologia 2012;32(1):15-9 M. Luisa Campos et al. The value of serum FLC assay 14. Abadie JM, van Hoeven KH, Wells JM. Are renal reference intervals required when screening for plasma cell disorders with serum free light chains and serum protein electrophoresis? Am J Clin Pathol 2009;131(2):166-71. 15. Bladé J, Fernández-Llama P, Bosch F, Montolíu J, Lens XM, Montoto S, et al. Renal failure in multiple myeloma: presenting features and predictors of outcome in 94 patients from a single institution. Arch Intern Med 1998;158(17):1889-93. 16. Hutchison CA, Cockwell P, Reid S, Chandler K, Mead GP, Harrison J, et al. Efficient removal of immunoglobulin free light chains by hemodialysis for multiple myeloma: in vitro and in vivo studies. J Am Soc Nephrol 2007;18(3):886-95. 17. Reyes GM, González IG, Alférez MJ, González JMM, Frutos MA. Cristales intracitoplasmáticos y síndrome de Fanconi en un paciente con mieloma IgA Kappa. Nefrologia 2001;XXI(2):213-6. 18. Herrera GA, Joseph L, Gu X, Hough A, Barlogie B. Renal pathologic spectrum in an autopsy series of patients with plasma cell dyscrasia. Arch Pathol Lab Med 2004;128(8):875-9. 19. Kariyawasan CC, Hughes DA, Jayatillake MM, Mehta AB. Multiple myeloma: causes and consequences of delay in diagnosis. QJM 2007;100(10):635-40. 20. Basnayake K, Cheung CK, Sheaff M, Fuggle W, Kamel D, Nakoinz S, et al. Differential progression of renal scarring and determinants of late renal recovery in sustained dialysis dependent acute kidney injury secondary to myeloma kidney. J Clin Pathol 2010;63(10):884-7. 21. Cockwell P, Hutchison CA. Management options for cast nephropathy in multiple myeloma. Curr Opin Nephrol Hypertens 2010;19(6):550-5. 22. Hutchison CA, Bridoux F. Renal impairment in multiple myeloma: time is of the essence. J Clin Oncol 2011;29(11):e312-313; author reply e314. editorial comments 23. Hutchison CA, Cockwell P, Stringer S, Bradwell A, Cook M, Gertz MA, et al. Early reduction of serum-free light chains associates with renal recovery in myeloma kidney. J Am Soc Nephrol 2011;22(6):1129-36. 24. Dimopoulos MA, Terpos E, Chanan-Khan A, Leung N, Ludwig H, Jagannath S, et al. Renal impairment in patients with multiple myeloma: a consensus statement on behalf of the International Myeloma Working Group. J Clin Oncol 2010;28(33):4976-84. 25. Clark WF, Stewart AK, Rock GA, Sternbach M, Sutton DM, Barrett BJ, et al. Plasma exchange when myeloma presents as acute renal failure: a randomized, controlled trial. Ann Intern Med 2005;143(11):777-84. 26. Leung N, Gertz MA, Zeldenrust SR, Rajkumar SV, Dispenzieri A, Fervenza FC, et al. Improvement of cast nephropathy with plasma exchange depends on the diagnosis and on reduction of serum free light chains. Kidney Int 2008;73(11):1282-8. 27. Ward RA. Protein-leaking membranes for hemodialysis: a new class of membranes in search of an application? J Am Soc Nephrol 2005;16(8):2421-30. 28. Hutchison CA, Bradwell AR, Cook M, Basnayake K, Basu S, Harding S, et al. Treatment of acute renal failure secondary to multiple myeloma with chemotherapy and extended high cutoff hemodialysis. Clin J Am Soc Nephrol 2009;4(4):745-54. 29. Martín-Reyes G, Toledo R, Torres A, et al. Tratamiento con hemodiálisis del fracaso renal agudo en el mieloma múltiple con filtros de alto poro. Nefrologia 2012;32(1):35-43. 30. Reyes GM, Valera A, Frutos MA, Ramos B, Ordónez V, Novales EL. Supervivencia de pacientes con mieloma tratados con diálisis. Nefrologia 2003;XXIII(2):131-6. Sent for review:20 Jul. 2011 | Accepted: 9 Nov. 2011 Nefrologia 2012;32(1):15-9 19 short review http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society Renal supportive care and palliative care: revision and proposal in kidney replacement therapy Juan P. Leiva-Santos1, Rosa Sánchez-Hernández2, Helena García-Llana3, M. José Fernández-Reyes2, Manuel Heras-Benito2, Álvaro Molina-Ordas2, Astrid Rodríguez2, Fernando Álvarez-Ude2 Cuidados Paliativos-MIR MFyC. Hospital General de Segovia. Spain Servicio de Nefrología. Hospital General de Segovia. Spain 3 Servicio de Nefrología. Hospital Universitario La Paz, IdiPAZ. Madrid. Spain 1 2 Nefrologia 2012;32(1):20-7 doi:10.3265/Nefrologia.pre2011.Nov.11065 ABSTRACT Patients with chronic kidney disease may receive sustained renal supportive care and renal palliative care (RPC) starting with the diagnosis of the disease, throughout the various stages of renal replacement therapy (RRT), the cessation of the RRT, and in the decision of whether to provide conservative treatment or non-initiation of RRT. This article reviews the literature on the development of renal palliative care and proposed RPC models. We describe the progression of disease in organ failure, which is very different from other areas of palliative care (PC). We describe important components of resident nephrology training in PC. We discuss the management of pain and symptom control, as well as communication skills and other psychological and ethical aspects in the renal patient. We conclude that in chronic renal patients, a palliative care approach can provide a positive impact on the quality of life of patients and their families, as well as optimizing the complex treatment of the renal patient. Keywords: Renal Palliative Care. Opioids and Renal Failure. Renal Palliative Care and Organization. Cuidados de soporte renal y cuidados paliativos renales: revisión y propuesta en terapia renal sustitutiva RESUMEN El paciente con enfermedad renal crónica es susceptible de recibir tratamiento de soporte y cuidados paliativos renales (CPR) desde el diagnóstico de la enfermedad, durante las distintas etapas de tratamiento sustitutivo renal (TSR), en el cese de dicho TSR y también si se decide tratamiento conservador o no inicio de TSR. Este artículo revisa la literatura referente al desarrollo de cuidados CPR y los modelos propuestos. Exponemos la trayectoria de la enfermedad en el fallo de órgano, que marca diferencias respecto a otros campos de los cuidados paliativos (CP). Se describen componentes de formación importantes para el residente de nefrología en CP. Abordamos el manejo del dolor y el control de síntomas, así como habilidades de comunicación y otros aspectos psicológicos y éticos en el paciente renal. Concluimos que en la atención al paciente renal crónico, un enfoque desde la medicina paliativa puede suponer un provechoso impacto en la calidad de vida del paciente y su familia, además de optimizar el complejo tratamiento nefrológico del paciente. Palabras clave: Cuidados paliativos renales. Opioides en insuficiencia renal. Organización en cuidados paliativos renales. INTRODUCTION The 21st century should be second nature to health professional has two obligations when performing his/her Correspondence: J.P. Leiva Santos Servicio de Nefrología. Hospital General de Segovia. Carretera de Ávila s/n. 40002 Segovia. Spain. [email protected] 20 job: to cure disease, and to alleviate suffering.1 In the case of chronic incurable diseases, the objective of treatment is to improve or preserve body function so as to avoid a premature death. This objective is carried out through palliative care (PC), defined by the World Health Organisation (WHO) in 20022 as total, active, and continued care of the patient and the patient’s family by a multiprofessional team when the medical expectation is no longer Juan P. Leiva-Santos et al. Renal supportive care and palliative care one of curing the disease. The primary objective of this medicine field is not to prolong survival, but to achieve the highest possible quality of life for patients and their families. As such, PC practitioners must cover their patients’ physical, psychological, social, and spiritual needs.2 If necessary, this support must also be extended to the grieving process. Palliative medicine (PM) is not synonymous with terminal illness, although it does include it. There is a large knowledge base for the application of PM, but this experience is not benefiting the majority of people that have a need for it. Despite the efforts made in the last two decades, the majority of patients that require PC do not receive it.3 Recently, dramatic changes have occurred in the demographics of patients with advanced chronic kidney disease (ACKD).4 In 2009, the incidence of RRT in Spain was 126 patients per million population (pmp).5 Currently, approximately 1.5 million patients are receiving haemodialysis globally, and this value is growing at a 7% yearly rate in those populations that have the privilege of access to this type of treatment.6 Chronic kidney disease (CKD) follows a progressive deterioration through various stages, in which RRT is the final stage that only a small portion of CKD patients arrive at7; with this in mind, we performed a review focusing on the development of renal palliative care (RPC) and renal supportive care (RSC), areas of new and growing interest for nephrology in Spain. For a better analysis of the subject, we will focus on the following aspects: 1) Concepts of RSC and RPC 2) Bioethics: fundamental basis for RRT 3) Progression and stages of the disease short review the concept of life expectancy previously held by the patient, placing them in a scenario of uncertainty where the perception of control is lost and quality of life is threatened. However, this psychological situation is rarely recognised or discussed. Experts in PM should work together with nephrologists in order to provide the best possible care and quality of life for patients and their families.9 The future course of this type of treatment will involve integrating the principles and practices of PC in the different areas of nephrology including haemodialysis, peritoneal dialysis, renal transplants when necessary, dealing with patients with CKD where the indicated approach is a conservative treatment, and, of course, patients that require end of life care. The objective is to ensure that all renal patients with a limited life expectancy receive high quality care with the help of the most appropriate professionals. The concept of RSC has been analysed by Noble et al, using the Rogers method, which is based on the idea that concepts are dynamic, with constant modification and change. With this in mind, the following attributes of the concept of RSC were laid down 8: 1) RSC must be available from the moment of diagnosis to patient death, with emphasis on a clear prognosis and the impact of the advanced renal disease. One difference with respect to oncology is that, in the case of oncology, some patients may recover but CKD patients have a very high probability of death due to the complications associated with their disease. 2) Interdisciplinary approach to treatment. This facilitates avoiding the medicalisation of the psychological and existential needs associated with RRT patients. 4) Organisational models 5) Guidelines for training in PC for nephrology residents 6) Pain and symptom control 7) Psychological aspects and counselling A NEW CONCEPT: RENAL SUPPORTIVE CARE The term RSC is emerging as a central topic in nephrology. It deals with a concept that is similar to PC, end of life care, and conservative patient management, 8 but with several differences. Although very similar, they are not synonymous, and as such it requires a definition. To tell a patient that he/she has advanced renal failure and needs dialysis involves very bad news that shatters Nefrologia 2012;32(1):20-7 In addition, the concept of a nephrologist with extensive training in RPC has emerged, with great importance at centres where palliative care units (PCU) do not cover all the patients with non-oncological pathologies. 3) Attention given to the patient’s caretaker and family. The support of patient caretakers is essential in RSC. The pressures felt by financial budgets are one important factor, making the patient the central focus.10 In family renal disease, unaffected caretakers tend to have an added emotional burden that may go unnoticed at first.11 4) Therapeutic communication skills, which ensure proper and opportune shared decision making. The objectives of RSC are not reachable without proper training in communication skills.12 21 short review BIOETHICS: A FUNDAMENTAL BASIS FOR RENAL REPLACEMENT THERAPY Within the sphere of RPC interventions, we must pay special attention to certain clinical situations with a high emotional impact that can generate difficulties in shared decision making, such as the withdrawal of a patient from dialysis or the decision not to start it, among others.6 At this point in the development of the field of nephrology, we know that not all that is technically possible is ethically admissible. Modern bioethics is based on the development of four fundamental principles that should be second nature to doctors attending to patients on RRT (Table 1). A detailed study of these principles falls outside the aims of this review. PROGRESSION AND STAGES OF DISEASE Most of patients with ACKD die due to non-renal causes, and although the renal disease may contribute to exitus, it is not directly responsible.12 Four different progressions have been described for disease (Figure 1).13,14 1) Sudden death may occur at any point during the progression of the disease, with no previous diagnosis (Figure 1A). 2) Following diagnosis of the terminal disease, function is preserved initially, followed by a rapid deterioration in advanced stages (Figure 1B); which is the typical pattern in oncology. Juan P. Leiva-Santos et al. Renal supportive care and palliative care 3) This pattern involves recurrences of acute episodes, requiring hospitalisation occasionally, with no recovery of previous functional states (Figure 1C), which poses a risk of death. This is the typical pattern in organ failure. 4) There is a gradual decrease in function prior to death, which is characteristic of dementia and advanced age (Figure 1D). These patterns may overlap in a patient on RRT. Implications of the disease progressions For ACKD patients and their families, as well as for the health professionals that treat them, it is of vital importance to understand these disease progressions towards death.12 This is consistently becoming more important for nephrologists, who currently manage patients on RRT at more advanced ages and with more associated comorbidities. The disease progressions allow health care providers to: realise that “to do all that one can do may be the wrong choice,” carry out a practical plan for a good death, support both the patients and their caregivers, and avoid a “prognostic paralysis”.15 These implications lead us to the following conclusions: 1) that one single model does not work for all cases: the typical oncological PC model may not be adjustable to gradual and progressive patient decline with unpredictable exacerbations. Patients with non-malignant diseases may have more prolonged needs than, although similar in nature to, those of cancer patients; 2) transferable lessons: PC in oncology Table 1. The four principles of modern bioethics A patient, to not do damage (primum non nocere). Tends to be interpreted as the lex artis and the obligation to carry out only indicated actions and never contraindicated actions. 2. Principle of justice: equitable distribution of the costs and benefits of treatment, with no discrimination of patients based on any cause that might have to do with their social condition, sex, race, etc. The principle of justice has to do with equal opportunity and equitable allocation of available resources. Level II, or maximum: 3. Principle of autonomy: all people are, inherently and unless otherwise demonstrated, capable of making their own decisions regarding Terminal illness High Functional state 1. Principle of non-maleficence: the obligation to do no harm to the B Sudden death C High Death Death Low Organ failure D Fragile elderly patient Functional state 1. Level I or minimal: Death Death Low Time Time whether or not to receive treatments that will affect their health. 4. Principle of beneficence: we are obliged to aid our patients, provi- (Source: Lunney et al20) ding them with the greatest possible benefit. (Source: TL Beauchamp y JF Childress, 1979). 22 Figure 1. Typical progressions of disease, identified in different patients. Nefrologia 2012;32(1):20-7 Juan P. Leiva-Santos et al. Renal supportive care and palliative care provides experiences that we can benefit from; 3) challenges for research: patients receiving PC are an especially vulnerable group, for which there is not usually a “second opportunity” that allows for improving the care given to them. As such, the idea of assigning patients to sub-optimal conditions of health care is unacceptable.15 Research is difficult to carry out in this field of medicine, but even so should be given more emphasis. ORGANISATIONAL MODELS Spanish authors have proposed a PC programme for terminal uraemia in patients with stage 5 CKD that do not respond to dialysis, with glomerular filtration rates (GFR) <15ml/min and who refuse RRT.16 In the model we propose in this review, we extend this concept, considering clinical and psychological criteria to be paramount above laboratory parameters for including a patient on RSC and RPC programmes; for example, a patient with a functioning renal graft and a GFR of 60ml/min but with musculoskeletal pain may benefit from the aid of an expert, multi-disciplinary team that manages this pain and immunosuppression therapy without compromising the patient’s GFR. A PC programme for terminal uraemia patients is only part of the proposed approach (Figure 2). Tejedor et al also propose the creation of a PCU for ACKD patients. We believe it to more economically sound, safer, and more effective to train nephrology teams in RSC and RPC. Anyway, the essential step is to incorporate expert Renal supportive care ACKD C K D HD R R T Conservative ACKD treatment Cessation of RRT S L Terminal uraemia D PD: peritoneal dialysis; CKD: chronic kidney disease; ACKD: advanced chronic kidney disease; HD: haemodialysis; SLD: situation during last days; RRT: renal replacement therapy; RTx: renal transplant. Shaded boxes show the scenarios in which supportive and palliative renal care intervention is most intense. Figure 2. Proposed model for the application of renal supportive care and renal palliative care. Nefrologia 2012;32(1):20-7 doctors in PM and psychologists, who can offer RSC and RPC from the hospital PCU, in the context of the nephrology department. They can provide aid to the patients and their families, as well as direct support for the nephrologist.9 As such, it is essential to adapt the PM model to the nephrology context and to incorporate RSC.4 This proposed model is in accordance with a recent publication in nephrology regarding the sustainability and equity of RRT in Spain,17 and is a humble addition to this shared task. PC could be developed and carried out by interdisciplinary teams that would also include the patient’s family and friends (Figure 3),4 who appreciate and recognise the importance of this type of care before and after the death of the patient.18 In some countries such as in Canada and the United States, the approval of recent guidelines and national publications have established the role of PC in the field of nephrology.19 Similar experiences have been described in England, Poland, Australia, Portugal, and Hong Kong. GUIDELINES FOR TRAINING NEPHROLOGY RESIDENTS IN PALLIATIVE CARE In Spain there are no recommendations from the national committee on nephrology for residents to carry out a specific rotation period for PC. A Canadian study concluded that residents that had contact with PM specialists had better training in situations dealing with the final stages of disease and are considered more competent for providing necessary end of life care,20 and proposed a programme for PM training directed towards nephrology residents that covers the following aspects21: Renal palliative care KTx PD short review G R I E V I N G Nephrologist Renal nurse Nutritionist Geriatrics Family and friends Primary care doctor Primary care nurse – Physiotherapist – Dentist – Neurologist – Ophthalmologist Renal patient Palliative medicine specialist Social worker Occupational therapist Psychologist priest/spiritual advisor Palliative care nurse (Source: Lichodziejewska et al2) Figure 3. Diagram of the interactions amongst the multidisciplinary team in renal palliative care. 23 short review 1) Impact on the patient upon reaching the final stage of renal disease: life expectancy, survival, mortality, and comorbidity 2) PM-based focus in haemodialysis units 3) Living wills and cardiopulmonary resuscitation 4) Communication skills: counselling 5) Pain management in renal patients Juan P. Leiva-Santos et al. Renal supportive care and palliative care and intensity of pain during dialysis is higher than outside of dialysis, according to the VAS scale.26 The revised Edmonton Staging System (rESS),27 designed for the evaluation of oncological pain, could be a clinically useful tool for the study and treatment of pain in renal patients. This system has the advantage of predicting analgesic response, considering that a “difficult” pain does not necessarily mean that it cannot be controlled. In order to make a simple of effective evaluation, it is enough to know whether the pain is neuropathic, incidental episodes, or if it associated with emotional suffering and/or altered cognitive state, or drug abuse.27 6) Controlling symptoms in renal patients 7) Transferral to specialised PC units and grieving support 8) Clinical, ethical, and legal issues when starting and withdrawing dialysis treatment PAIN AND SYMPTOM CONTROL Arteriovenous fistulas are punctured an average of 300 times per year in haemodialysis patients, and are qualified as medium-intensity pain using the VAS scale.28 The postoperative treatment of pain in patients with ACKD poses a problem due to the fear of accumulating metabolites that would depress the respiratory system. This also has been described by anaesthesiologists, and several drugs are being researched for their efficacy and safety in postoperative ACKD patients.29 Pain Chronic pain is suffered by 50%-63% of dialysis patients, and 42%-55% of them classify their pain as severe.22 The evidence provided by dialysis guidelines suggest that little recognition is given to this pain and other symptoms in dialysis patients, with consequent under-treatment.23 The use of opioid analgesics is not very widespread, due to the threat of toxicity and a lack of experience in their use. This leads to inadequate treatment of pain in CKD patients. In a recent review on the use of opioids and benzodiazepines in dialysis patients, the effectiveness of treatment was variable and low; in 17%-38% of patients, the years spent on dialysis were correlated with the presence of pain, and 72%-84% of patients with severe pain did not receive any analgesia.24 Recent research into the use of cannabinoids has motivated the evaluation of their potential use in controlling symptoms in renal patients on RRT. Although they had promising results, at the moment this option is limited due to the absence of long-term results.25 Evaluation of pain The quantification and later follow-up of pain using scales should be an added parameter for monitoring the management of renal patients. Several different validated pain measurement scales have been applied to groups of haemodialysis patients: the Visual Analogue Scale (VAS), the Pain Management Index (PMI), the McGill Pain Questionnaire (MGPQ), and the Brief Pain Inventory (BPI). The most common aetiology of pain observed during dialysis is ischaemic pain, and the most common extradialysis pain is chronic musculoskeletal pain. The prevalence 24 Opioids in chronic kidney disease We have dedicated this section solely to opioid compounds listed for analgesia by the WHO, given the difficulty in their use and their notable usefulness in renal patients. The complex pharmacokinetics of opioid analgesics in patients with kidney disease, as well as a lack of familiarity with the medical use of these drugs, presents a barrier to the effective alleviation of pain.30 The development of renal failure as a consequence of opioid use has not been described.31 A prospective study performed with 12 patients on haemodialysis concluded that hydromorphone could provide a safe and effective option in certain patients on haemodialysis.32 Similar conclusions were reached in a study comparing the use of hydromorphone in patients with normal urea and creatinine vs patients with renal failure.33 The pharmacokinetic and pharmacodynamic profiles of hydromorphone, methadone, and fentanyl are apparently safe in patients with renal failure, making them recommendable in these cases. The dose should be adjusted when GFR<10ml/min.31 It has also been recommended to reduce the dose and/or increase the dosing interval in renal failure patients on dialysis. Frequent patient monitoring is also indicated, and may even include home telephone follow-up. It is also recommended not to use morphine or codeine due to the difficulty of managing the adverse effects and complications produced in these patients.31 Morphine metabolites could accumulate between dialysis sessions. Methadone metabolites are inactive and are not dialysed, and so do not require adjusted doses in dialysis patients.31 These studies are limited to populations with poor statistical significance, and so we must use the recommended drugs Nefrologia 2012;32(1):20-7 Juan P. Leiva-Santos et al. Renal supportive care and palliative care under close monitoring and perform clinical/laboratory evaluation of our patients. a complete short review Table 2. Stressors in advanced chronic kidney disease treatable through psychological interventions 1. Secondary effects of ACKD and its treatments Symptom control 2. Diet management and hydration restrictions 3. Anxiety and depression The general load of symptoms in CKD patients is high and similar to those produced at the end of life in cancer patients.34 4. Insomnia In patients with stage 5 CKD that are treated without the use of dialysis, the prevalence and severity of symptoms during the last month prior to death have been studied using the Memorial Symptom Assessment Scale-Short Form (MSAS-SF) 35; of the 74 patients that participated in the study, with a mean age of 81 years, 86% had severe asthenia, 84% had pruritus, 82% had somnolence, 80% had dyspnoea, 76% had difficulties concentrating, 73% suffered from pain, 71% had reduced appetite, 71% had oedema of the arms or legs, 69% had dry mouth, 65% had constipation, and 59% had nausea. The SMILE (Symptom Management Involving End-Stage Renal Disease) randomised clinical trial demonstrated that patients suffered from pain, sexual dysfunction, and depression when on haemodialysis, these being underdiagnosed symptoms even though they are very prevalent.36 7. Main caregiver burnout PSYCHOLOGICAL ASPECTS AND COUNSELLING The interest in incorporating psychological aspects into the care of these patients and their families has been part of PC since its beginning. However, the integration of psychologists into nephrology teams is not widespread. As such, the importance of these variables in PC of renal patients is shown at an earlier stage at the end of their lives. The field of RPC poses an ideal space in which psychologists could exercise their specialty.37 Depression is a psychological issue with a very high prevalence (10%-66%) in ACKD patients, and can be approached through psychological and/or combined intervention. 38 In certain scenarios, such as in peritoneal dialysis units, depression is under-diagnosed and difficult to treat. 38 Few controlled studies and even fewer randomised studies have considered this issue. Renal patients at the end of their lives are a special high-risk group, given their high vulnerability and dependence.39 In a similar manner, anxiety disorders can be quite common among renal patients at the end of their lives if preventative treatment is not given. The prevalence rate of anxiety disorders in haemodialysis patients is approximately 45.7%, with negative effects on patient quality of life.40 Nefrologia 2012;32(1):20-7 5. Social alienation 6. Functional limits and dependency 8. Spiritual crisis 9. Complicated grieving process: prevention and treatment 10. Exhaustion of the health care team. Prevention of burnout ACKD: advanced chronic kidney disease According to Cukor et al, ACKD entails a large number of stressors that the patient and his/her close friends and relatives must face (Table 2). Preventative measures in RPC must be the gold standard for avoiding psychopathologyzation at the end of patients’ lives. Renal patients have a history of disease associated with multiple losses that intensifies upon reaching advanced phases. It is normal to encounter scenarios such as those in hospital haemodialysis units, where “biographical death,” associated with reduced quality of life, can occur before “biological death.” In order to deal with these differences, psychological intervention in chronic advanced diseases requires protocols for maximising resources rather than models orientated towards psychological pathologies.41 Psychological therapies in RPC include support for the family and after the patient has died.23 In 10%-20% of cases, those grieving can have significant difficulties and even associated physical comorbidities in the process of adapting to the loss.42 One therapeutic tool that has demonstrated its effectiveness in PC is counselling, since this is an ideal method for therapeutic communication in circumstances of intense emotional reactions.43 There are several definitions of counselling available. According to Dietrich,44 at its core, it is a form of a helpful, interventional, and preventative relationship in which an advisor, through communication, attempts in a relatively short span of time to provoke an active process of cognitiveemotional learning in a disorientated or overloaded person, during the course of which this person can improve his/her disposition towards self-help, his/her capacity for selfdirection, and operational competence. 25 short review Juan P. Leiva-Santos et al. Renal supportive care and palliative care KEY CONCEPTS 1. The introduction of PC and its values within the field of nephrology generates a significant impact on the quality of life of renal patients and their families. 2. Clinical, psychological, and laboratory criteria are orientated towards including patients in RSC and RPC programmes, although this is not exclusively for terminal stage patients. 3. RSC and RPC involve a multidisciplinary team and can be improved by the inclusion of lessons learned in other fields of PC. 4. Training in PC for nephrology residents should be promoted within the teaching and research areas. 5. The study and management of pain in renal patients is a priority that should be promoted both within clinical practice and research. 6. Nephrology teams should be trained to develop communication skills such as counselling. To our knowledge, there is no term in Spanish that translates the full conceptual richness of the English word. Counselling is not an assessment, nor is it assisted or clinical advice, but it is the opposite of giving advice. Perhaps the most closely related term would be “helping relationship.” Conflicts of interest Counselling is not just an interventional tool based on problem solving and emotional management, but also a way of conceiving interpersonal relationships through an ethical concept based on recognising the capacity in others to make their own decisions. Counselling is the art and science of making open, focused questions in order to explore threats and detect resources. It is based on listening rather than talking, empathy rather than judgement, and a fundamental respect of the patient’s values rather than imposing our own. All of this is of the utmost importance during the end of a renal patient’s life. REFERENCES CONCLUSIONS It has been clearly demonstrated that the application of the principles of PC in the field of nephrology generates major benefits for the patient, the patient’s family, and the nephrologist. We are faced with a significant challenge in carrying out studies that offer better safety in the use of paincontrolling drugs and those used to treat other symptoms in these patients. The nephrologist also should learn to masterfully use the different options available for managing pain and suffering in advanced phases of renal disease, as well as communication skills for supporting the patients and their families. The development of PC in nephrology departments is a large and important development that we must strive for, involving the dedication of several different disciplines. 26 Authors declare no potential conflicts of interest . 1. Callahan D. Death and the research imperative. N Engl J Med 2000;342:654-6. 2. World Health Organisation. Cancer Pain relief and Palliative Care, Technical Report Series 804. Geneva: World Health Organisation; 1990. 3. Stjernswärd J, Clark D. Palliative medicine: A global perspective. En: Doyle D, Hanks G, et al. Oxford Textbook of Palliative Medicine, 3rd ed. New York: Oxford University Press; 2004. p. 1199-224. 4. Lichodziejewska M, Rutkowski B. Paliative care in nephrology. J Nephrol 2008; 21 (suppl 13):S153-S157. 5. Sociedad Española de Nefrología: Registros de diálisis y trasplante 2009, Available at: http://www.senefro.org/modules.php?name=webstructure&idwebstructure=128 6. Smith C, Da Silva-Gane M, Chandna S, Warwicker P, Greenwood R, Farrington K. Chooosing not to dialyse: evaluation of planned nondialytic management in a cohort of patients with end-stage renal failure. Nephron Clin Pract 2003;95(2):c40-6. 7. Alcázar R, De Francisco ALM. Acción estratégica de la SEN frente a la enfermedad renal crónica. Nefrologia 2006;26(1):1-4. 8. Noble H, Kelly D, Rawlings-Anderson K, Meyer J. A concept analysis of renal supportive care: the changing world of nephrology. J Adv Nurs 2007;59(6):644-53. 9. Arnold RM, Solomon L. Editorial: Renal palliative care: supporting our colleagues, patients and family. J Palliat Med 2006;9(4):975-6. 10. Moss AH. A new clinical practice guideline on initiation and withdrawal of dialisys that makes explicit the role of palliative medicine. J Palliat Med 2000;3:253-60. Nefrologia 2012;32(1):20-7 Juan P. Leiva-Santos et al. Renal supportive care and palliative care 11. Álvarez-Ude F, Valdés C, Estébanez C, Rebollo P. Health-related quality of life of family caregivers of dialysis patients. FAMIDIAL Study Group. J Nephrol 2004;17(6):841-50. 12. Holley JL. Paliative care in end stage renal disease: illness trajectories, comunication and hospice use. Adv Chronic Kidney Dis 2007;14:402-8. 13. Lunney JR, Lynn J, Foley DJ, Lipson S, Guralnik JM. Patterns of functional decline at the end of life. JAMA 2003;289:2387-92. 14. Murtagh FE, Murphy E, Sheerin NS. Illness trajectories: an important concept in the management of kidney failure. Nephrol Dial Transplant. 2008;23:3746-8. 15. Holley JL. Palliative care in end-stage renal disease. Focus on advance care planning, hospice referral, and bereavement. Semin Dial 2005;18:154-6. 16. Tejedor A, De las Cuevas X. Cuidado paliativo en el paciente con enfermedad renal crónica avanzada (Grado 5), no susceptible de tratamiento dialítico. Nefrologia 2008;3:129-36. 17. De Francisco ALM. Sostenibilidad y equidad del tratamiento sustitutivo de la función renal en España. Nefrologia 2011;31:241-6. 18. Levy JB, Chambers EJ, Brown EA. Supportive care for the renal patient. Nephrol Dial Transplant 2004;19(6):1357-60. 19. Renal Physicians Association and American Society of Nephrology. Shared Decision-Making in the Appropriate Initiation of and Withdrawal from Dialysis: Clinical Practice Guideline: 2. Washington, DC: Renal Physicians Association; 2000. 20. Holley JL, Carmody SS, Moss AH, Cohen LM, Block SD, Arnold RM. The need for end of life training in nephrology: national survey results of nephrology. Am J Kidney Dis 2003;42(4):813-20. 21. Moss AH, Holley JL, Davidson SN, Dart AR, Germain MJ, Cohen L, et al. Core curriculum in Nephrology. Palliative Care. Am J Kidney Dis 2004;43(1):172-85. 22. Moss AH. Revised dialysis clinical practice guidelines promoted more informed decision making. J AM Soc Nephrol. 2010;5(12):2380-3. 23. Calls J, Rodríguez Calero MA, Hernández Sánchez D. Evaluación del dolor en hemodiálisis mediante diversas escalas de medición validadas. Nefrologia 2009;29(3):236-43. 24. Wyne A, Rai R, Cuerden M, Clark WF, Suri RS. Opiodes and benzodiacepinas use in end stage renal disease: a systematic review. Clin J Am Soc Nephrol 2011;6:326-33. 25. Davison SN, Davison JS. Is there a legitimate role for the therapeutic use of cannabinoids for symptomatic management in chronic kidney disease. J Pain Symptom Manage 2011;41(4):768-78. 26. Calls J, Rodríguez Calero MA, Hernández Sánchez D. Evaluación del dolor en hemodiálisis mediante diversas escalas de medición validadas. Nefrologia 2009;29(3):236-43. 27. Fainsinger RL, Nekolaichuck CL, Lawlor PG, Neumann CM, Hanson J, Vigano A. Multicenter study of the revised Edmonton staging system for calsifying cancer pain in advanced cancer patients. J Pain Symptom Manage 2005;29:224-37. short review 28. Figueiredo AE, Viegas A, Monteiro M, Poli-de-Figuereido CE. Research into pain perception with arteriovenous fistula cannulation. Journal of Renal Care 2008;34(4):169-72. 29. Mimoz O, Chauvet S, Grégoire N, Marchand S, Le Guern ME, Saleh A, et al. Nefopam pharmacocinetic in patien with end stage renal disease. Anesth Analg 2010;111(5):1146-53. 30. Davison SN. Pain in hemodialysis patients: Prevalence, cause, severity, and management. Am J Kidney Dis 2003;42:1239-47. 31. Dean M. Opiods in renal failure and dialysis patients. J Pain Symptom Manage 2004;28:497-504. 32. Davison SN, Mayo PR. Pain management in chronic kidney disease: The pharmacokinetics and pharmacodynamics of hidromorphone and hidromorphone-3-glucuronide in hemodialysis patients. J Opioid Manag 2008;4(6):335-44. 33. Lee MA, Leng ME, Tiernan EJ. Restrospective study of the use of hydromorphone in palliative care patients with normal and abnormal urea and creatinine. Palliat Med 2001;15:26-34. 34. Davison SN, Jhangri GS, Johnson JA. Cross-sectional validity of modified Edmonton symptom assessment of symptom burden. Kidney Int 2006;69:1621-5. 35. Murtagh FE, Addington-Hall J, Edmonds P, Donohoe P, Carey I, Jenkins K, et al. Symptoms in advanced renal disease: a cross sectional survey of symptom prevalence in stage 5 chronic kidney disease managed without dialysis. J Palliat Med 2007;10(6):1266-76. 36. Weisbord SD, Shields AM, Mor MK, Sevick MA, Peternel J, Porter P, et al. Methodology of randomized clinical trial of symptom management strategies in patients receiving chronic hemodialysis: The SMILE study. Contemp Clin Trials 2010;31(5):491-7. 37. Barbero J. Psicólogos en cuidados paliativos. El largo camino de la extrañeza a la integración. Clínica Contemporánea 2010;1:39-48. 38. Wuerth D, Finkelstein SH, Kliger AS, Finkelstein FO. Chronic peritoneal dialysis patients diagnosed with clinical depression: results of pharmacologic therapy. Semin Dial 2003;16:424-7. 39. Cohen LM, Poppel DM, Cohn GM, Reiter GS. A very good death: measuring quality of diying in end-stage renal disease. J Palliat Med 2001;4(2):167-72. 40. Cukor D, Cohen SD, Peterson RA, Kimmel P. Psychosocial aspects of chronic disease. ESRD as a paradigmatic illness: J Am Soc Nephrol 2007;18:3042-3055. 41. Albee GW. A competency model to replace the defect model. En: Gibbs MS, Lachenmeyer JR, Sigal J (eds.). Community psychology. Nueva York: Gardner Press; 1980. p. 213-38. 42. Prigerson HG, Bierhals AJ, Kasl SV, Reynolds CF, Shear MK, Day N, et al. Traumatic grief as a risk factor for mental and physical morbidity. Am J Psychiatry 1997;154(5):616-23. 43. Arranz P. La comunicación en cuidados paliativos. Medicina Paliativa (Madrid) 2001;8(1):26-3. 44. Dietrich G. Psicología general del Counseling. Barcelona: Herder; 1986. Sent for review: 28 Jun. 2011 | Accepted: 27 Aug. 2011 Nefrologia 2012;32(1):20-7 27 special article http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society Start-up of a clinical sample processing, storage and management platform: organisation and development of the REDinREN Biobank Laura Calleros*1, María A. Cortés*1, Alicia Luengo1, Inés Mora1, Brenda Guijarro1, Paloma Martín1, Alberto Ortiz-Arduán2, Rafael Selgas3, Diego Rodríguez-Puyol4, Manuel Rodríguez-Puyol1 Departamento de Fisiología. Facultad de Medicina. Universidad de Alcalá. Instituto Reina Sofía de Investigaciones Nefrológicas (IRSIN). Madrid. Spain 2 Laboratorio de Investigación Renal y Vascular. IIS-Fundación Jiménez Díaz. Universidad Autónoma de Madrid. Instituto Reina Sofía de Investigaciones Nefrológicas (IRSIN). Madrid. Spain 3 Servicio de Nefrología. Hospital Universitario La Paz-IdiPAZ. Instituto Reina Sofía de Investigaciones Nefrológicas (IRSIN). Madrid. Spain 4 Sección de Nefrología y Unidad de Investigación. Hospital Príncipe de Asturias. Instituto Reina Sofía de Investigaciones Nefrológicas (IRSIN). Madrid. Spain * Both authors have contributed equally to this work 1 Nefrologia 2012;32(1):28-341(6):XX doi:10.3265/Nefrologia.pre2011.Oct.11121 ABSTRACT Background: The creation of the Biobank, a resource pertaining to the Spanish Renal Research Network (REDinREN) promotes advances in clinical research on kidney disease in Spain. The Biobank’s aims are to generate an archive of clinical samples and associated data, furnish those samples to research teams, and coordinate with European biobanks. Method: Applicable legislation had to be complied with in order to launch the Biobank project (Biomedical Research Law, Data Protection Law and Biological Sample Transport Regulations). A strict work protocol and a new database for the Network’s clinical data were also implemented. Results: Over time, the Biobank has acquired additional infrastructure and qualified personnel. In 2010, 2953 new patient samples were collected, giving a total of 37 043 stored vials containing different types of samples. Furthermore, the Biobank is currently participating in eleven research projects. Discussion: Although the Biobank was originally designed for REDinREN use, we must take joint action to make this biological sample storage system and the many possibilities it offers available to the entire nephrological community with a view to promoting kidney disease research. Puesta en marcha de una plataforma de proceso, almacenamiento y gestión de muestras clínicas: organización y desarrollo del Biobanco de REDinREN RESUMEN Antecedentes: La creación del Biobanco, como una plataforma dentro de la Red de Investigación Renal (REDinREN), impulsa el avance de la investigación clínica de la enfermedad renal en España. Los objetivos del Biobanco son la generación del archivo de muestras clínicas y de datos asociados, para su cesión a los grupos de investigación, y la coordinación con biobancos europeos. Métodos: Para su puesta en marcha, fue necesaria la implementación de la normativa vigente (Ley de Investigación Biomédica y de Protección de Datos y la Normativa del Transporte de Sustancias Biológicas), un estricto protocolo de trabajo y la creación de una base de datos clínicos de la Red. Resultados: En su evolución, el Biobanco ha adquirido infraestructura y personal cualificado, lo que permitió que en el año 2010 se obtuviera un total de 2.953 pacientes, lo que hace un total de 37.042 viales almacenados con muestras de diferentes naturalezas. Además, hasta la fecha, el Biobanco está incluido en 11 proyectos de investigación. Discusión: Aunque el Biobanco fue diseñado como una plataforma de soporte de la REDinREN, es necesario con una acción conjunta poner a disposición de toda la comunidad científica nefrológica las posibilidades que otorga este sistema de almacenamiento de muestras biológicas para potenciar la investigación de la enfermedad renal. Key Words: Biobank. Chronic kidney disease. Network Palabras clave: Biobanco. Enfermedad renal crónica. Support. Medical samples. Plataforma de red. Muestras clínicas. Correspondence: Laura Calleros Departamento de Fisiología. Facultad de Medicina. Universidad de Alcalá, Ctra. Madrid-Barcelona km 33.600. 28871 Alcalá de Henares, Madrid. Spain. [email protected] INTRODUCTION 28 The Biobank of the REDinREN (Spanish Renal Research Network) is a public, non-profit entity that stores a collection Laura Calleros et al. Evolution of the REDinREN Biobank of samples intended for use in biomedical research into kidney disease. Its purpose is to contribute to advancing scientific knowledge of nephrological diseases as one of the activities coordinated by REDinREN, which is dependent on the Carlos III Institute of Health (RD6/0016/0002). This network is made up of groups of researchers from different regions in Spain. It enables researchers to undertake interdisciplinary, multi-centre research projects in strategic areas that focus on gaining a better understanding of kidney disease, with an emphasis on providing solutions for real, current problems in the field of nephrology. The Biobank is currently an essential platform for undertaking joint research activities in REDinREN. The Biobank’s initial list of objectives is found in Table 1, and all of its objectives are still in force. The Biobank is therefore a cross-sectional resource that provides support to nephrological research, and it may be considered as a complement to other biobanks that were created in the frameworks of a range of institutional initiatives. METHOD Aspects fundamental to the creation of the REDinREN Biobank The REDinREN Biobank’s organizational structure and workings are based on four basic pillars: compliance with legislation, dedicated staff members, the development of standardised technical procedures, and a formal system to record and access clinical data. Each of these components is listed below. 1) Legal requirements: The REDinREN Biobank was designed according to the regulations stated in the Spanish Law for Biomedical Research (Law 14/2007 of 3 July).1 This Law states that patients must be informed by their doctors and give written consent before any samples may be sent to the Biobank. As a result, in one of the first steps toward creating the Biobank, the Ethics Committees at each hospital belonging to the network approved the Informed Consent forms that are provided to patients. Information is given in Table 1. Biobank objectives Create the REDinREN archive of biological samples by processing all extracted samples. Coordinate technical procedures that health centres must follow in order to donate samples to the Biobank. Create a system to organise samples and provide a computerised platform for their associated information. Coordinate action procedures with similar European Union organisations. Nefrologia 2012;32(1):28-34 special article writing, and it covers the details, significance, implications and risks of donating samples to the Biobank, and mentions the donor’s right to revoke consent at any time. To obtain the samples, the Biobank depends on the free will of people who may choose to donate their biological samples to science. The Biobank reserves the right to audit the samples that are sent in to ensure that consent was properly given. On a parallel note, the clinical database containing coded data pertaining to patients whose samples are stored in the Biobank has been registered with both the Spanish Data Protection Agency and the Madrid Data Protection Agency. The Biobank complies with the security requirements set forth by the Spanish regulation on security measures for computer files containing personal data, approved by Royal Decree 994/1999 of 11 June, in fulfilment of Organic Law 15/1999 of 13 December on Personal Data Protection.2 Lastly, all technical procedures described below were designed in accordance with the legally established requirements. All biological samples are stored in such a way that long-term preservation, quantity, and quality are assured, so that they can be used for current research projects and any future projects that may be undertaken as our knowledge of kidney disease and biotechnology increases. 2) Dedicated staff and associated Committee: The Biobank’s organisational structure is shown in Figure 1. During an early step in the process, the Biobank also trained specific personnel from each centre that would be sending samples. This was done so that the process would be standardised according to the protocols established for the entire network. The Biobank continues to train researchers and support staff that are new to the Network and will be providing samples. 3) Technical procedures: In order to guarantee quality sample processing and storage, the Biobank has established a Standard Working Procedure based on a quality management system similar to UNE-EN-ISO 9001:2008. Figure 2 shows a sample processing diagram. The Biobank receives blood and urine samples. At times, depending on the active lines of research, it also receives tissue samples. These samples are sent to the Biobank by courier. Taken from whole blood samples, it routinely stores plasma aliquots and cell samples from which DNA and proteins can be extracted. If there are enough cells with good viability (more than 80%), some of them are cryopreserved. By using the Qiagen automated system, RNA is isolated from blood that was extracted with PAXgene® tubes. Vials are stored in such a way as to guarantee that all aliquots can be found in the Biobank’s locator database. For purposes of storing samples, the Biobank has storage systems set to –80 ºC and –180 ºC. Storage units are protected by a three-fold safety system: uninterruptible 29 Laura Calleros et al. Evolution of the REDinREN Biobank special article Technical and Scientific Committee Lead Researcher Dossier Authority Technical Subdirector Technical Assistant 1 Technical Assistant 2 Technical Assistant 3 Technical Assistant 4 Figure 1. Biobank organisational structure. power supply (UPS), temperature sensors with remote alarm monitoring, and a connection to the Faculty of Medicine’s generator set. In this way, biological samples are guaranteed to be completely protected in the event of an electrical or computer system failure. 4) Quality of the clinical data record system and linking data correctly with stored samples: While the biological sample collection was being created, the Biobank also made a clinical database for the Network. It covers all of the patients with samples stored in the Biobank and contains all information needed in order to make full use of the biological samples. Although the initial design allowed for a single document (Figure 3), supplementary documents may be included in the different specific studies. Patients selected to send samples to the Biobank are assigned a Biobank Number which allows entering their clinical data in the database. Patients’ personal data are not linked to clinical data, and they are not included in the same database. This archive is found online within the intranet of the Network’s webpage (www.redinren.eu). Members can gain access by using their personal passwords. Each hospital’s access is limited to the clinical data of its own patients, and the hospital providing the data is the only one to know the patient’s name and clinical history number. Every time a sample is extracted from a patient, it is given a Label Number and sent to the Biobank. Upon reaching the Biobank, the samples are coded (the samples are disassociated from identified donors, or made unidentifiable 30 by using a code to substitute or disassociate the donor’s identifying information. The code is also used to recover that information. The clinical database on the Network’s webpage records the Biobank Number and Label Number. The link between the sample and the patient is maintained in order to have the possibility of contacting the patient to obtain samples representing different moments in the progression of his or her kidney disease. In cases in which more than one sample will be extracted from the same patient, there will be multiple Label Numbers corresponding to the same Biobank Number (the opposite cannot occur.) The researcher who gains access to the sample receives only the Label Number, and never the Biobank Number. This provides additional protection for patient confidentiality throughout the entire procedure and complies with current security regulations.1-2 5) Sending and requesting samples: Meanwhile, the Biobank has finished designing both its webpage and its intranet area. Here, all Network members enjoy a range of computerised mechanisms for accessing Biobank information: forms for sending or requesting samples, the calendar to reserve a dispatch date, and a summary of the Biobank’s inventory available to research teams. Collecting, transporting, handling and dispatch of samples all take place under biosecure conditions that comply with applicable legislation (WHO regulations on the Transport of Infectious Substances).3 The Biobank guarantees the following in sample transport: sample Nefrologia 2012;32(1):28-34 Laura Calleros et al. Evolution of the REDinREN Biobank Urine in 50ml tubes with protease inhibitors special article Whole blood tubes (EDTA) 3 10ml-tubes PAXgene tubes (2.5ml blood) ® Each patient's sample arriving at the Biobank Biobank action protocol Urine centrifuged and divided into small aliquots RNA extraction (automated method) Stored at -80ºC Stored at -80ºC 3 aliquots of 4x106 cells (DNA) Stored at -80ºC 20ml of whole blood. Separate mononuclear cells (Ficoll method) 10ml of whole blood. Separated into 4 plasma aliquots (500µl) Stored at -80ºC 3 aliquots of 4x106 cells (Proteins) Stored at -80ºC Cryotubes of viable cells (8-10x106) (Proteins) Stored at -180ºC The whole blood is processed by diluting and separating peripheral mononuclear blood cells, which are subjected to Ficoll density gradient. Figure 2. Biobank action protocol. identification and traceability, a definition of the preparation conditions, handling and transport conditions required by each type of sample, and proper training for the personnel handling and preparing the samples. When transporting samples abroad, the Biobank will transmit all of the required documents in accordance with Spanish Royal Decree 65/2006 4 for exporting biological samples and sending them through customs checkpoints. purposes of the approved project; not to store, return, or transfer it to a third party; and to use the minimum quantity possible), the possibility of Biobank running an audit to verify that contractual conditions have been met, and the requirement of including Biobank’s name in publications (material and methods). Lastly, upon completion the project, researchers are required to report their final results. In addition, the Biobank’s Scientific and Technical Committee has drawn up and approved the documents needed by researchers in order to donate and/or provide Biobank samples (Figure 4). Requests for samples and/or data will be evaluated by the Biobank Scientific and Technical Committee; a Clinical Research Ethics Committee must necessarily support the study in question. Biological samples stored in the Biobank may be used in research projects undertaken by REDinREN members. If permission is granted by the Scientific and Technical Committee, they may be used in research projects with third-parties, provided that a REDinREN member is participating in the project. The provision of a sample can be accompanied by the associated clinical information from the Network’s clinical database, with the necessary guarantees for personal data protection. If provision of the sample is approved following the evaluation, a cession agreement is signed. This agreement stipulates the regulations for collecting samples, the samples for which access is provided, the lead researcher and the project in which the samples will be used, the researcher’s obligations (use for exclusive RESULTS Nefrologia 2012;32(1):28-34 Evolution and description of the current state of the Biobank 1) Infrastructures: The REDinREN Biobank is located within the Faculty of Medicine of the University of Alcalá (Madrid). Launching the biobank involved facility remodelling, purchase of a complete set of equipment and start-up of the entire laboratory. From the time it was created to the present, the REDinREN Biobank infrastructure has grown considerably. Since its inception, it has added 2 new –80 ºC vertical laboratory freezers to bring its total to 3, as well as the corresponding temperature probes, a higher-powered UPS system and 3 times as much physical capacity as was found in the original laboratory. 2) Personnel: The substantial increase in the workload arising from the greater number of samples and requests for material for a variety of projects has resulted in the need for more supporting technicians. These tasks were 31 special article Laura Calleros et al. Evolution of the REDinREN Biobank samples were added to the Biobank at the end of 2010. These include plasma and serum samples that are separated at the hospital of origin and arrive at the Biobank ready to be stored. This guarantees the preservation of a number of biomarkers that may change in the transport of whole blood samples. Lastly, based on data entered by researchers on the Website, we have a list of all pathologies for which samples are kept in the Biobank (Figure 5C). Those in white were gathered by the Nefrona project. Research projects linked to the Biobank 1) General strategy: prospective inclusion of patients meeting the following criteria: although patient inclusion criteria were originally very general, samples are currently being taken from patients with specific characteristics that are matched to each project. (Source: www.redinren.eu) Figure 3. Clinical data sheet for the REDinREN biobank 2) Specific strategies: the Biobank as a resource for specific research projects. The Biobank is currently involved in several collaborative scientific projects with Network members. Collaborative projects in the framework of the Spanish National Plan: 6 projects; Collaborative projects with a European Network: 1; research projects receiving private funding: 3; clinical trials: 1. The Biobank has also applied to be included in a European Biobank Network for dialysis patients (ERAEDTA. EURECA-m BIOBANK). DISCUSSION originally carried out by 1 person, but we currently have 4 staff members dedicated to the different phases of sample processing. Maintaining qualified personnel is a continuous training task that is essential to make the Biobank work properly. 3) Stored samples and recorded patients: The dispatch, collection, processing and storage system is currently working correctly; having received biological samples from all hospitals within the Network. As shown in Figure 5A, the Biobank stored 518 vials in 2007; this number has increased considerably, reaching 37 042 in 2010. Of the total of vials that are categorised by type (Figure 5B), we have plasma vials (30%), vials for DNA isolation (28%), and vials for protein extraction (28%). We must consider that urine vials (1%) are only collected in specific cases and that as RNA extraction with the PAXgene ® system is very expensive, it is only performed in a few selected patients (4%). Meanwhile, through Biobank’s collaborative efforts with the Nefrona project, new 32 Future prospects Our Biobank was designed to strictly comply with all applicable legislation. This will grant us access to the National Biobank Register as soon as it is created, as provided for by Law 14/2007, and the relevant Royal Decree has been issued. The register will aid us in publicising our Biobank on a national level and foster access to biological material by the scientific community. Furthermore, all of our procedures are managed with a view to obtaining top quality, and we are now beginning to launch our quality management programme so as to be certified by AENOR under ISO standard 9001:2008 in the near future. Although the Biobank was initially designed as a support platform for the REDinREN, the aim of the Network members and Biobank administrators is to place all of the possibilities contained in this biological sample storage system at the disposal of the nephrological research community. Nefrologia 2012;32(1):28-34 Laura Calleros et al. Evolution of the REDinREN Biobank special article Hospital Patient NETWORK Visit Signature I.C. Biobank Processing and storage Sample provision process (DOC. 5) (DOC. 6) Sample Extraction (DOC. 2) Data Recorded in network database Hospitals and/or research centres Request material and/or clinical data from biobank Extraction, quantification and preparation of nucleic acids and proteins Scientific and technical committee (DOC. 3) Project evaluation (DOC. 4) I.C.: inform consent; DOC. 2: sample transfer agreement with donor centre; DOC. 3: application form for samples or data from the Biobank; DOC. 4: Committee evaluation; DOC. 5: cession agreement between the Biobank and the researcher; DOC. 6: results report by the researcher. Red arrows indicate the sample transfer process to the Biobank. Green arrows indicate the process to gain access to the Biobank samples. Figure 4. Summary of the main steps for donating and gaining access to biobank samples Lastly, integrating our Biobank in a European network of biobanks with kidney patient samples may be a key step towards facilitating the export of samples to other countries This would promote collaborative efforts in international research projects. 37042 A 12582 7447 518 2007 2008 2009 2010 C B DNA Paxgene RNA Cells Urine Plasma Plasma by origin Proteins Serum by origin Underlying kidney disease Amyloidosis Glomerulonephritis Nephroangiosclerosis Diabetic nephropathy Ischaemic nephropathy Lupus nephritis Tubulo-interstitial nephritis None Other Polycystosis Vasculitis (left blank) Total 3 128 83 121 19 5 78 28 131 57 6 2 294 Total samples 2 953 A) Total of vials that the Biobank has stored over years. B) Vials by type of sample. C) Total of samples by patient pathology as entered into the Website. Data to January 2011. Figure 5. REDinREN Biobank inventory. Nefrologia 2012;32(1):28-34 33 special article ECONOMIC CONSIDERATIONS IN CREATING THE BIOBANK Most of REDinREN’s Biobank infrastructures and personnel are financed by the Biobank’s parent entity Carlos III Health Institute (REDinREN, Spanish Renal Research Network from the Carlos III Institute of Health, FEDER funds, RD6/0016/0002). With the permission of this institution, the Biobank has applied for specific grants from private organisations or projects. These include FRIAT (Íñigo Álvarez Renal Foundation of Toledo) and the Nefrona project which is supported by the S.E.N. (Spanish Society of Nephrology) and by Abbott Laboratories. On the other hand, Laura Calleros et al. Evolution of the REDinREN Biobank the cost of processing the samples requested for a specific project, whether public or private, is borne by the project itself. Acknowledgements Carlos III Health Institute: (REDinREN, Spanish Renal Research Network from the Carlos III Institute of Health, FEDER funds, RD6/0016/0002). FRIAT (Íñigo Álvarez Renal Foundation of Toledo). Reina Sofía Institute of Renal Research (IRSIN). Nefrona project, supported by the S.E.N. and Abbott laboratories. REFERENCES 1. Ley 14/2007, de 3 de julio, de Investigación Biomédica. MadridEspaña: Boletín Oficial del Estado N.º 159; 2007. p. 28826-48. 2. Ley orgánica 15/1999, de 13 de diciembre, de Protección de Datos de Carácter Personal. Madrid-España: Boletín Oficial del Estado N.º 298; 1999. p. 43088. 3. WHO/EMC/97.3. Guía para el transporte seguro de substancias infecciosas y especímenes diagnósticos. Organización Mundal de la Salud. División para la Vigilancia y el Control de Enfermedades Emergentes y Otras Enfermedades Transmisibles. Transporte seguro de Substancias Infecciosas 2005:1-15. 4. Real Decreto 65/2006, de 30 de enero, por el que establecen requisitos para la importación y exportación de muestras biológicas. Madrid-España: Boletín Oficial del Estado N.º 32; 2006. p. 4626-36. Sent to:17 Aug. 2011 | Accepted: 22 Oct. 2011 34 Nefrologia 2012;32(1):28-34 11094-I 8/2/12 12:36 Página 35 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society originals See editorial comment on page 15 Haemodialysis using high cut-off dialysers for treating acute renal failure in multiple myeloma Guillermo Martín-Reyes1, Remedios Toledo-Rojas1, Álvaro Torres-Rueda1, Eugenia Sola-Moyano1, Lourdes Blanca-Martos1, Laura Fuentes-Sánchez1, M. Dolores Martínez-Esteban1, M. José Díez-de los Ríos2, Alicia Bailén-García3, Miguel González-Molina1, Isabel García-González4 Servicio de Nefrología. Hospital Regional Universitario Carlos Haya. Málaga. Spain Laboratorio de Análisis Clínicos. Hospital Regional Universitario Carlos Haya. Málaga. Spain 3 Servicio de Hematología. Hospital Regional Universitario Carlos Haya. Málaga. Spain 4 Servicio de Anatomía Patológica. Hospital Regional Universitario Carlos Haya. Málaga. Spain 1 2 Nefrologia 2012;32(1):35-431(6):XX doi:10.3265/Nefrologia.pre2011.Nov.11094 ABSTRACT Introduction: Acute renal failure (ARF) occurs in 12%-20% of all multiple myeloma (MM) cases, and the survival of these patients depends on renal function recovery. Renal function is not recovered in 75% of dialysis-dependent patients, and their mean survival with replacement therapy is less than one year. Renal tubular disease is the most frequent cause of renal failure. It is present in more than 55% of renal failure cases and in 75% of those requiring dialysis. Rapid reduction of free light chain levels in the blood is necessary in order to recover renal function. One coadjuvant measure in treating the disease is reducing light chain levels with plasmapheresis, but its efficacy has not yet been clearly proven. Our proposal was therefore to use extended haemodialysis sessions with high cut-off dialysers (HCO-HD), obtaining a recovery rate of more than 60%. We present the progress of 6 patients with myeloma and acute renal failure who were treated with HCO-HD and the complications associated with using this type of haemodialysis. Then, we review the pros and cons of this technique. Method: Six patients diagnosed with MM and ARF requiring dialysis and with serum free light chain levels above 500mg/l were treated with 8-hour haemodialysis sessions with an HCO-HD filter. Before and after each session, serum free light chain levels were measured by nephelometry; other parameters were recorded as well. At the same time, patients underwent chemotherapy according to protocols. Results: The symptom onset times of the Correspondence: Guillermo Martín Reyes Servicio de Nefrología. Hospital Regional Universitario Carlos Haya. Almirante Enríquez, 10, Bajo D, 29017 Málaga. Spain. gmartí[email protected] 3 men and 3 women diagnosed with MM and ARF were highly variable, from 7 days to more than 1 year. We performed 90 extended sessions with HCO-HD filters, and each patient underwent between 6 and 40 sessions. Free light chain levels decreased by a mean of 65% between treatment onset and completion, except in one patient who experienced a 12.6% reduction. The mean percentage of reduction of light chain levels per session was 54.98%±17.27%. A complication occurred during 28% of the sessions. Of these complications, 48% were due to system coagulation. There were no major changes in predialysis albumin, calcium, phosphorous or magnesium levels, although lower values that were not clinically relevant were recorded in one case. Renal function was recovered in 3 patients, they are alive and dialysis-free. In biopsied cases that recovered renal function, the specimen showed tubular nephropathy only. Those patients who took longer to be diagnosed did not recover their renal function, and when biopsied, they were diagnosed with renal tubular disease and light chain deposition disease. Conclusion: We found extended haemodialysis with HCOHD filters to be a reasonable alternative in ARF caused by renal tubular disease, and achieved a recovery rate of 50% in our cases. Function recovery was influenced by the elapsed time between symptom onset and myeloma diagnosis, histological findings, promptness of starting chemotherapy, response to chemotherapy, and effectiveness of light chain extraction. In any case, further studies are needed to examine new chemotherapy agents and new direct free light chain removal techniques. Keywords: Haemodialysis, High cut-off dialysers, Acute renal failure, Multiple myeloma 35 11094-I 8/2/12 12:36 Página 36 originals Tratamiento con hemodiálisis del fracaso renal agudo en el mieloma múltiple con filtros de alto poro (high cut-off) RESUMEN Introducción: El fracaso renal agudo (FRA) en el mieloma múltiple (MM) se presenta entre el 12-20% de los casos y la supervivencia de estos pacientes depende de la recuperación de la función renal. El 75% de los pacientes dependientes de diálisis no recuperan la función renal y su supervivencia media en situación de tratamiento sustitutivo es inferior al año. La nefropatía por cilindros es la causa más frecuente de fracaso renal y acontece en más del 55% de los casos, y en el 75% de aquellos que requieren diálisis. Para facilitar la recuperación de la función renal es imprescindible la disminución rápida de los niveles en sangre de cadenas ligeras. Una medida coadyuvante al tratamiento específico de la enfermedad ha sido la reducción de estas cadenas ligeras con plasmaféresis, sin que se haya demostrado claramente su eficacia, por lo que se ha propuesto el uso de hemodiálisis largas con filtros de alto poro (HCO), consiguiendo una tasa de recuperación superior al 60%. Presentamos la evolución en seis casos de pacientes con mieloma y fracaso renal agudo que fueron tratados con dichos filtros HCO, las complicaciones con este tipo de hemodiálisis y revisamos los pros y los contras de esta técnica. Metodología: Seis pacientes diagnosticados de MM y FRA con necesidad de diálisis y niveles circulantes de cadena ligera por encima de 500 mg/l fueron tratados con hemodiálisis de 8 horas con filtro HCO. Al comienzo y al final de cada sesión se medían las cadenas ligeras séricas por nefelometría, así como otros parámetros. Al mismo tiempo los pacientes fueron tratados con quimioterapia según protocolos. Resultados: A tres hombres y tres mujeres diagnosticados de MM y FRA, con inicio de los síntomas muy variable, desde 7 días a más de un año, se les realizó 90 sesiones de hemodiálisis largas con filtros HCO con un rango de entre 6 y 40 sesiones. El porcentaje de reducción de las cadenas ligeras desde el inicio del tratamiento hasta su finalización fue el 65% de media, excepto en un paciente, que fue del 12,6%. La media del porcentaje de reducción de la cadena ligera por sesión fue de 54,98 ± 17,27%. En el 28% de las sesiones se registró alguna complicación. El 48% de las complicaciones se debieron a la coagulación del sistema. No hubo grandes cambios en los niveles de albúmina prediálisis, calcio, fósforo y magnesio, aunque en algún caso se registraron valores disminuidos que no comportaron relevancia clínica. En tres pacientes la función renal se recuperó y permanecen vivos e independientes de la diálisis. En los casos biopsiados y que recuperaron función renal, la nefropatía por cilindros fue pura. Los pacientes que tardaron más en ser diagnosticados fueron los pacientes que no recuperaron función renal, y cuando se les efectuó biopsia el diagnóstico fue de nefropatía por cilindros más enfermedad por depósitos. Conclusión: En nuestra experiencia, la hemodiálisis larga con filtros HCO es una alternativa razonable en el FRA causado por nefropatía por cilindros, alcanzando en nuestros casos una tasa de recuperación del 50%. En la recupera36 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma ción influyeron: el tiempo transcurrido desde el inicio de los síntomas al diagnóstico de mieloma, los hallazgos histológicos, la rapidez de instauración del tratamiento quimioterápico y su respuesta y la eficacia en la extracción de cadenas ligeras. En cualquier caso, son necesarios nuevos estudios con nuevos agentes quimioterápicos y las nuevas técnicas de extracción directa de cadenas ligeras. Palabras clave: Mieloma múltiple. Fracaso renal agudo. Hemodiálisis. Filtros de alto poro. INTRODUCTION Multiple myeloma (MM) is a plasma cell neoplasm that causes acute renal failure (ARF) in 12%-20% of cases, whether as an initial manifestation of the myeloma or during the course of evolution of the disease following diagnosis.1 The survival of these patients will depend on whether or not they recover renal function, not only due to the complications derived from the renal failure itself, but also from the reduced possibility of access to more effective treatments.2,3 Renal function is not recovered in 75% of dialysis-dependent patients,4-7 and their mean survival on renal replacement therapy is less than 1 year.8 Cast nephropathy is the most common cause of ARF and plays a role in over 55% of cases, and 75% of those that require dialysis.9,10 The formation of casts in the distal tubules, caused by the deposition of light chains and Tamm-Horsfall protein, is the main cause of renal failure in these patients11-14; as such, it is essential to rapidly reduce blood levels of free light chains in order to facilitate the recovery of renal function.15 Chemotherapy treatments for MM have improved greatly in the last decade, and regimens involving bortezomib, melphalan, thalidomide, and lenalidomide have improved the prognosis of these patients. 16 One coadjuvant therapy for this disease has been reducing free light chain levels using extracorporeal purifying techniques. Plasmapheresis has been used for many years to reduce the circulating levels of light chains, although the efficacy of this treatment in recovering renal function has not been clearly demonstrated. 17 This has caused researchers to search for new techniques that can effectively remove light chains and improve the recovery rate of renal function. To this end, Hutchison et al, using high cut-off (HCO) dialysers and long haemodialysis sessions (8 hours), have achieved good results in these patients, with recovery rates over 60%. 18 We present the evolution of six patients with myeloma and acute renal failure that were treated with these HCO filters, as well as the complications that arise with this type of haemodialysis, and review the pros and cons of this technique. Nefrologia 2012;32(1):35-43 11094-I 8/2/12 12:36 Página 37 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma originals MATERIAL AND METHOD We also used a Congo red stain test. Six patients diagnosed with MM and ARF requiring dialysis and with serum light chain levels >500mg/l were treated with haemodialysis using HCO 1100® (1m2) or Theralite® (2.1m2) high cut-off filters made from polyaryl ether sulfone/polyvinyl pyrrolidone (Gambro Dyalisatorem, Henchingen, Germany).19 These filters are designed to increase the permeability of substances smaller than 60kD. The only difference between them is the greater surface area of the membrane, which increases the efficacy in removing light chains and produces a greater loss of albumin. We used Theralite® (2.1m2) filters when they were commercially available. The haemodialysis sessions involved standard haemodialysis monitors, lasted for 8 hours, involved a blood flow of 200ml/min-250ml/min, and had an ultrapure dialysate flow rate of 500ml/min. Sodium heparin was applied at 3000IU to start with, and 500IU at each hour. At the end of each session, 20% albumin was administered (100cc) along with monosodium phosphate (10ml) and magnesium sulphate (10ml), following the protocol set out by Hutchison et al.20 The goal was to perform 5 sessions on consecutive days, followed by sessions on alternating days until renal function was recovered or until light chain levels fell below 500mg/l. Ultrafiltration was programmed according to the clinical needs of each patient. At the same time, patients were treated with chemotherapy regimens according to the protocols of the haematology department. Before and after each session, mean free light chain levels were measured in terms of mg/l using nephelometry (FREELITE®; The Binding Site, Birmingham, UK),21 and creatinine, albumin, phosphorous, calcium, and magnesium were measured before each session. These measurements were also taken after each session in the first patient. In cases requiring a renal biopsy, an analysis was performed using a light microscope and immunofluorescence with standard techniques, including the detection of anti-kappa and lambda light chain antibodies by immunofluorescence. RESULTS Three men and three women were admitted to the nephrology department for ARF of an unknown cause and were finally diagnosed with MM. The characteristics of the patients are summarised in Table 1. Three patients were younger than 60 years, and the other three were older than 70. The initial symptoms were bone pain in four cases, one patient sought treatment for constitutional syndrome, and the sixth patient was admitted under the diagnosis of unconfirmed pancreatitis and renal failure. The start of symptoms varied greatly, between 7 days and over a year (6th patient). In this patient, a review of the clinical history revealed that a monoclonal gammopathy and bone infarcts had been detected one year before, requiring health care, but with an incomplete analysis. The patients that took the longest time to be diagnosed were those that did not recover renal function. Three patients had kappa light chain myeloma, two had lambda IgG myeloma, and one had IgA kappa myeloma. The bone marrow was infiltrated to a varying degree by plasma cells of an anomalous phenotype that are characteristic of the disease. Four patients underwent a renal biopsy: two were diagnosed with cast nephropathy and the other two were diagnosed with cast nephropathy and light chain deposition disease. Immunofluorescence was positive in glomerular samples for light chains in two of the deposition disease cases, and in the tubules and cylinders of all four cases. Interstitial damage was mild, and tubular Table 1. Characteristics of the six patients diagnosed with multiple myeloma Patient Age Sex Syntoms Time Type of Renal Bone marrow, from start of chain biopsy % plasma cells symptoms Free light Recovery chain of renal levels mg/l function 12 600 Yes to admission 1 54 M Lumbar pain 7 days Kappa CN 72% 2 3 52 F Constitutional syndrome 79 M Shoulder pain 120 days Kappa CN + DD 15% 683 No 15 days IgA Kappa NB NB 1080 Yes 4 70 5 82 F M Side pain 60 days IgG Lambda NB 80% 13 000 No Lumbar pain 45 days IgG Lambda CN 15% 3470 Yes 6 50 F Pancreatitis, 1 year Kappa CN + DD 10% 3140 No renal failure DD: deposition disease; M: male; F: female; CN: cast nephropathy; NB: no renal or bone marrow biopsy Nefrologia 2012;32(1):35-43 37 11094-I 8/2/12 12:36 Página 38 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma originals reactivity with marked regenerative changes was similar in all cases. None of the Congo red stain tests resulted positive. One patient refused the biopsy (case 3), and the health of another (case 4) negated that possibility. Proteinuria ranged between 6680mg/dl and 570mg/dl, with no correlation observed with serum free light chain levels or histological findings. The number of days of hospitalisation, days to diagnosis, days to start of HCO-HD, as well as the number of days elapsed between hospitalisation and the start of chemotherapy are shown in Table 2. The first three patients were treated with vincristine, adriamycin, and dexamethasone, and the last three were given bortezomib and melphalan and/or prednisone, according to the protocols of the haematology department. It total, the six patients underwent 90 sessions of haemodialysis, with a range of 6-40 sessions each. The HCO-HD was interrupted due to: a) renal function recovery in three cases (cases 1, 3, and 5); b) in case 2, the start of HCO-HD was delayed several days since the patient had a large inguinal haematoma following catheterisation of the femoral vein, and a posteriori analysis showed that light chain levels had fallen below 500mg/l; c) in case 4, treatment was interrupted upon finding that the patient’s light chain levels remained above 700mg/l and anuria continued even after 7 sessions of haemodialysis; and d) in case 6, treatment was discontinued after 40 sessions of HCO-HD and three cycles of bortezomib and prednisone in the absence of a response, with light chains remaining at 2760mg/l and persistent oliguria. The percentage reduction in light chain levels from the start of treatment to the end of treatment in each patient is summarised in Figure 1, and was very high in all patients except for patient 6, who did not respond to chemotherapy treatment, with a small reduction in light chain levels (12.6%). In the other five patients, levels decreased by over 65%. In 56 sessions in which pre and post-HD light chain levels were measured, the mean percentage reduction in light chain levels was 54.98%±17.27% (Figure 2 and 3). There were no differences in the reduction of lambda and kappa light chains (kappa: 53.52%±18.6% vs lambda 57.82%±14.2%), or between results when using 1.1m2 HCO or 2.2m2 Theralite® filters (56%±19% vs 53%±15%, respectively). In 25 haemodialysis sessions (28%), several complications/events were registered. These are discussed in Table 3. On 12 occasions (48%), the complications were due to system coagulations (8 episodes in patient 6), requiring a suspension of the session on three occasions. Reduced flow and catheter obstruction were the causes of 8 other episodes (32%). Other complications were irrelevant. There were no major changes in predialysis albumin levels when using this method, although patients that were treated using the 2.1m2 Theralite® filter had a greater decrease in albumin, requiring increased doses after dialysis (cases 5 and 6). Although calcium, phosphorous, and magnesium levels were slightly reduced in several cases, this change had no clinical significance (Table 4). Other complications in the evolution of our study patients were: two infections (one resulted in a positive blood culture for Escherichia coli, the other one testing negative) and one stroke with complete recovery. In three patients, renal function was recovered (cases 1, 3, and 5), and these patients remain alive and free of dialysis. Case 1 received a bone marrow transplant and has had favourable evolution for three and a half years; case 3 has Table 2. Days to diagnosis and to treatment with HCO, chemotherapy used in the six patients diagnosed with multiple myeloma Patient Days Days to Days from No. of HCO Days on Days from in hospital diagnosis admission to sessions HCO admission to 1 36 6 15 8 12 8 VAD 2 56 16 26 6 11 22 VAD 3 19 5 6 13 12 6 VAD 4 24 3 7 9 27 9 Bortezomib- 5 57 6 13 14 28 14 Bortezomib- 6 36 8 11 40 86 14 start of HCO-HD Chemotherapy start of chemoth melphalan dexamethasone Bortezomibdexamethasone CT: chemotherapy; VAD: vincristine, adriamycin, and dexamethasone 38 Nefrologia 2012;32(1):35-43 11094-I 8/2/12 12:36 Página 39 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma 100% 90% 80% 70% 60% 50% 40% 30% 20% 10% 0% Case 1 Case 2 Case 3 Case 4 Case 5 Case 6 Figure 1. Percentage reduction in free light chain levels in the six patients treated with HCO haemodialysis at the end of treatment. been monitored for two years 8 months, and case 5 for 8 months. Of the three cases that did not recover renal function, one patient died 4 months after starting haemodialysis; in the other two cases, the patients are still alive, although they continue to depend on dialysis (case 2: 2 years, 10 months; case 6: 4 months). In the two cases that did not recover renal function and had undergone a renal biopsy, cast nephropathy was accompanied by deposition disease, whereas in the two cases that underwent a biopsy and recovered renal function, the cast nephropathy was the sole diagnosis. DISCUSSION Based on our experience, patients with MM and ARF with cast nephropathy can recover renal function through combined treatment with chemotherapy and long haemodialysis sessions with HCO filters, as occurred in at least 50% of the patients in our small study. Recent studies have demonstrated that a rapid decrease in free light chain levels with chemotherapy and extracorporeal techniques is associated with recovered renal function.15,22-24 Several studies have evaluated the efficacy of plasmapheresis in the treatment of myeloma-related renal failure. The study by Zucchelli et al,23 and more importantly, the study by Johnson24 demonstrated a better patient evolution in those cases that were treated with plasmapheresis vs those that were not; however, the small sample size, the fact that not all patients had cast nephropathy, and the use of different dialysis methods significantly limited the conclusions made in this study. Afterwards, Clark17 published a randomised, controlled study involving 97 patients, the largest study available in the literature. In this study, plasmapheresis did not achieve superior results in terms of death, dependence on dialysis, and glomerular filtration rate (<30%ml/min/1.73m2). However, Leung15 found that plasmapheresis was effective at recovering renal function (mean time: 2 months) if the renal damage was due to cast nephropathy and the level of free Nefrologia 2012;32(1):35-43 originals light chains decreased by at least 50%. In summary, the efficacy of plasmapheresis in myeloma kidney is uncertain at best, and has yet to be fully upheld as a valid treatment after the publication of only a small number of studies with few patients. The reason for this is that plasmapheresis only purifies the intravascular space (17%) and not at the quantity necessary to provide a benefit to many patients, since the reduction in light chain levels depends primarily on the level of production.5 Furthermore, plasmapheresis cannot be sustained for a very long time. Since 2007, the use of long haemodialysis sessions with membranes that are permeable to proteins has been proposed in order to increase the extraction rate of light chains up to 90% during three weeks.20 In this study, HCO filters (Gambro) with a cut-off of 45kD were superior to four different high-flux dialysers (10kD) and two super-flux dialysers (20kD) in clearing light chains (22.5-31kD). In this same study, the HCO filter was tested in 11 patients with myeloma and requiring haemodialysis who underwent different chemotherapies. Of the 11 patients diagnosed with cast nephropathy based on a renal biopsy, 5 underwent dialysis on a daily basis for one week and afterwards on alternating days; 3 recovering renal function. In the 2 patients that did not recover renal function, infections forced an interruption of the chemotherapy provided, with a consequent rebound in light chain levels. Later, these same authors reported their experience with 19 patients with renal biopsies showing cast nephropathy, 13 of which recovered renal function. Of the 6 patients in which chemotherapy had to be suspended, only one recovered renal function, which appears to suggest that the extraction of light chains using this method is only useful when accompanied by a response to chemotherapy.18 Following the first experience by Hutchison et al, we started treating patients with MM and ARF from cast nephropathy with this method of haemodialysis, with similar results to those from previous authors. In our six patients, the extraction of light chains in each long haemodialysis session with HCO filters reduced light chain levels by 53%-57%, similar to the results reported by Hutchison. At the end of the treatment, our patients had experienced decreases of over 70% over the initial values, except for patient number 6, who, despite the fact that haemodialysis was effective in reducing light chain levels, did not respond to chemotherapy and never recovered renal function, as occurred in Hutchison’s patients.18 We did not observe severe complications during haemodialysis sessions in our study, and with the replacement of lost albumin, calcium, phosphates, and magnesium, we reached acceptable levels in all these parameters. The use of 2.1m2 filters did result in a greater loss of albumin, which required increased doses to replenish albumin levels after dialysis sessions, using a double filter.25 39 11094-I 8/2/12 12:36 Página 40 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma originals Patient 2 Patient 1 160 14 000 140 12 000 120 10 000 100 8000 pre K FLC mg/L 6000 post FLC mg/L pre K FLC mg/L 80 post FLC mg/L 60 4000 40 2000 20 0 0 1 2 3 4 5 6 7 1 8 2 3 5 6 Patient 4 Patient 3 1200 14 000 1000 12 000 10 000 800 600 pre K FLC mg/L 8000 pre K FLC mg/L post FLC mg/L 6000 post FLC mg/L 400 4000 200 2000 0 0 1 2 3 4 5 6 7 8 1 9 2 3 Patient 5 3500 3000 2500 pre K FLC mg/L 1500 post FLC mg/L 1000 500 1 2 3 4 5 6 7 4 5 6 7 8 9 10 11 Patient 6 4000 0 4 8 9 10 11 12 13 14 5000 4500 4000 3500 2500 2000 1500 1000 500 0 pre K FLC mg/L post FLC mg/L 1 3 5 7 9 11 13 15 17 19 21 23 25 27 FLC: free light chains Figure 2. Serum free light chain levels before and after haemodialysis in the six patients The two infectious complications produced were not significant, and did not require halting the chemotherapy regimen. With regard to the recovery of renal function, our results are similar to Hutchison’s18: approximately 60% of patients recovered renal function using this technique.22 Several factors influenced these results: 1) the time elapsed between the start of symptoms and the diagnosis of myeloma. In our study, the three patients that did not recover renal function were those that had suffered symptoms for the longest amount of time (60 days, 120 days, and 1 year). 2) Histological findings: the 2 patients that underwent a renal biopsy and did not recover renal function had deposition disease, in addition to cast nephropathy. This situation involves a poor prognosis and short life expectancy.26 3) The 40 speed with which chemotherapy treatment is started and the response to this treatment. 4) The efficacy in removing light chains in haemodialysis using high cut-off filters. The possibility of recovery was 80% in patients whose light chain levels decreased by 60% after three weeks of treatment.18 There is still no proven evidence that this technique clearly contributes to improve survival in patients with MM and cast nephropathy, or that this type of treatment is superior to plasmapheresis, which has not demonstrated its effectiveness either. Currently, a randomised controlled study (EuLITE) is underway, involving 90 newly diagnosed patients with myeloma caused by cast nephropathy and dialysis-dependent renal failure. All patients will receive bortezomib and will be randomised to treatment with standard haemodialysis or Nefrologia 2012;32(1):35-43 11094-I 8/2/12 12:36 Página 41 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma originals Patient 2 Patient 1 60 90 80 50 70 40 60 50 30 40 30 20 20 10 10 0 1 2 3 4 5 6 7 0 8 1 2 3 35 70 30 60 25 50 20 40 15 30 10 20 5 10 0 2 3 4 5 5 6 Patient 4 Patient 3 1 4 6 7 8 0 9 1 2 3 4 Patient 5 5 6 7 8 9 10 11 Patient 6 80 70 60 50 40 30 20 10 0 1 2 3 4 5 6 7 8 9 10 11 12 13 1 14 3 5 7 9 11 13 15 17 19 21 23 25 27 Figure 3. Percentage reduction in serum free light chains in the haemodialysis sessions of all six patients HCO filter haemodialysis. The primary variable is independence from dialysis after three months of treatment.27 If results are favourable for this technique, it will then be compared with plasmapheresis. Additionally, one limitation for the use of this technique is its high economic cost. Grima28 recently presented a costeffectiveness model comparing the treatment of myeloma kidney with HCO filters vs standard haemodialysis. This study found that the treatment of myeloma kidney with long haemodialysis sessions using HCO filters could substantially improve renal recovery in patients with multiple myeloma, Nefrologia 2012;32(1):35-43 Table 3. Complications in HCO haemodialysis Complication No. % Low flow 7 28% Catheter obstruction 1 4% System coagulation 12 48% Other 5 20% Total 25 100% 41 11094-I 8/2/12 12:36 Página 42 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma originals Table 4. Maximum, minimum, mean, and standard deviation of phosphorous, calcium, magnesium, albumin, and creatinine values before haemodialysis sessions Phosphorous (mg/dl) Patient Max Min M 1 ± Calcium (mg/dl) SD Max Min M ± ± Magnesium (mg/dl) SD Max Min M ± Albumin (mg/dl) SD Max Min M ± Creatinine (mg/dl) SD Max Min M ± SD 9.4 8.9 9.1 ± 0.2 2.1 1.7 1.9 ± 0.1 3.2 2.7 2.9 ± 0.2 8.2 5.4 6.4 ± 1.0 2 3.0 1.3 2.0 ± 0.7 9.7 7.7 8.5 ± 0.7 2.3 1.7 2.0 ± 0.3 4.4 3.2 3.8 ± 0.5 5.9 2.7 3.9 ± 1.2 3 6.4 4.6 5.4 ± 0.8 10.0 7.8 9.0 ± 0.9 2.8 1.8 2.1 ± 0.4 4.5 2.6 3.5 ± 0.7 2.6 1.1 1.7 ± 0.6 4 6.7 1.9 3.7 ± 1.5 7.5 6.3 6.8 ± 0.4 2.3 2.0 2.1 ± 0.1 3.3 1.9 2.5 ± 0.4 6.5 1.7 3.2 ± 1.4 5 6.1 2.7 3.9 ± 1.1 8.0 6.1 7.0 ± 0.6 2.6 1.4 2.1 ± 0.3 2.3 1.7 2.0 ± 0.2 9.2 3.5 5.8 ± 1.8 6 6.3 2.3 3.8 ± 1.1 9.5 6.3 8.4 ± 0.7 3.8 2.0 2.6 ± 0.5 3.3 2.7 3.0 ± 0.2 7.1 2.4 4.6 ± 1.4 SD: standard deviation; Max: maximum values; M: mean; Min: minimum values compared to the standard haemodialysis. This results in an improved life expectancy and overall savings, without the need for chronic haemodialysis. The model predicts a mean survival of 19.92 months in patients treated with standard haemodialysis vs 33.9 months using the new technique, with a total savings of 6000 pounds sterling per patient. 4. 5. 6. In conclusion, the poor prognosis of patients with MM and ARF due to cast nephropathy requiring dialysis, the relationship between reduced light chain levels and the recovery of renal function suggest the potential benefit of early chemotherapy combined with a method for the extra-corporeal removal of light chains that is effective and can be maintained over time. Long haemodialysis sessions with HCO filters are a reasonable option, although new studies involving new chemotherapy agents and new direct extraction methods of light chains are needed. 7. 8. 9. 10. Conflicts of interest The authors declare potential conflicts of interest: 11. The author(s) are paid for conferences. The author(s) receive a travel bursary/allowance. 12. REFERENCES 13. 1. Kyle RA, Gertz MA, Witzig TE, Lust JA, Lacy MQ, Dispenzieri A, et al. Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin Proc 2003;78:21-33. 2. Knudsen LM, Hjorth M, Hippe E. Renal failure in multiple myeloma: reversibility and impact on prognosis. Eur J Haematol 2000;65:175-81. 3. Blade J, Fernández-Llama P, Bosch F, Montolíu J, Lens XM, Montoto S, et al. Renal failure in multiple myeloma: presenting 42 14. 15. features and predictors of outcome in 94 patients from a single institution. Arch Intern Med 1998;158:1889-93. Winearls CG. Acute myeloma kidney. Kidney Int 1995;48:134761. Magee C, Vella JP, Tormey W, Walshe JJ. Multiple myeloma and renal failure: One center’s experience. Ren Fail 1998;20:597-606. Innes A, Cuthbert RJ, Russell NH, Morgan AG, Burden RP. Intensive treatment of renal failure in patients with myeloma. Clin Lab Haematol 1994;16:149-56. Montseny JJ, Kleinknecht D, Meyrier A, Vanhille P, Simon P, Pruna A, et al. Long-term outcome according to renal histological lesions in 118 patients with monoclonal gammopathies. Nephrol Dial Transplant 1998;13:1438-45. Martín Reyes G, Valera A, Frutos MA, Ramos B, Ordóñez V, López de Novales E. Survival of myeloma patients treated with dialysis. Nefrologia 2003;23(2):131-6. Sanders PW. Renal involvement in plasma cell dyscrasias. Curr Opin Nephrol Hypertens 1993;2(2):246-52. Ivany. B. Frequency of light chain deposition nephropathy relative to renal amyloidosis and Bence Jones cast nephropathy in a necropsy study of patients with myeloma. Arch Pathol Lab Med 1990;114:986-7. Bouvet JP, Frot JC, Ducaylar A, Benlarhache C, Muller F. [Gammopathies with oligoclonal electrophoretic patterns. Incidence, immunochemical nature and association with neoplastic pathology]. Presse Med 1983;12:2511-4. Sanders PW, Herrera GA, Chen A, Booker BB, Galla JH. Differential nephrotoxicity of low molecular weight proteins including Bence Jones proteins in the perfused rat nephron in vivo. J Clin Invest 1988;82:2086-96. Leboulleux M, Lelongt B, Mougenot B, Touchard G, Makdassi R, Rocca A, et al. Protease resistance and binding of Ig light chains in myeloma-associated tubulopathies. Kidney Int 1995;48:72-9. Basnayake K, Stringer SJ, Hutchison CA, Cockwell P. The biology of immunoglobulin free light chains and kidney injury. Kidney International 2011;79:1289-301. Leung N, Gertz MA, Zeldenrust SR, Rajkumar SV, Dispenzieri A, Fervenza FC, et al. Improvement of cast nephropathy with plasma exchange depends on the diagnosis and on reduction of serum free light chains. Kidney Int 2008;73:1282-8. Nefrologia 2012;32(1):35-43 11094-I 8/2/12 12:36 Página 43 Guillermo Martín-Reyes et al. Acute renal failure in multiple myeloma 16. Dimopoulos MA, Richardson PG, Schlag R. VMP (Bortezomib, Melphalan, and Prednisone) is active and well tolerated in newly diagnosed patients with multiple myeloma with moderately impaired renal function, and results in reversal of renal impairment: cohort analysis of the phase III VISTA study. J Clin Oncol 2009;27(36):6086-93. 17. Clark, WF, Stewart, AK, Rock, GA, Sternbach M, Sutton DM, Barrett BJ, et al. Plasma exchange when myeloma presents as acute renal failure: a randomized, controlled trial. Ann Intern Med 2005;143:777. 18. Hutchison CA, Bradwell A, Cook M, Basnayake K, Basu S, Harding S, et al. Treatment of Acute Renal Failure Secondary to Multiple Myeloma with Chemotherapy and Extended High CutOff Hemodialysis. Clin J Am Soc Nephrol 2009;4(4):745-54. 19. Morgera S, Haase M, Kuss T, Vargas-Hein O, Zuckermann-Becker H, Melzar C, et al. Pilot study on the effects of high cutoff hemofiltration on the need for norepinephrine in septic patients with acute renal failure. Crit Care Med 2006;34(8):2099-104. 20. Hutchison CA, Cockwell P, Reid S, Chandler K, Mead GP, Harrison J, et al. Efficient removal of immunoglobulin free light chains by hemodialysis for multiple myeloma: In-vitro and in-vivo studies. J Am Soc Nephrol 2007;18:886-95. 21. Bradwell AR, Carr-Smith HD, Mead GP, Tang LX, Showell PJ, Drayson MT, et al. Highly sensitive automated immunoassay for immunoglobulin FLCs in serum and urine. Clin Chem 2001;47:673-80. originals 22. Hutchison CA, Cockwell P, Stringer S, Bradwell A, Cook M, Gertz MA, et al. Early reduction of serum-free light chains associates with renal recovery in myeloma kidney. J Am Soc Nephrol 2011;22:1129-36. 23. Zucchelli P, Pasquali S, Cagnoli L, Ferrari G. Controlled plasma exchange trial in acute renal failure due to multiple myeloma. Kidney Int 1988;33:1175. 24. Johnson WJ, Kyle RA, Pineda AA, O’Brien PC, Holley KE. Treatment of renal failure associated with multiple myeloma. Plasmapheresis, hemodialysis, and chemotherapy. Arch Intern Med 1990;150:863-9. 25. Hutchison CA, Harding S, Mead G, Goehl H, Storr M, Bradwell A, et al. Serum free-light chain removal by high cutoff hemodialysis: optimizing removal and supportive care. Artif Organs 2008;32(12):910-7. 26. Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman Wh, Appel GB, et al. Renal monoclonal immunoglobulin deposition disease: the disease spectrum. J Am Soc Nephrol 2001;12:1482-92. 27. Hutchison CA, Cook M, Heyne N, Weisel K, Billingham L, Bradwell A, et al. European trial of free light chain removal by extended haemodialysis in cast nephropathy (EuLITE): a randomized controlled trial. Trials 2008;9:55. 28. Grima DT, Hutchison CA. Modelled cost-effectiveness of high cutoff haemodialysis compared to standard haemodialysis in the management of myeloma kidney. Curr Med Res Opin 2011;27(2):383-91. Sent for review:19 Jul. 2011 | Accepted: 3 Nov. 2011 Nefrologia 2012;32(1):35-43 43 originals http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society Early biomarkers of acute kidney failure after heart angiography or heart surgery in patients with acute coronary syndrome or acute heart failure I. Torregrosa1, C. Montoliu2, A. Urios2, N. Elmlili2, I. Juan1, M.J. Puchades1, M.A. Solís1, R. Sanjuán3, M.L. Blasco3, C. Ramos1, P. Tomás1, J. Ribes1, A. Carratalá4, A. Miguel5 Nephrology Department. Valencia University Clinical Hospital. Spain Research Fundation of the Valencia University Clinical Hospital. INCLIVA. Spain 3 Coronary Unit. Valencia University Clinical Hospital, Spain 4 Laboratory. Valencia University Clinical Hospital. Spain 5 Nephrology Department. Valencia University Clinical Hospital. University of Valencia. Spain 1 2 Nefrologia 2012;32(1):44-52 doi:10.3265/Nefrologia.pre2011.Sep.10988 ABSTRACT Background: Acute kidney injury (AKI) is a common complication in cardiac surgery and coronary angiography, which worsens patients’ prognosis. The diagnosis is based on the increase in serum creatinine, which is delayed. It is necessary to identify and validate new biomarkers that allow for early and effective interventions. Aims: To assess the sensitivity and specificity of neutrophil gelatinase-associated lipocalin in urine (uNGAL), interleukin-18 (IL-18) in urine and cystatin C in serum for the early detection of AKI in patients with acute coronary syndrome or heart failure, and who underwent cardiac surgery or catheterization. Methods: The study included 135 patients admitted to the intensive care unit for acute coronary syndrome or heart failure due to coronary or valvular pathology and who underwent coronary angiography or cardiac bypass surgery or valvular replacement. The biomarkers were determined 12 hours after surgery and serum creatinine was monitored during the next six days for the diagnosis of AKI. Results: The area under the ROC curve (AUC) for NGAL was 0.983, and for cystatin C and IL-18 the AUCs were 0.869 and 0.727, respectively. At a cut-off of 31.9ng/ml for uNGAL the sensitivity was 100% and the specificity was 91%. Conclusions: uNGAL is an early marker of AKI in patients with acute coronary syndrome or heart failure and undergoing cardiac surgery and coronary angiography, with a higher predictive value than cystatin C or IL-18. Biomarcadores precoces de fracaso renal agudo tras angiografía coronaria o cirugía cardíaca en pacientes con síndrome coronario o fallo cardíaco agudos RESUMEN Antecedentes: El fracaso renal agudo (FRA) es una complicación frecuente tras la cirugía cardíaca y la angiografía coronaria que ensombrece el pronóstico de estos pacientes. El diagnóstico se basa en el ascenso de la creatinina sérica, que es tardío. Es necesaria la identificación y validación de nuevos biomarcadores precoces que permitan intervenciones más tempranas y eficaces. Objetivos: Evaluar la sensibilidad y especificidad de interleuquina-18 (IL-18) en orina, neutrophil gelatinase-associated lipocalin en orina (uNGAL) y cistatina C en suero para la detección precoz del FRA en una población de pacientes con síndrome coronario agudo o fallo cardíaco y sometidos a cirugía cardíaca o cateterismo. Métodos: Se incluyeron en el estudio 135 pacientes ingresados en una unidad de cuidados intensivos por síndrome coronario agudo o fallo cardíaco por patología coronaria o valvular y a los que se realizaron una angiografía cardíaca o una cirugía cardíaca de revascularización o recambio valvular. Se determinaron los biomarcadores a las 12 horas de la intervención y se monitorizó la creatinina sérica durante los siguientes seis días para el diagnóstico del FRA. Resultados: Para NGAL se obtuvo un área bajo la curva ROC (AUC) de 0,983 y para cistatina C e IL-18 de 0,869 y 0,727, respectivamente. Para un punto de corte de NGAL en orina de 31,9 ng/ml la sensibilidad fue del 100% y la especificidad del 91%. Conclusiones: uNGAL es un marcador precoz de FRA en pacientes con síndrome coronario o fallo cardíaco agudo y sometidos a cirugía cardíaca y angiografía cardíaca, con una capacidad predictiva superior a cistatina o a IL-18. Keywords: Acute kidney injury. NGAL. Interleukin-18. Cystatin C. Cardiac surgery. Coronary angiography. Acute coronary syndrome. Heart failure. Palabras clave: Fracaso renal agudo. NGAL. Interleukina18. Cistatina-C. Cirugía cardíaca. Angiografía coronaria. Síndrome coronario agudo. Fallo cardíaco. Correspondence: Isidro Torregrosa Servicio de Nefrología. Hospital Clínico Universitario de Valencia. Spain. Avda. Blasco Ibáñez, 17. 46010 Valencia. [email protected] INTRODUCTION 44 Acute kidney injury (AKI) is a common complication following cardiovascular surgery, with varying incidence rates evidenced I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients in the medical literature that depend both on the definition of AKI used and the population studied, but most estimates are close to 30%.1-3 AKI prolongs the duration of hospital stays and increases the risk of mortality by three to nine times, depending on its severity.2,3 Even small increases in creatinine levels worsen the situation for these patients.4 The mechanisms that contribute to kidney injury include exogenous and endogenous toxins, metabolic factors, ischaemia and reperfusion, neurohumoral activation, inflammation, and oxidative stress.5 The diagnosis of AKI is based on the detection of increased serum creatinine levels, which is delayed and does not properly show glomerular filtration rates in acute patients.6 However, experimental studies have demonstrated that although AKI can be prevented or treated with several different therapies, it should be started immediately after the renal damage is produced.7 The ineffectiveness of these interventions in clinical trials with humans has been attributed to significant delays in diagnosing AKI. As such, it is clear that in order to be effective, the treatment of AKI must start as early as possible. The incidence of AKI following percutaneous coronary operations can be placed between 5% and 20%, according to the definition of AKI and the population studied, and constitutes a potentially serious complication.8,9 Peak serum creatinine values tend to arise within the first 5 days following the surgery, and return to baseline values within 3 weeks (although permanent damage can be caused). As in AKI following heart surgery, increased serum creatinine values are late markers of kidney damage. To try and resolve these problems, different research groups have attempted in recent years to identify new markers for the early diagnosis and stratification of the AKI risk. Despite the effort invested and the progress made, the actual usefulness of these markers has yet to be established in the different clinical contexts of AKI.10,11 The most commonly studied markers are cystatin-C, which is not a marker of kidney damage but rather glomerular filtration rate, neutrophil gelatinase-associated lipocalin (NGAL), interleukin-18 (IL18), kidney injury molecule-1 (KIM-1), and liver fatty acidbinding protein (L-FABP), in serum or urine samples. The objective of this study is to evaluate the sensitivity and specificity of IL-18 in urine, NGAL in urine (uNGAL), and serum cystatin for the early detection of acute kidney injury in a population of patients with acute coronary syndrome or heart failure and undergoing heart surgery or catheterization. MATERIAL AND METHOD Study sample Our study included 135 patients admitted between May 2008 and December 2009 to the intensive care unit of the Valencia Nefrologia 2012;32(1):44-52 originals University Clinical Hospital for acute coronary syndrome or heart failure due to coronary or valvular pathologies, and who underwent a heart angiography with or without angioplasty and/or an implanted endoprosthesis (group 1, n=89), or cardiac surgery involving revascularization or valve replacement (group 2, n=46). The exclusion criteria were: being younger than 18 years of age, pre-existing chronic kidney disease on renal replacement therapy with dialysis or transplant, and acute renal failure due to cardiogenic shock upon hospitalisation. We also excluded patients that had consecutively undergone both angiography and surgery in order to avoid confusion. All patients were monitored prospectively starting at their inclusion in the study. We monitored creatinine levels from the day prior to the procedure to six days after, and followed the clinical evolution of each patient until they were discharged from the hospital. We also took serum and urine samples 12 hours after the procedure for assessing the biomarkers. Baseline creatinine levels were established as the value measured the day before the intervention. Blood and urine samples were centrifuged for 10 minutes at 1500g. Several 0.5ml aliquots were taken from each serum and urine sample for storage at -80ºC for later analysis. We took urine samples from 20 healthy volunteers in order to obtain normal urine NGAL values. Table 1. Clinical and demographic characteristics of patients that underwent coronary angiography Patients without AKI Patients with AKI No. patients 77 12 Age 61 (13) 73 (9) Sex (M/F) 59/18 8/4 81 (24) 58 (18)a Previous creatinine (mg/dl) 0.94 (0.22) 1.2 (0.3)ª Maximum creatinine (mg/dl) 1.0 (0.23) 2.03 (0.56)a Previous eGFR (MDRD) (ml/min/1.73m2) RIFLE (R/I/F) 6/2/1 AKI (day) 4 (3) Deaths 3.9% 25% Hospital stay (days) 10 (6) 21 (15) AKI: acute kidney injury; previous eGFR (MDRD): estimated glomerular filtration rate using MDRD before intervention. AKI (day): day of the diagnosis of AKI using creatinine levels. Values are given as mean (SD). The values for patients with AKI that were significantly different from those of patients without AKI had P-values of a P<.01; b P<.001. 45 I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients originals The following information was also collected from each patient: demographic variables and comorbidities, parameters of the surgical procedure, and complications during or after the catheter placement or heart surgery (Table 1 and Table 2). The endpoint used for evaluating our patients was the appearance of AKI, defined as creatinine levels increasing by 50% or more, in keeping with the RIFLE (Risk, Injury, Failure, Loss, End-stage renal disease) classification system.12 The secondary criteria were in-hospital mortality rates and duration of stays in the ICU and hospital. We measured IL-18 levels from urine samples using ELISA kits (Human IL-18 ELISA, Bender MedSystems), and used 50µl of each urine sample for the analysis. The detection limit for this test was 9pg/ml. Measurements of biomarkers Statistical analysis NGAL and IL-18 in urine We analysed the results using SPSS (version 17.0) and GraphPad PRISM (version 4.0) software. The KolmogorovSmirnov tests were used for verifying that the variables had a normal distribution. We compared the means of more than two variables using a one-way ANOVA test with post-hoc Bonferroni analysis, and used Student’s t-tests for comparing two means, or Mann-Whitney U-tests in the case of nonnormal distributions. We used Pearson’s correlation coefficients to analyse the relationship between the different variables. The significance level was set at P<.05. We evaluated the sensitivity and specificity of each marker using ROC (Receiver Operating Characteristic) curves. NGAL was measured from urine samples using ELISA kits (Human NGAL ELISA, Hycult biotechnology b.v.). The urine samples were diluted at 1:20 in a dilution buffer provided by the manufacturing laboratory, and 0.1ml aliquots were taken for analysis. The minimum detection level for this test is 0.4ng/ml. Table 2. Clinical and demographic characteristics of patients that underwent cardiac surgery Patients without AKI Patients with AKI No. patients 32 14 Age 70 (9) 66 (15) Sex (M/F) 22/10 12/2 (ml/min/1.73m2) 70 (32) 64 (28) Previous creatinine (mg/dl) 1.1 (0.4) 1.23 (0.5) Maximum creatinine (mg/dl) 1.2 (0.4) 2.12 (0.7)a Previous eGFR (MDRD) RIFLE (R/I/F) 9/5/0 AKI (day) 3 (1) Deaths 0 29% Type of surgery: - By-pass 19 4 - Valvular 13 9 - By-pass and valvular 1 0 Time on ECC (min) 65 (25) 91 (59) Hospital stay (days) 14 (8) 30 (24) AKI: acute kidney injury; previous eGFR (MDRD): estimated glomerular filtration rate using MDRD before intervention. ECC: time on extracorporeal circulation (minutes). AKI (day): day of the diagnosis of AKI using creatinine levels. Values are given as mean (SD). The values for patients with AKI that were significantly different from those of patients without AKI had a P-value of a P<.001. 46 Cystatin-C in serum We measured cystatin-C in serum samples using a standardised immunonephelometry analysis using a BNII nephelometer (Siemens Healthcare Diagnostics). RESULTS Clinical characteristics The clinical and demographic characteristics of our study patients are summarised in Table 1 and Table 2. A total of 26 patients developed AKI, 12 of which were in the angiography group (13%, 12/89) and 14 were from the cardiac surgery group (30%, 14/46). Six patients that underwent catheterization died, three of which had developed AKI, and four patients died from the cardiac surgery group, all of which had AKI. The patients with AKI had a longer mean hospital stay than those that did not in both groups of patients. In the group of patients that underwent catheterization, creatinine values prior to the procedure were significantly higher in patients with AKI (P<.001) (Table 1), and we did not observe any significant differences in the group of patients that underwent cardiac surgery. NGAL values in urine samples from healthy controls and patients We observed a significant difference in the values of NGAL from urine samples between patients that developed AKI and Nefrologia 2012;32(1):44-52 I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients originals those that did not, allowing us to clearly distinguish between the two groups (P<.001) (Figure 1). The mean urine level of NGAL in the control group (healthy volunteers) was 18 (5) ng/ml. We observed a significant difference in NGAL (P<.001) between patients with AKI and the control group (catheters: 96 [24]; cardiac surgery: 129 [28]) (Figure 1A, Figure 1B, and Table 3). The NGAL values in the group of patients without AKI were not significantly different from the control group (catheter: 17.4 [1.5]; cardiac surgery: 27.4 [3.7]) (Figure 1A, Figure 1B, and Table 3). Figure 1C shows how patients without AKI did not have significantly different urine NGAL values from the control group. NO AKI AKI NO AKI AKI NO AKI AKI Cystatin-C serum values The cystatin-C serum values were significantly different (P<.001) between AKI and non-AKI patients. In the catheter group, the mean value for patients with AKI was 1.22 (0.16) mg/dl, and 0.71 (0.02) mg/dl in patients without AKI. In the cardiac surgery group, the values for patients with and without AKI were 1 (0.1) mg/dl and 0.78 (0.04) mg/dl, respectively. The overall results were 1.1 (0.09) mg/dl and 0.7 (0.02) mg/dl in patients with and without AKI, respectively (Table 3). Urine IL-18 values In the case of the IL-18 inflammatory marker, AKI patients had significantly higher values than those without AKI (Table 3). In both the catheter and cardiac surgery groups, we observed a significant difference between patients with and without AKI (P<.05). This significant difference was greater when we analysed all values together (P<.001). Relationship between cystatin, IL-18, and NGAL We tested for correlations between these parameters and observed a significant correlation between NGAL in urine samples and cystatin in serum samples (r=0.311; P=.001) and between NGAL and IL-18 in urine samples (r=0.448; P<.001). A: patients undergoing catheterization; B: patients that underwent cardiac surgery; C: graph corresponding to both study groups together (catheter + cardiac surgery). Values are given as mean (SD). Asterisks (***) indicate that the difference between the two groups is significant (P<.001). NGAL: Neutrophil Gelatinase-Associated Lipocalin. . Figure 1. Measurements of NGAL in urine samples from controls, patients without acute kidney injury (no AKI), and patients with acute kidney injury (AKI) in the different study groups ROC curves for NGAL, cystatin-C, and IL-18 In order to determine whether these markers are good predictors for the appearance of AKI in patients that undergo catheterization or cardiac revascularisation surgery, we Nefrologia 2012;32(1):44-52 carried out a sensitivity/specificity analysis using ROC curves (Figure 2). Figure 2A shows the ROC curve of the three markers that were analysed in the catheter group. NGAL, cystatin-C, and IL-18 were good markers for AKI, 47 The ROC curve for patients that underwent cardiac surgery is displayed in Figure 2B. In these patients, although the incidence of AKI was higher than in the catheter group (Table 1 and Table 2), the ROC AUC were lower for all three markers (Table 4). NGAL continued to be the best predictor (AUC: 0.773) with sensitivity and specificity values of 64% and 80% respectively, considering 31.9ng/ml as the cut-off point for urine NGAL. Cystatin-C produced an AUC of 0.675, with sensitivity and specificity of 64% for the cut-off point of 0.8mg/l, and the ROC curve for IL-18 produced an AUC of 0.676, with the best cut-off point for urine IL-18 at 249pg/ml, with a sensitivity of 64% and specificity of 60% (Table 4). Figure 2C shows the ROC curve for the three markers, considering the two groups of patients studied. NGAL produced an AUC of 0.881, and was capable of detecting 80% of AKI cases with a specificity of 86%, considering the cut-off point for urine NGAL to be 31.9ng/ml. Cystatin-C produced an AUC of 0.774, detecting 75% of AKI cases with a specificity of 71% for a cut-off point of 0.8mg/l of cystatin-C in serum samples. The cut-off point for IL-18 was 201pg/ml, with sensitivity and specificity values lower than the other markers (70% and 61%, respectively) and an AUC of 0.722 (Table 4). DISCUSSION In this study we evaluated the usefulness of NGAL and IL18 in urine samples and cystatin-C in serum samples for the early (within 12 hours of the intervention) detection of AKI in a group of emergency patients in the ICU with acute coronary syndrome or heart failure, who underwent cardiac surgery or coronary angiography with or without angioplasty or endoprosthesis implantation with the following results: Sensitivity NGAL CISTATINE C IL 18 1-Specificity Sensitivity and NGAL was the best predictor, with an area under the curve (AUC) of 0.983, followed by cystatin-C (0.869) and IL-18 (0.727) (Table 4). For a cut-off point of urine NGAL at 31.9ng/ml, sensitivity was 100% and specificity was 91%. For cystatin-C, the cut-off point of 0.8mg/l in serum samples yielded a sensitivity of 89% and specificity of 76%. The cutoff point of 202pg/ml for IL-18 yielded a sensitivity of 67% and a specificity of 73%. I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients NGAL CISTATINE C IL 18 1-Specificity Sensitivity originals NGAL CISTATINE C IL 18 1-Specificity Cystatin in serum samples and IL-18 in urine samples also offer good options, although not quite as strong (AUC of 0.774 and 0.722, respectively). A: patients undergoing catheterization (AUC for NGAL: 0.983; cystatin: 0.869; IL-18: 0.727); B: patients that underwent cardiac surgery (AUC for NGAL: 0.773; cystatin: 0.675; IL-18: 0.676); C: ROC curve corresponding to both study groups together (catheter + cardiac surgery) (AUC for NGAL: 0.881; cystatin: 0.774; IL-18: 0.722). ROC: Receiver Operating Characteristic; AUC: area under the curve; NGAL: Neutrophil Gelatinase-Associated Lipocalin. Of the 46 patients that underwent cardiac surgery, 14 (30%) developed AKI, 9 of which had a status of R on the RIFLE classification system, and five of which were I. This 30% is Figure 2. ROC curves for the different markers analysed in each patient group uNGAL is useful for the early detection of AKI with an AUC of 0.881. 48 Nefrologia 2012;32(1):44-52 I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients originals Table 3. Values for the three markers analysed Catheter Cardiac surgery No AKI AKI NGAL (ng/ml) 17.4 (1.5) Cystatin-C (mg/ml) IL-18 (pg/ml) No AKI AKI 95.8 (24) 27.4 (3.7) 0.71 (0.02) 1.22 (0.16)b 196 (18) 292 (39)a b Catheterization + cardiac surgery No AKI AKI 129 (28) 20.5 (1.6) 115 (19)b 0.78 (0.04) 1.01 (0.12)b 0.73 (0.02) 1.10 (0.09)b 217 (20) 319 (38)a 203 (14) 305 (27)b b No AKI: patients that do not develop acute kidney injury; AKI: patients that do develop acute kidney injury. Cystatin-C was measured in serum samples; NGAL and IL-18 were measured in urine samples. Values are given as mean (SDM). The values for patients with AKI that were significantly different from those of patients without AKI had P-values of a P<.05; b P<.001. in accordance with the data previously published on the subject. Of the 89 patients that underwent angiography, 12 (13%) developed AKI, six of which were stage R, two were I, and one was F. These results were also within expected ranges. The delay for AKI diagnosis using creatinine took 3 (1) days in the cardiac surgery group and 4 (3) days in the angiography group. Of all the biomarker for AKI that are being studied, NGAL has probably inspired the greatest amount of interest. NGAL is a 25 kD protein that is covalently bound to neutrophil gelatinase. It is normally expressed in very low concentrations in several different tissues such as the kidneys, lungs, stomach, and colon, but is found at very high levels in kidneys with ischaemic or toxic damage.13-15 It has been most heavily researched as a marker for AKI following cardiac surgery. In a study published in The Lancet in 2005,16 Mishra et al carried out a study using paediatric patients that underwent cardiac surgery with extracorporeal circulation, and showed that NGAL is useful both in serum and urine samples for the early (in the first few hours following the procedure) detection of patients that will develop AKI in the coming days, with extremely high sensitivity and specificity levels. The usefulness of NGAL (in blood or urine samples) for the early detection of AKI following cardiac surgery was later confirmed both in children17,18 and adults,19-24 although the results are more varied and are clearly worse in adults. In other studies, however, NGAL was predictive for AKI in urine samples, but not in serum,25,26 and some studies have even produced negative results from using urine NGAL.27 Probably, the reason for the lower specificity observed in adults than in children is related to the existence of other associated conditions. For instance, it has been shown that NGAL values are higher in hypertensive28 and diabetic29 patients, and that they also increase with age.30 Sepsis can also affect NGAL values.31 It has also been shown that the relationship between NGAL and AKI after cardiac surgery varies according to baseline glomerular filtration rate, and the predictive capacity of NGAL is lost in patients with an estimated glomerular filtration rate (eGFR) below 60ml/min.32 Although the strength of the results are variable in both urine and blood tests, it appears that urine samples Table 4. Diagnostic capacity of the markers analysed for the detection of acute kidney injury in the different study groups N-GAL Study group Cystatin C IL-18 Cut-off pointa AUC Cut-off pointa AUC Cut-off pointa AUC (S-s)b (95% CI)c (S-s)b (95% CI) (S-s)b (95% CI) Angiography Cardiac surgery Angiography + cardiac surgery 31.9 0.983 0.8 0.869 202 0.727 (100-91) (0.954-1.012) (89-76) (0.676-1.06) (67-73) (0.566-0.888) 31.9 0.773 0.8 0.675 249 0.676 (64-80) (0.580-0.96) (64-64) (0.46-0.88) (64-60) (0.49-0.86) 31.9 0.881 0.8 0.774 201 0.722 (80-86) (0.784-0.977) (75-71) (0.626-0.92) (70-61) (0.609-0.835) Cut-off points for each marker are expressed in ng/ml for NGAL, mg/l for cystatin-C, and pg/ml for IL-18. S-s: sensitivity (%) specificity (%), respectively, for the cut-off point indicated. AUC: area under the ROC curve. c 95% CI: 95% confidence interval a b Nefrologia 2012;32(1):44-52 49 originals I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients are generally superior.25,26 The best time to obtain and analyse the samples has not been defined yet. 2. Whether blood or urine samples should be used, and the optimal moment for obtaining the sample. NGAL has also been shown to be useful for the early diagnosis of AKI following coronary angiography,33-38 although fewer studies have been published in support, and the predictive power of this analysis, the cut-off point, and the optimal time for measurement are not well established. 3. Extrinsic factors that may influence the results and viability of the measurements in different clinical contexts. In our study, the results from the overall group of patients were very positive for the measurement of uNGAL 12 hours after the procedure, with an AUC of 0.881. In the cardiac surgery group, AUC was 0.773, somewhat higher than the results published in other studies with adult cardiac surgery patients (0.62-0.72).19-25 The AUC for patients that underwent coronary angiography was higher too (0.983). The cut-off point for better sensitivity and specificity in the overall group was 31.9ng/ml, with a sensitivity of 80% and a specificity of 86%. Urine IL-18 has also been studied as a possible early marker for the detection of AKI in patients that undergo cardiac surgery, yielding both positive39 and negative40 results, as well as following coronary angiography,36 although this measurement has inspired less interest as a possible biomarker for AKI. In contrast to NGAL and IL-18, cystatin is not a marker of kidney damage, but rather glomerular filtration rate. CystatinC is a small endogenous protein that is freely filtered in the glomerulus, and is reabsorbed and catabolised in the proximal tubule, such that only a small quantity is excreted in the urine. Its usefulness as a possible substitute for creatinine as a method of measuring glomerular filtration rate has been examined in recent years, both in chronic and acute kidney disease, because of its lower dependence on muscle mass. However, its value does depend on other variables such as age, sex, race, and the presence of diabetes or inflammation.41 The results using this measure are also variable in patients that undergo cardiac surgery, with positive,21 modest,42 and negative results,26 although in this latter study, urine cystatin was capable of predicting the appearance of AKI, compared to serum samples. The published studies with cardiac catheters are also inconsistent, with varying and occasionally contrasting results.35, 43-45 In our study, the AUC for cystatin was 0.774 in all patients, 0.869 in the coronary angiography group, and 0.675 in the cardiac surgery group. The results for IL-18 were quite similar (Table 4). Therefore, the search for the ideal biological marker for the early detection of AKI still has the following important questions pending: 1. The best biomarker (or combination of biomarkers) and cut-off point that offers optimal sensitivity and specificity. 50 4. The prognostic power in addition to the diagnostic capacity. In this study, we have attempted to contribute a response to some of these questions. This study differs from those previously published by including both cardiac surgery and catheter patients, as well as the fact that all patients were admitted to intensive care units for coronary syndrome or acute heart failure, making them unstable patients. This is important because we need to know how these biomarkers work in large groups of the population with different characteristics, not just in homogeneous groups, and also because of the need to observe how other factors influence these measurements. We took all of our samples 12 hours after the procedure. We were interested in testing the usefulness of samples at this time since it is the simplest option for collecting and processing samples in a normal clinical context (if the procedure takes place in the afternoon, the samples would be taken first thing the next morning). We decided not to take samples for the biomarkers before the procedure, since the ultimate objective is to have a reliable marker in a specific situation, regardless of baseline values. For instance, when troponin or D-dimer are measured in clinical practice, it is without a baseline value for comparison. In any case, NGAL was measured in a population of healthy individuals with normal renal function in order to provide a reference value for comparison with the study groups. The results from the markers in urine samples were not adjusted to urinary creatinine levels since, although some authors do defend this normalisation, the majority of studies have preferred not to do so because the evidence is not clear. We preferred to measure NGAL in urine samples over serum samples since, although currently there are rapid methods for determining NGAL in blood samples with a good correlation with the results obtained in blood samples by ELISA,16 the results from previously published studies show a better yield in urine samples. In conclusion, urine NGAL taken 12 hours after the procedure is a good marker for the early detection of AKI in adult patients with acute heart failure or coronary syndrome that undergo cardiac surgery or coronary angiography, with a predictive power superior to cystatin and IL-18. Nefrologia 2012;32(1):44-52 I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. REFERENCES 1. Rosner MH, Okusa MD. Acute kidney injury associated with cardiac surgery. Clin J Am Soc Nephrol 2006;1:19-32. 2. Del Duca D, Iqbal S, Rahme E, Goldberg P, De Varennes B. Renal failure after cardiac surgery: timing of cardiac catheterization and other perioperative risk factors. Ann Thorac Surg 2007;84:1264-71. 3. Karkouti K, Wijeysundera DN, Yau TM, Callum JL, Cheng DC. Acute kidney injury after cardiac surgery: focus on modifiable risk factors. Circulation 2009;119:495-502. 4. Lassnigg A, Schmidlin D, Mouhieddine M. Minimal changes of serum creatinine predict prognosis in patients after cardiothoracic surgery: a prospective cohort study. J Am Soc Nephrol 2004;15:1597-605. 5. Bellomo R, Auriemma S, Fabbri A, D´Onofrio A, Katz N. The pathophysiology of cardiac surgery-associated acute kidney injury (CSA-AKI). Int J Artif Organs 2008;31:166-78. 6. Stevens LA, Coresh J, Greene T, Levey AS. Assesing kidney function. Measured and estimated glomerular filtration rate. N Engl J Med 2006;354:2473-83. 7. Devarajan P. Update on mechanisms of ischemic acute kidney injury. J Am Soc Nephrol 2006;17:1503-20. 8. McCullough PA. Contrast-induced acute kidney injury. J Am Coll Cardiol 2008;51:1419-28. 9. Marenzi G, De Metrio M, Rubino M, Lauri G, Cavallero A. Acute hyperglycemia and contrast-induced nephropathy in primary percutaneous coronary intervention. Am Heart J 2010;160:1170-7. 10. Coca SG, Yalavarthy R, Concato J, Parikh CR. Biomarkers for the diagnosis and risk stratification of acute kidney injury. Kidney Int 2008;73:1008-16. 11. Malyszko J. Biomarkers of acute kidney injury in different clinical settings: a time to change the paradigm? Kidney Bllod Press Res 2010;33:368-82. 12. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P, and the ADQI Workgroup. Acute renal failure-definition, outcome measures, animal models, fluid therapy and information technology needs: the second international consensus conference of the acute dialysis quality initiative (ADQI) group. Crit Care 2004;8:R204-R212. 13. Supavekin S, Zhang W, Kucherlapti R, Kaskel FJ, Moore LC, Devarajan P. Differential gene expression following early renal ischemia/reperfusion. Kidney Int 2003;63:1714-24. 14. Mishra J, Ma Q, Prada A, Mitsnefes M, Zahedi K. Identification of neutrophil gelatinase-associated lipocalin as a novel early urinary biomarker for ischemic renal injury. J Am Soc Nephrol 2003;14:2534-43. 15. Mishra J, Mori K, Ma Q, Kelly C, Barasch J, Devarajan P. Neutrophil gelatinase-associated lipocalin: A novel early urinary biomarker for cisplatin nephrotoxicity. Am J Nephrol 2004;24:307-15. 16. Mishra J, Dent C, Tarabishi R, Mitsnefes MM, Ma Q. Neutrophil gelatinase-associated lipocalin (NGAL) as a biomarker for acute renal injury after cardiac surgery. Lancet 2005;365:1231-8. 17. Dent C, Ma Q, Dastrala S, Bennett M, Mitsnefes MM. Plasma neuNefrologia 2012;32(1):44-52 originals trophil gelatinase-associated lipocalin predicts acute kidney injury, morbidity and mortality after pediatric cardiac surgery: a prospective uncontrolled cohort study. Crit Care 2007;11:R127. 18. Bennett M, Dent CL, Ma Q, Dastrala S, Grenier F. Urine NGAL predicts severity of acute kidney injury after cardiac surgery: a prospective study. Clin J Am Soc Nephrol 2008;3:665-73. 19. Wagener G, Gubitosa G, Wang S, Borregaard N, Kim M. Urinary neutrophil gelatinase-associated lipocalin and Acute Kidney Injury after cardiac surgery. Am J Kidney Dis 2008;52:425-33. 20. Xin C, Yulong X, Yu C, Changchun C, Feng Z. Urine neutrophil gelatinase-associated lipocalin and interleukin-18 predict acute kidney injury after cardiac surgery. Ren Fail 2008;30:904-13. 21. Haase-Fielitz A, Bellomo R, Devarajan P, Story D, Matalanis G. Novel and conventional serum biomarkers predicting acute kidney injury in adult cardiac surgery-A prospective cohort study. Crit Care Med 2009;37:553-60. 22. Tuladhar SM, Püntmann VO, Soni M, Punjabi PP, Bogle RG. Rapid detection of acute kidney injury by plasma and urinary neutrophil gelatinase-associated lipocalin after cardiopulmonary bypass. J Cardiovasc Pharmacol 2009;53:261-6. 23. Han WK, Wagener G, Zhu Y, Wang S, Lee HT. Urinary biomarkers in the early detection of acute kidney injury after cardiac surgery. Clin J Am Soc Nephrol 2009;4:873-82. 24. McIlroy DR, Wagener G, Lee HT. Neutrophil gelatinase-associated lipocalin and acute kidney injury after cardiac surgery: the effect of baseline renal function on diagnostic performance. Clin J Am Soc Nephrol 2010;5:211-9. 25. Xin C, Yulong X, Yu C, Changchun C, Feng Z, Xinwei M. Urine neutrophil gelatinase-associated lipocalin and interleukin-18 predict acute kidney injury after cardiac surgery. Ren Fail 2008;30:904-13. 26. Koyner JL, Bennett MR, Worcester EM, Ma Q, Raman J, Jeevanandam V, et al. Urinary cystatin C as an early biomarker of acute kidney injury following adult cardiothoracic surgery. Kidney Int 2008;74:1059-69. 27. Liangos O, Tighiouart H, Perianayagam MC, Kolyada A, Han WK. Comparative analysis of urinary biomarkers for early detection of acute kidney injury following cardiopulmonary bypass. Biomarkers 2009;14:423-31. 28. Malyszko J, Bachorzewska-Gajewska H, Malyszko JS, Pawlak K, Dobrzycki S. Serum neutrophil gelatinase-associated lipocalin as a marker of renal function in hypertensive and normotensive patients with coronary artery disease. Nephrology (Carlton) 2008;13:153-6. 29. Malyszko J, Bachorzewska-Gajewska H, Poniatowski B, Malyszko JS, Dobrzycki S. Urinary and serum biomarkers after cardiac catheterization in diabetic patients with stable angina and normal serum creatinine. Ren Fail 2009;31:910-9. 30. Przybylowski P, Malyszko j, Malyszko JS. Kidney function assessed by eGFR, cystatin C and NGAL (neutrophil gelatinase-associated lipocalin) in relation to age in heart allograft recipients. Med Sci Monit 2010;16:440-4. 31. Martensson J, Bell M, Oldner A, Xu S, Venge P, Martling CR. Neutrophil gelatinase-associated lipocalin in adult septic patients with and without acute kidney injury. Int Care Med 2010;36:1333-40. 32. Koyner JL, Vaidya VS, Bennett MR, Ma Q, Worcester E. Urinary biomarkers in the clinical prognosis and early detection of acute kidney injury. Clin J Am Soc Nephrol 2010;5:2154-65. 51 I. Torregrosa et al. Biomarkers of AKI in acute cardiac patients originals 33. Hirsch R, Dent C, Pfriem H, Allen J, Beekman RH. NGAL is an early predictive biomarker of contrast-induced nephropathy in children. Pediatr Nephrol 2007;22:2089-95. 34. Ling W, Zhaohui N, Ben H, Levi G, Jianping L. Urinary IL-18 and NGAL as early predictive biomarkers in contrast-induced nephropathy after coronary angiography. Nephron Clin Pract 2008;108:176-81. 35. Bachorzewska-Gajewska H, Malyszko J, Sitniewska E, Malyszko JS, Pawlak K. Could neutrophil gelatinase-associated lipocalin and cystatin C predict the development of contrast-induced nephropathy after percutaneous coronary interventions in patients with stable angina and normal serum creatinine values? Kidney Blood Press Res 2007;30:408-15. 36. Ling W, Zhaohui N, Ben H, Leyi G, Jianping L. Urinary IL-18 and NGAL as early predictive biomarkers in contrast-induced nephropathy after coronary angiography. Nephron Clin Pract 2008;108:176-81. 37. Bachorzewska-Gajewska H, Poniatowski B, Dobrycki S. NGAL (neutrophil gelatinase-associated lipocalin) and L-FABP after percutaneous coronary interventions due to unstable angina in patients with normal serum creatinine. Adv Med Sci 2009;54:221-4. 38. Malyszko J, Bachorzewska-Gajewska H, Poniatowski B, Malyszko JS, Dobrycki S. Urinary and serum biomarkers after cardiac cathe- 39. 40. 41. 42. 43. 44. 45. terization in diabetic patients with stable angina and without severe chronic kidney disease. Ren Fail 2009;31:910-9. Parikh CR, Mishra J, Thiessen-Philbrook H, Dursun B, Ma Q. Urinary IL-18 is an early predictive biomarker of acute kidney injury after cardiac surgery. Kidney Int 2006;70:199-203. Haase M, Bellomo R, Story D, Davenport P, Haase-Fielitz A. Urinary interleukin-18 does not predict acute kidney injury after adult cardiac surgery: a prospective observational cohort study. Critical Care 2008;12:R96. Stevens LA, Schmid CH, Greene T, Li I, Beck GJ. Factors other than glomerular filtration rate affect serum cystatin C levels. Kidney Int 2009;75:652-60. Wald R, Liangos O, Perianayagam MC, Kolyada A, Herget-Rosenthal S. Plasma cystatin C and acute kidney injury after cardiopulmonary bypass. Clin J Am Soc Nephrol 2010;5:1373-9. Rickli H, Benou K, Ammann P, Fehr T, Brunner-La Rocca HP. Time course of serial cystatin C levels in comparison with serum creatinine after application of radiocontrast media. Clin Nephrol 2004;61:98102. Briguori C, Visconti G, Rivera NV, Focaccio A, Golia B. Cystatin C and contrast-induced acute kidney injury. Circulation 2010;121:2117-22. Merkle M, Sauter M, Argirov M, Wörnle M. Cystatin C and creatinine as markers for radio contrast-induced nephropathy in patients treated with N-Acetylcysteine. Ren Fail 2010;32:85-90. Sent for review: 11 Jun. 2011 | Accepted: 21 Sep. 2011 52 Nefrologia 2012;32(1):44-52 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society originals The calculation of baseline serum creatinine overestimates the diagnosis of acute kidney injury in patients undergoing cardiac surgery A.M. Candela-Toha1, M. Recio-Vázquez2, A. Delgado-Montero2, J.M. del Rey3, A. Muriel4, F. Liaño5, T. Tenorio6 Anaesthesiology and Resuscitation Department. Ramón y Cajal University Hospital. Ramón y Cajal Institute of Health Research (IRYCIS). Acute Kidney Failure Research Consortium (CIFRA). Madrid, Spain 2 Cardiology Department. Ramón y Cajal University Hospital. Madrid, Spain 3 Biochemistry Department. Ramón y Cajal University Hospital. Madrid, Spain 4 Biostatisitics Unit. IRYCIS. Networked Consortium of Biomedical Research on Epidemiology and Public Health (CIBERESP). Ramón y Cajal University Hospital. Madrid, Spain 5 Nephrology Department. Ramón y Cajal University Hospital. IRYCIS. CIFRA. Medicine Department, University of Alcalá. Madrid, Spain 6 Nephrology Department. Ramón y Cajal University Hospital. CIFRA. Madrid, Spain 1 Nefrologia 2012;32(1):53-8:XX doi:10.3265/Nefrologia.pre2011.Oct.11102 ABSTRACT Introduction and objectives: The current definition and classification of acute kidney injury is based on consensus criteria (RIFLE and AKIN systems). Creatinine is the most commonly used of the recommended parameters (creatinine, glomerular filtration rate and diuresis). If the baseline value is not known, it can be calculated based on the simplified MDRD equation, assuming a filtration rate of 75ml/min/1.73m2 for the calculation. The aim of this study was to evaluate the diagnostic impact of using estimated baseline creatinine compared to the actual value measured in patients undergoing cardiac surgery. Methods: Analysis of patients undergoing major cardiac surgery, who were prospectively included in a database. The maximum RIFLE stage reached was calculated for each patient using the measured and estimated baseline creatinine levels. The impact on the diagnosis was analysed using intraclass correlation coefficients, concordance analysis and Bland-Altman plots. Results: The incidence of postoperative acute kidney injury in 2103 cases between 2002 and 2007 was 29.1%, according to estimated creatinine (14.3% with the measure). This represents an overestimation of 104%, with an intraclass correlation Correspondence: A.M. Candela-Toha Servicio de Anestesia y Reanimación. Hospital Universitario Ramón y Cajal. Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS). Consorcio de Investigación del Fracaso Renal Agudo. Ctra. de Colmenar, km 9,1. 28034 Madrid. Spain. [email protected] of 0.12. By excluding patients with known chronic kidney disease (glomerular filtration rate [<60ml/min/1.73m2), both the overestimation (2.4%) and the correlation (0.57) improved. Conclusions: The calculation of baseline creatinine using the MDRD equation overestimates the incidence of acute kidney injury after cardiac surgery, and is an inadequate method for detection when the baseline value is unknown. Keywords: Cardiac surgery. Acute renal failure. Acute kidney injury. Serum creatinine. RIFLE. El cálculo de la creatinina sérica basal sobrestima el diagnóstico de alteración renal aguda en pacientes operados de cirugía cardíaca RESUMEN Introducción y objetivo: La definición y clasificación actual de insuficiencia renal aguda se basa en criterios de consenso (sistemas RIFLE y AKIN). De los parámetros recomendados (creatinina, tasa de filtración glomerular y diuresis), la creatinina es el más empleado. En ausencia de valor basal conocido se recomienda su estimación a partir de la ecuación MDRD simplificada, asumiendo en el cálculo una tasa de filtración de 75 ml/min/1,73 m2. El objetivo del presente trabajo fue evaluar la repercusión diagnóstica del empleo de la creatinina basal estimada frente al valor real medido en pacientes operados de cirugía cardíaca. Métodos: Análi53 originals sis de pacientes operados de cirugía cardíaca mayor incluidos de forma prospectiva en una base de datos. Para cada paciente se calculó el estadio RIFLE máximo alcanzado usando la creatinina basal medida y la estimada. Se analizó la repercusión sobre el diagnóstico mediante coeficientes de correlación intraclase, análisis de concordancia y gráficas de Bland y Altman. Resultados: La incidencia de insuficiencia renal aguda postoperatoria en 2.103 casos operados entre 2002 y 2007 fue del 29,1% al utilizar la creatinina estimada (14,3% con la medida). Esto supone una sobrestimación del 104%, y la correlación intraclase es de 0,12. Excluyendo a los pacientes con insuficiencia renal crónica conocida (tasa de filtrado glomerular [TFG] <60 ml/min/1,73 m2), tanto la sobrestimación (2,4%) como la correlación (0,57) mejoraron. Conclusiones: El cálculo de la creatinina basal a partir de la ecuación MDRD sobrestima la incidencia de insuficiencia renal aguda tras la cirugía cardíaca, y es un método inadecuado para su detección cuando el valor basal se desconoce. Palabras clave: Cirugía cardíaca. Insuficiencia renal aguda. Alteración renal aguda. Creatinina sérica. RIFLE. A.M. Candela-Toha et al. RIFLE-AKIN in cardiac sugery and perhaps for non-scientific reasons,8 the RIFLE classification is very popular. The number of publications exceeds 150, and a fairly recent systematic review included 24 studies covering more than 70 000 patients.9 Among the problems associated with this classification is the diagnosis and classification of patients whose baseline serum creatinine is unknown. This occurs quite often in patients with an altered SCr value who are admitted to critical care units for acute problems (e.g., septic shock, acute coronary syndrome and heart failure). The ADQI recommendation is to assume that patients with a previously unknown renal function, with no known kidney disease, have a GFR of 75100ml/min/1.73m2, and then to back-calculate the SCr value using the simplified MDRD equation.10 This method has been used in several epidemiological studies despite its lack of validation.11,12 The aim of this study is twofold: 1) To assess the agreement between estimated baseline SCr (est SCr) against the known baseline SCr in patients undergoing cardiac surgery, and 2) To analyse the impact of differences in baseline SCr on AKI frequency and severity for those episodes observed in cardiac surgery. INTRODUCTION MATERIAL AND METHODS Acute kidney injury (AKI) is a common complication following cardiac surgery. Its appearance leads to a significant increase in both morbid-mortality and hospital stay. AKI frequency varies depending on the definition used. When the most severe forms, those requiring renal replacement therapy (RRT), are considered the range is 2%-5%.1 In recent years there has been a consensus in new definitions for this syndrome and a change in the nomenclature.2,3 The English term acute renal failure, traditionally translated into Spanish as insuficiencia or fracaso renal agudo has changed to acute kidney injury to broaden the spectrum of the syndrome and include milder cases. Liaño et al. suggested that AKI should be coined as alteración renal aguda (ARA) in Spanish.4 Since Lassnig’s study on patients undergoing cardiac surgery, it is known that even slight increases (0.3mg/dl) in serum creatinine (SCr) with respect to baseline have an impact on postoperative morbidity and mortality.5,6 These results have been confirmed in hospitalised patients.7 In 2004, the Acute Dialysis Quality Initiative (ADQI) proposed the use of the RIFLE system (Risk, Injury, Failure, Loss and End stage renal disease) with 3 diagnostic (R, I and F) and 2 prognostic (L and E) categories. The diagnosis is made based on SCr increases compared to baseline, decreases in the glomerular filtration rate (GFR), or absolute decreases in diuresis.2 Patients are diagnosed and classified using the criteria that placed them in the most severe stage. These criteria were reviewed in 2007 and modified by the Acute Kidney Injury Network (AKIN), this latter classification being recognised as the current one.3 However, 54 Perioperative data from cardiac surgery patients operated on in our hospital were compiled prospectively in a database kept by the Anaesthesiology Department. We used data from patients operated on between 2002 and 2007. All patients older than 14 years who underwent major cardiac surgery with or without extracorporeal circulation were included. Patients were excluded if they were on dialysis prior to surgery, were kidney transplant recipients, had died within 24 hours of surgery or if they underwent second major surgery during the same hospitalisation period. The measured baseline SCr (act SCr) was considered as the last SCr measurement before surgery, usually within 24 hours for patients undergoing elective surgery. The kinetic Jaffe method was used. The GFR was estimated from the simplified MDRD formula using four variables (SCr, age, sex and race). To calculate baseline serum creatinine (est SCr) from this equation, considering a GFR of 75ml/min/1.73m2 (the lower normal limit), the ADQI recommendations were followed by using the following formula: Estimated SCr=(75/[186x(age-0,203)x(0.742 for women)x(1.21 for black people)])-0,887 Patients were assigned the highest stage by the RIFLE classification from SCr determinations during the first week after surgery. The criterion based on percentage GFR decreases was not used, due to its lack of linearity Nefrologia 2012;32(1):53-8 A.M. Candela-Toha et al. RIFLE-AKIN in cardiac sugery and consistency when compared with increases in SCr.13 The diuresis criterion was also not used due to lack of data. The agreement between act SCr and est SCr was assessed by the intraclass correlation coefficient 14 and Bland and Altman plot. 15 The bias, the overall mean of the differences between act SCr and est SCr, was also calculated; as well as precision, defined as the bias standard deviation. The estimated error was calculated as the percentage of patients diagnosed with AKI using est SCr (estAKI) with respect to those diagnosed from act SCr (actAKI), using the formula below: originals Table 1. Main demographic variables, comorbidity, surgical variables, postoperative variables and hospital stay in the studied cohort (n=2103) Age, years, mean (SD) 66.1 (11.3) Females, % 40.7 Baseline SCr observed, mg/dl, mean (SD) 1.12 (0.38) eGFR, ml/min/1.73m , mean (SD) 68.26 (19.9) 2 - Normal: GFR> _90ml/min/1.73m (%) 221 (10.5) - Slightly impaired: GFR 60-89ml/min/1.73m2 (%) 1187 (56.4) 2 - Moderately impaired: GFR 30-59ml/min/1.73m2 (%) (estAKI – actAKI)/actAKI 46 (2.2) - End stage renal disease: GFR<15ml/min/1.73m (%) 4 (0.2) 2 A positive value showed an overestimation while a negative one implied underestimation. An initial analysis of the global sample and a second analysis, excluding patients with stage 3 or higher chronic kidney disease (CKD) according to the National Kidney Foundation K/DOQI classification, were performed. 16 The mean and standard deviation or median and interquartile range, depending on the distribution of the quantitative variables studied, were used as measures of central tendency and dispersion. A statistical analysis of the data and plots was performed using SPSS version 18.0. RESULTS A total 2167 operations were performed on 2131 patients during the study period. Those patients excluded were: 31 patients (32 surgeries) for undergoing preoperative dialysis or being kidney transplant recipients; 27 who died soon after surgery (<24 hours); and 5 for having a second surgery during the same hospitalisation period. The final sample consisted of 2103 operations on 2067 patients, and the main features are shown in Table 1. 645 (30.7) - Severely impaired: GFR 15-29ml/min/1.73m (%) 2 Hypertension, % 53.6 CHF History, % 22.1 COPD, % 6.4 Diabetes, % - With medical treatment 19.6 - Insulin-dependent 7.7 Previous cardiac surgery, % 11.7 Type of surgery, % - Isolated CABG 34.4 - Valvular 47.7 - Combined (CABG and valvular) 10.2 - Aortic 4.5 - Others 3.2 Off-pump ECC, % 10 Ischaemia time, min, mean (SD) 59.4 (33.5) Perfusion time, min, mean (SD) 81.5 (43.2) Mechanical ventilation, h, median (P25-P75) Need for postoperative RRT, % 9 (7-20) 3.4 Hospital stay, days, median (P25-P75) Table 2 shows the number and percentage of patients who reached each RIFLE stage during the first week after surgery, according to the type of baseline SCr used, either actual or estimated; the correlation between both; and the bias and precision. The global overestimation rate was 104% (29.1% vs 14.6%). The diagnosis of AKI whith est SCr showed a false positive rate of 14.8%. After excluding patients with a GFR<60ml/min/1.73m2, the correlation was better. The overestimation decreased to 2.4% with a false positive rate of 0.28%. Figure 1 shows the correlation between est SCr and act SCr via Bland and Altman plots. The point cloud arrangement indicates that the degree of agreement decreases when the baseline SCr increases. Graphically, there is greater agreement between both creatinine values after excluding patients with any CKD stage prior to surgery (Figure 2). Nefrologia 2012;32(1):53-8 - In ICU - Outside ICU 3 (2-5) 10 (7-15) CHF: chronic heart failure;COPD: chronic obstructive pulmonary disease; est GFR: estimated glomerular filtration rate; GFR: glomerular filtration rate; ICU intensive care unit; CABG: coronary artery bypass graft; RRT: renal replacement therapy; SCr: serum creatinine; SD: standard deviation. DISCUSSION This study shows that the recommendation to calculate baseline SCr from the MDRD-4 equation assuming a GFR of 75ml/min/1.73m2 overestimates the incidence of AKI in 55 A.M. Candela-Toha et al. RIFLE-AKIN in cardiac sugery originals Table 2. Renal function stratified by RIFLE category RIFLE stage Act SCr Est SCr sepsis, trauma or cardiac arrest. Our study was the first to focus on postoperative cardiac surgery patients, therefore it is not possible to make direct comparisons with other studies. Overall cohort (n=2103) None 1803 (85.7%) 1493 (71%) R 178 (8.5%) 319 (15.2%) I 77 (3.7%) 191 (9.1%) F 45 (2.1%) 100 (4.8%) Any criteria 300 (14.3%) 610 (29.1%) Total 2103 (100%) 2103 (100%) Correlation 0.605 Bias (mg/dl) 0.16 Precision (mg/dl) 0.38 Patients with eGFR >_60ml/min/1.73m2 (n=1408) None 1247 (88.6%) 1243 (88.3%) R 103 (7.3%) 103 (7.3%) I 43 (3.1%) 51 (3.6%) F 15 (1.1%) 11 (0.8%) Any criteria 161 (11.5%) 165 (11.7%) Total 1408 (100%) 1408 (100%) Correlation 0.845 Bias (mg/dl) -0.02 Precision (mg/dl) 0.13 Both the prevalence of CKD or preoperative renal dysfunction and the incidence of AKI have a clear impact on the percentage of overestimation. To some extent, the first may be influenced by age (generally higher in surgical patients) and other cardiovascular risk factors (particularly hypertension and diabetes), which were significantly present in our sample. It is worth noting that the best definition of preoperative renal dysfunction for patients undergoing cardiac surgery was established by Wijeysundera,21 as creatinine clearance (CrCl) lower than 60ml/min. The odds ratio for developing severe AKI (with a need for RRT) in a random sample of 2000 patients operated on over a period of 16 months was 5 (confidence interval [CI] 95%; 2-12.6) when presenting this degree of renal dysfunction. The prevalence of preoperative renal dysfunction in this sample was 27%. The prevalence of moderate or severe CKD (GFR<60ml/min/1.73m2) in the Pickering study cited above was 28%, while that of Bagshaw was 46%, which left our study between them (33.1%). Excluding these patients the 6 5 4 56 3 Act Cr – est Cr (mg/dl) patients undergoing cardiac surgery. A similar conclusion was reached by several authors using more heterogeneous cohorts than ours. In the BEST Kidney study, Bagshaw17 found an overestimation of 42% using the same formula to calculate baseline SCr from a prospective study of over 1300 patients from 54 intensive care units (ICUs) in 23 countries. In a cohort study of almost 5000 patients hospitalised during a year, Siew18 compared 3 different methods to estimate the baseline serum creatinine with values obtained from preadmission renal function for diagnosing AKI. The methods were SCr calculated from MDRD with a GFR of 75ml/min, the minimum SCr value during admission and the SCr value on admission. The first 2 methods overestimated the incidence by almost 50%, while the use of SCr on admission underestimated the diagnosis by 46%. More recently, Závada19 found similar results when comparing 3 different methods, including the one recommended by ADQI, in nonselected patients from 3 different hospital ICUs. Finally, after prospectively analysing 224 patients from 2 general ICUs used as controls in the EARLYRAF study, Pickering and Endre20 found an overall overestimation of 35.7% with an assumed GFR of 75ml/min, which increased to 92.8% when a baseline GFR of 100ml/min was considered. Unlike our cohort study, only 23% of the patients underwent cardiac surgery, and the rest had thoracic surgery, vascular surgery, 2 1 0 -1 -2 -3 -4 -5 -6 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 (Act Cr – est Cr)/2 (mg/dl) The thick line represents the average difference (bias), and the two thin lines above and below it show its standard deviation (precision). The higher the baseline creatinine, the greater the difference between both values, i.e. the degree of agreement decreases. Figure 1. Bland and Altman plot for the global cohort (n=2103) showing the degree of agreement between both creatinine values Nefrologia 2012;32(1):53-8 A.M. Candela-Toha et al. RIFLE-AKIN in cardiac sugery originals 0.8 0.6 Act Cr – est Cr (mg/dl) 0.4 0.2 0.0 –0.2 –0.4 –0.6 –0.8 0.5 0.6 0.7 0.8 0.9 1.0 1.1 1.2 1.3 (Act Cr – est Cr)/2 (mg/dl) The agreement between both creatinine values improves significantly. Note the difference in scales compared to Figure 1. CKD: chronic kidney disease Figure 2. Bland and Altman plot after excluding patients with impaired renal function, stage 3 CKD (n=1408) overestimation decreased in all 3 studies. This effect is easily explained, as a significant number of cases where the patient is given a GFR above the real one are excluded. Another concept worth mentioning, which was also described by Wijeysundera, is that of occult renal insufficiency.22 This is defined as preoperative renal function with a CrCl<60ml/min, but with a preoperative baseline SCr<1.13mg/dl. In a cohort of over 10 000 patients undergoing cardiac surgery, the prevalence of occult renal failure was 9%, and was very similar to our sample (8.5%). This concept stresses the importance of estimating GFR or CrCl in addition to baseline SCr in a preoperative analytical evaluation. The risk factors for AKI in the context of cardiac surgery have been extensively studied, with several grading systems established to calculate RRT preoperative risk.23-25 Preoperative renal function appears constantly among the factors considered. The incidence of AKI was 14.2% in our sample, lower than both the Pickering (31.2%) and BEST (44.9%) studies. Since the number of diagnosed cases is the denominator in the overestimation calculation, the smaller this number is, the higher the overestimation. Together with the different prevalence of CKD, this situation may explain our findings. We believe that the main limitation of this study is the use of baseline SCr taken as part of the preoperative evaluation: this value need not necessarily reflect the subject’s SCr at Nefrologia 2012;32(1):53-8 baseline. A significant number of patients admitted to the emergency department with symptoms of ischaemic heart disease or heart failure undergo cardiac surgery several days later, once the patient has been stabilised. However, similar approaches have been used in the above studies. Although the current recommendation is to use a SCr value obtained in the 3 months before admission, there is no known best method for calculating baseline SCr when a baseline measurement is not available. In the study by Pickering and Endre,20 the random assignment of SCr values from a lognormal distribution curve adjusted to the parameters of central tendency and dispersion of the source population was the method that most closely approximated to the actual incidence (with an AKI underestimation of 2.9%). Failing that, and as a more practical method, the authors recommend using the lowest SCr observed during the first 7 days of hospital stay (an AKI overestimation of 5.7%). This recommendation, of course, should only be done for an epidemiological analysis; as such a delay in diagnosis is not acceptable from a healthcare point of view. A second limitation of this study is the use of SCr criterion only for AKI diagnosis, excluding patients diagnosed from a decrease in diuresis. This is a problem for all retrospective analyses using the RIFLE system, which are the vast majority of those published. This is because this parameter is rarely reflected in databases with sufficient detail to be applied (i.e., on an hourly basis). Therefore, we did not give sensitivity and specificity values, as all criteria must be used to make the diagnosis reliably and to classify those patients with the most severe stage. CONCLUSIONS Using baseline creatinine calculated from the MDRD equation in the diagnosis of AKI overestimates the incidence in patients undergoing cardiac surgery. This is common in populations with an increased prevalence of mild CKD, with the intensity depending both on this factor and the incidence of AKI. However, this is acceptable in patients whose premorbid GFR is normal or nearly normal. This observation must be considered, especially in hospital-based epidemiological studies. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. REFERENCES 1. Rosner MH, Okusa MD. Acute kidney injury associated with cardiac surgery. Clin J Am Soc Nephrol 2006;1:19-32. 2. Bellomo R, Ronco C, Kellum J, Mehta R, Palevsky P. Acute renal failure-definition, outcome measures, animal models, fluid therapy and 57 originals 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004;8:R204-R212. Mehta R, Kellum J, Shah S, Molitoris B, Ronco C, Warnock D, et al. Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care 2007;11:R31. Liaño F, Candela A, Tenorio M, Rodríguez-Palomares J. La IRA en la UCI: Concepto, clasificaciones funcionales, epidemiología, biomarcadores, diagnóstico diferencial y pronóstico. In: Poch E, Liaño F, Gaínza F, eds. Manejo de la disfunción aguda del riñón del paciente crítico en la páctica clínica (first ed.). Madrid: Ergon; 2011. p. 1-21. Lassnigg A, Schmidlin D, Mouhieddine M, Bachmann LM, Druml W, Bauer P, et al. Minimal changes of serum creatinine predict prognosis in patients after cardiothoracic surgery: a prospective cohort study. J Am Soc Nephrol 2004;15:1597-605. Lassnigg AM, Schmid ERM, Hiesmayr MM, Falk C, Druml W, Bauer P, et al. Impact of minimal increases in serum creatinine on outcome in patients after cardiothoracic surgery: Do we have to revise current definitions of acute renal failure? Crit Care Med 2008;36:1129-37. Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J Am Soc Nephrol 2005;16:3365-70. Liaño F, Tenorio M. Fracaso renal agudo: conceptos y epidemiología. In: Hernando Avendaño L, ed. Nefrología Clínica (third ed.). Buenos Aires: Editorial Médica Panamericana; 2008. p. 733-8. Ricci Z, Cruz D, Ronco C. The RIFLE criteria and mortality in acute kidney injury: a systematic review. Kidney Int 2007;73:538-46. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D, for the Modification of Diet in Renal Disease Study Group. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Ann Intern Med 1999;130:461-70. Bagshaw SM, George C, Bellomo R, for the ANZICS Database Management Committe. A comparison of the RIFLE and AKIN criteria for acute kidney injury in critically ill patients. Nephrol Dial Transplant 2008;23:1569-74. Joannidis M, Metnitz B, Bauer P, Schusterschitz, Moreno R, Druml W, et al. Acute kidney injury in critically ill patients classified by AKIN versus RIFLE using the SAPS 3 database. Intens Care Med 2009;35:1692-702. Pickering JW, Endre ZH. GFR shot by RIFLE: errors in staging acute kidney injury. Lancet 2009;373:1318-9. A.M. Candela-Toha et al. RIFLE-AKIN in cardiac sugery 14. Latour J, Abraira V, Cabello JB, López Sánchez J. Métodos de investigación en cardiología clínica (IV). Las mediciones clínicas en cardiología: validez y errores de medición. Rev Esp Cardiol 1997;50:11728. 15. Martin Bland J, Altman D. Statistical methods for assessing agreement between two methods of clinical measurement. Lancet 1986;327:307-10. 16. National Kidney Foundation. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification and stratification. Am J Kidney Dis 2002;39:S1-S266. 17. Bagshaw SM, Uchino S, Cruz D, Bellomo R, Morimatsu H, Morgera M, et al. A comparison of observed versus estimated baseline creatinine for determination of RIFLE class in patients with acute kidney injury. Nephrol Dial Transplant 2009;24:2739-44. 18. Siew ED, Matheny ME, Ikizler TA, Lewis JB, Miller RA, Waitman LR, et al. Commonly used surrogates for baseline renal function affect the classification and prognosis of acute kidney injury. Kidney Int 2010;77:536-42. 19. Závada J, Hoste E, Cartin-Ceba R, Calzavacca P, Gajic O, Clermont G, et al. A comparison of three methods to estimate baseline creatinine for RIFLE classification. Nephrol Dial Transplant 2010;25:39118. 20. Pickering JW, Endre ZH. Back-calculating baseline creatinine with MDRD misclassifies acute kidney injury in the Intensive Care Unit. Clin J Am Soc Nephrol 2010;5:1165-73. 21. Wijeysundera DNM. Evaluating surrogate measures of renal dysfunction after cardiac surgery. Anesth Analg 2003;96:1265-73. 22. Wijeysundera DNM. Improving the identification of patients at risk of postoperative renal failure after cardiac surgery. Anesthesiology 2006;104:65-72. 23. Thakar CV, Arrigain S, Worley S, Yared JP, Paganini EP. A clinical score to predict acute renal failure after cardiac surgery. J Am Soc Nephrol 2005;16:162-8. 24. Wijeysundera DNM, Karkouti KM, Dupuis JYM, Rao V, Chan CT, Granton JT, et al. Derivation and validation of a simplified predictive index for renal replacement therapy after cardiac surgery. JAMA 2007;297:1801-9. 25. Mehta RHM, Grab JDM, O’Brien SMP, Bridges CR, Gammie JS, Haan CK, et al. Bedside tool for predicting the risk of postoperative dialysis in patients undergoing cardiac surgery. Circulation 2006;114:2208-16. Sent for review: 25 Jul. 2011 | Accepted: 3 Oct. 2011 58 Nefrologia 2012;32(1):53-8 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society originals Early detection of chronic kidney disease: Collaboration of Belgrade nephrologists and primary care physicians Ljubica Djukanović1, Visnja Lezaić2, Nada Dimković2, Gordana Perunicić Peković3, Danica Bukvić4, Sanja Bajcetić5, Jelena Pavlović6, Ana Bontić6, Nadezda Zec3, Danijela Momcilović7, Marina Stojanović Stanojević8 8 Academy of Medical Science. Serbian Medical Society. Belgrade (Serbia) Medical Faculty. Belgrade University. Belgrade (Serbia) 3 Department of Nephrology. Clinical Centre Zemun. Belgrade (Serbia) 4 Special Hospital for Endemic Nephropathy. Lazarevac (Serbia) 5 Department of Nephrology. Clinical Centre Zvezdara. Belgrade (Serbia) 6 Department of Nephrology. Clinical Centre of Serbia. Belgrade (Serbia) 7 Hemodialysis Center of Health. Obrenovac (Serbia) 8 Hemodialysis Center of General Hospital. Mladenovac (Serbia) 1 2 Nefrologia 2012;32(1):59-66 doi:10.3265/Nefrologia.pre2011.Oct.11031 ABSTRACT Background: Belgrade screening study was undertaken in order to detect persons with CKD markers in at risk populations and to educate primary care physicians how to carry out CKD screening. Methods: The study was performed by primary care physicians from thirteen Belgrade health centers in collaboration with nephrologists from clinical centers. Subjects without previously known kidney disease were enrolled: 1316 patients with hypertension without diabetes, 208 patients with type 2 diabetes and 93 subjects older than 60 years without hypertension or diabetes. The survey consisted of an interview, estimation of glomerular filtration rate (eGFR–MDRD), single urine dipstick detection of proteinuria, hematuria, glucosuria, microalbuminuria. Results: Microalbuminuria with or without proteinuria in combination with eGFR>60ml/min/1.73m2 was detected in 17% , 41% and 24% of patients with hypertension, diabetes and those above 60 years, respectively. Reduced eGFR (<60 ml/min/1.73m2 ) was found in 23%, 12% and 22% of the same patient groups. The prevalence of CKD markers increased with increasing number of risk factors. Conclusion: High prevalence of CKD markers in at risk population detected by primary care physicians in this collaborative study seems to be the best way to encourage primary care physicians to carry out regular CKD screening. Keywords: Chronic kidney disease. Screening. Primary care. Correspondence: Ljubica Djukanović Academy of Medical Science. Serbian Medical Society. Pasterova 2. 11000 Belgrade. Serbia. [email protected] [email protected] Detección precoz de la enfermedad renal crónica: colaboración entre nefrólogos y especialistas de atención primaria de Belgrado RESUMEN Antecedentes: El estudio de Belgrado se realizó para detectar personas con marcadores de ERC en poblaciones de riesgo y formar a los especialistas de atención primaria sobre cómo realizar proyecciones de ERC. Métodos: El estudio fue realizado por especialistas de atención primaria de trece centros de salud en colaboración con nefrólogos de centros clínicos. Se incluyó a personas sin enfermedad renal previa conocida: 1316 pacientes con hipertensión sin diabetes, 208 pacientes con diabetes tipo 2 y 93 pacientes de más de 60 años sin hipertensión ni diabetes. El estudio consistía en una entrevista, determinación de la tasa de filtración glomerular estimada (TFGe-MDRD) y detección de proteinuria, hematuria, glucosuria y microalbuminuria con una única tira reactiva de orina. Resultados: Se detectó microalbuminuria con o sin proteinuria en combinación con una TFGe >60 ml/min/1,73m2 en el 17%, el 41% y el 24% de los pacientes con hipertensión, diabetes y mayores de 60 años, respectivamente. Se encontró una TFGe reducida (<60 ml/min/1,73m2 ) en el 23%, el 12% y el 22% de estos mismos grupos de pacientes. La prevalencia de los marcadores de ERC aumentaba cuanto mayor era el número de factores de riesgo. Conclusión: La elevada prevalencia de marcadores de ERC en una población de riesgo detectada por los médicos de atención primaria en este estudio de colaboración parece ser la mejor forma de motivar a estos especialistas para que realicen cribados de ERC con regularidad. Palabras clave: Enfermedad renal crónica. Proyección. Atención Primaria. INTRODUCTION The steady increase in the incidence of patients on renal replacement therapy (RRT) was first noted in developed 59 originals countries and thereafter all around the globe.1-3 Over the same period diabetes and hypertension became the leading causes of chronic kidney disease (CKD) and the number of elderly patients in end-stage renal disease increased steadily3. It became obvious that more attention should be directed to prevention and early detection of CKD. Recent data indicated stabilization of incidence rates of RRT patients in many developed countries.4-6 Although different factors might have led to this stabilization, greater emphasis on early detection and prevention of CKD is doubtless one of them. However, our country is among those European countries where the incidence rate of patients on RRT is continuing to increase.7,8 The experience of developed countries in primary and secondary prevention of CKD has taught us that attention must be moved from treating advanced stages of CKD towards treatment of the early stages. As early stage of CKD in most patients is asymptomatic and undiagnosed, detection can be achieved only by active screening. It was proposed that prevention and early detection of CKD would be best managed in a partnership between primary and secondary care.9 Therefore, we undertook the first study for early detection of CKD in Serbia in which primary care physicians from thirteen Belgrade health centers collaborated with nephrologists from Belgrade clinical centers. The aim was to detect persons with CKD markers in populations at risk and also to educate primary care physicians how to carry out screening for CKD and how to interpret the results and manage subsequent treatment alone or in collaboration with nephrologists. SUBJECTS AND METHODS The present paper presents the results of screening for CKD carried out in Belgrade under the leadership of the Academy of Medical Science of the Serbian Medical Society. The study included three steps: (1) Education. Educative meetings for primary care physicians on prevention and early detection of CKD were organized by the Academy in 2008. (2) Organization of study for detection of persons with CKD markers. At the beginning of 2009 primary care physicians from all 13 Belgrade Health Centers were invited to participate in the screening study. After their positive response nephrologists from three clinical centers presented the study design to primary care physicians who carried out investigations from April to June 2009. (3) Results presentation and guideline distribution. In November 2009, the Academy organized a meeting where the results from each health center were presented by general practitioners and the overall findings were collated and reported by nephrologists, coordinators of the study. At the same time a guideline for early detection and treatment of chronic kidney disease, prepared by members of the Academy was presented and distributed to all participants.10 This study enrolled 1617 adult patients without previously known renal disease who came for regular check-ups to their primary care physicians in Belgrade Health centers 60 Ljubica Djukanović et al. Belgrade screening study over a three month period. The patients were enrolled in the study according to the following criteria: patients with hypertension for more than 5 years, patients with type 2 diabetes mellitus for more than 5 years regardless of the presence or not of hypertension, and persons older than 60 years without hypertension or diabetes. Exclusion criteria involved: previously known kidney diseases, malignant disease, congestive heart disease, pregnancy, any acute illness, as well as persons younger than 18 years. Informed consent was obtained from all patients and the Ethics Committee of the Clinical Center of Serbia approved the study. The survey started with an interview in which the participants answered a detailed questionnaire on demographic issues, personal medical and family history with special attention to duration and treatment of hypertension and diabetes and data on smoking. After the interview the primary care physicians examined all selected subjects physically, including measurement of body weight, height and blood pressure. In addition, they analyzed the medical records of each subject and calculated the average systolic and diastolic blood pressure as well as the average serum glucose level using all values registered in the year preceding the study. These data were also noted in the questionnaire. All participants were sent to the laboratory where serum creatinine level was measured by a kinetic Jaffé method and glomerular filtration rate (eGFR) estimated using the original Modification of Diet in Renal Disease (MDRD) Study formula.11 Proteinuria, hematuria, glycosuria and microalbuminuria were assessed semiquantitatively in spot urine samples using the urine dipstick test. Proteinuria, hematuria, glycosuria were defined when dipstick analysis quantified them as 1+ or more. Microalbuminuria (MAU) was detected with Micral-test® strips (ACCU-CHEK product, Roche Diagnostics). These immunoassay reagent strips (monoclonal antibodies to human albumin) reveal a distinctive color corresponding to a scale on the vial label giving a range of albumin concentrations as follows: negative, 20mg/L, 50mg/L and 100mg/L. According to the manufacturer’s instructions albumin concentrations detected as _> 20mg/L are consistent with microalbuminuria _> 30mg/day. This was confirmed in 100 patients with MAU detected by the Micral-dipstick test during the screening and subsequent determination of albumin in a 24h urine collection by immunonephelometry and Micral-dipstick. Therefore, the finding of MAU _> 20mg/L was considered as the presence of MAU. Nephrologist coordinators collected the questionnaire lists and laboratory results from all health centers participating in the study and analyzed the results. A person was considered as having arterial hypertension if systolic blood pressure was _>140mmHg and/or diastolic Nefrologia 2012;32(1):59-66 Ljubica Djukanović et al. Belgrade screening study pressure was _>90mmHg or if antihypertensive treatment had been previously prescribed. Patients with known and treated type 2 diabetes were considered diabetic regardless of their glycemic control. Body mass index (BMI) was calculated using the formula: weight (kg)/height2 (m2) and, depending on the obtained BMI value, patients were classified according to the WHO recommendations.12 Smoking was defined as actual use of cigarettes (not ex-smokers). The estimated GFR (eGFR) was used to classify subjects into Kidne Disease Outcomes Quality Initiative (K/DOQI) stages of CKD.13 In addition, stage 3 CKD was divided into two substages:14 sub-stage 3a with a GFR between 45 mL/min/1.73m2 and 59 mL/min/1.73m2 and sub-stage 3b with a GFR between 30 mL/min/1.73m2 and 44 mL/min/1.73m2. Descriptive statistics were presented as mean values ± standard deviation (SD) for the continuous variables, or as frequencies for categorical variables. Analysis of variance accompanied by Bonferroni multiple comparison tests and chi-square analysis were used for between-group comparisons of continuous and categorical variables, respectively. Pearson correlation coefficients were used to detect associations among eGFR and other variables. A p value of <0.05 was considered to be statistically significant. All analyses were performed using the SPSS statistical software package (Version 10; SPSS). RESULTS Out of 1617 subjects enrolled into the study, 1316 were patients with hypertension without diabetes, 208 patients had type 2 diabetes and 93 were subjects older than 60 years who had neither hypertension nor diabetes. Thus, patients with hypertension formed the largest group, but the relatively low number of individuals with type 2 diabetes enrolled in the study could have been due to way their health care is organized in our health centers. Namely, the majority of patients with diabetes are not controlled by the general practitioners who recruited patients for our screening studies but in special advice centers for diabetics. In addition, the number of enrolled subjects older than 60 years without hypertension and diabetes was also low, because people of that age frequently had hypertension or a comorbidity excluding them from the present study. Inclusion criteria allowed patients with hypertension older than 60 years to be included in the hypertension group, while patients with type 2 diabetes regardless of having hypertension or not or being above 60 years old were included in the diabetes group. Therefore, some subjects could have had more than one of the three risk factors for CKD. The Venn diagram presented in figure 1 shows that 759 (57.6%) of the patients with hypertension were older Nefrologia 2012;32(1):59-66 originals than 60 years, while among patients with diabetes 93 (44.7%) were above 60 years old and had hypertension, 61 (29.3%) had hypertension but were younger than 60 years and only 10 (5%) diabetics were normotensive and younger than 60 years. The demographic and clinical characteristics of the study population are shown in table 1. Patients with diabetes had significantly higher mean BMI and, as expected, the groups of patients with hypertension and diabetes had significantly higher systolic and diastolic blood pressure than the group of subjects older than 60 years. The results of laboratory analyses are presented in table 2. Patients with diabetes had significantly higher mean eGFR and most of them were in stage 1 and the least in stage 3 of CKD. When the patients in stage 3 of CKD were divided into two substages only one seventh of them belonged to substage 3b. Table 2 also shows that patients with diabetes had the highest prevalence of both MAU and proteinuria, but patients with hypertension had a significantly lower prevalence of MAU than the other two groups. Calculation of the Pearson correlation coefficient revealed significant associations between eGFR and systolic blood pressure (r=-0.163, p=0.023), duration of hypertension (r= -0.276, p=0.0001) and age (r=-0.383, p=0.0001). Different combinations of laboratory findings were detected in the examined subjects (figure 2). The most frequent pathological finding was MAU with or without proteinuria in combination with eGFR above 60ml/min/1.73m2. In patients with diabetes this was detected in the majority (41%) of patients with pathological laboratory findings, while in two other groups this percent was about two times lower. In the group with hypertension and with subjects older than 60 years the percentage of patients with Patients with diabetes Patients with hypertension Subjects > 60 years Figure 1. Venn diagram presenting the distribution of patients depending on the presence of three risk factors. 61 Ljubica Djukanović et al. Belgrade screening study originals Table 1. Demographic and clinical characteristics of the study groups Variable 1 2 3 Patients with hypertension (HTA) Patients with type 2 diabetes (DM) > 60 yrs without HTA and DM 1316 208 93 Total number Gender: females, no (%) 801 (60.8) 101 (48.5) 54 (58.1) Age, years 62.4±10.4 64.1±10.2 69.6±5.8a Body mass index, kg/m2 27.5±5.1b 33.9±3.6 25.4±5.2c 15.61-46.88 19.37-49.95 18.38-49.59 Range <18.5 4 (0.3) 0 2 (2.3) 18.5–24.9 328 (24.9) 37 (18.1) 42 (44.7) 25–29.9 616 (46.2) 95 (45.2) 39 (42.3) >30 368 (28.0) 76 (36.6) 10 (10.6) Systolic 140.1±17.8 138.0±19.34 125.3±10.1c Diastolic 85.7±9.9b 82.0±8.67 78.4±6.3c - 126 (60.6%) - 11.2±7.4 8.03±7.7 980 (74.5) 138 (66.3) 0 266 (25) 53 (25) 17 (18.3) Hypertension 171 (14) 124 (59.6) 23 (24.7) Type 2 diabetes mellitus 936 (77) 110 (52.8) 17 (18.3) Blood pressure, mmHg HbA1c > 7.5% Duration of Hypertension, yrs Type 2 diabetes mellitus, yrs No of patients treated with ACEi Smokers 9.4±6.7 Positive family history for Kidney disease 22 (10.6) Data are means ± SD or numbers (percentages). a p=0.0001, 1 vs.3; b p=0.005, 1 vs.2; c p<0.0001 3 vs.1,2. MAU/proteinuria and eGFR above 60ml/min/1.73m2 was similar to that of patients with normal urinary finding and eGFR below 60 ml/min/1.73m2. As many of our subjects had more than one of the three risk factors for CKD (hypertension, diabetes, older age), the frequency of MAU and reduced eGFR was examined depending on the number of risk factors. The analysis showed that in subjects with one, two or three risk factors MAU was present in 135 (21.0%), 215 (25.2%) or 44 (46.3%) subjects, but eGFR below 60ml/min/1.73m2 in 101 (15.4%), 241 (28.4%) or 26 (26.8%) subjects, respectively. DISCUSSION The present study aimed to detect persons with CKD markers in at risk population was carried out in Belgrade through the collaboration of nephrologists and primary care physicians and involved 1617 subjects. Among them there were 1316 patients with hypertension without diabetes, 208 patients with type 2 diabetes and 93 62 subjects older than 60 years without either hypertension or diabetes. Pathological findings were revealed in 4656% of the subjects the most frequent being MAU with or without proteinuria in combination with eGFR above 60ml/min/1.73m 2 that was found in 17%, 41% and 24% of the subjects with hypertension, diabetes and those older than 60 years, respectively The dramatic increase of CKD prevalence has directed the attention of nephrologists towards prevention and early detection, so numerous screening programs have been running all over the world. Nevertheless, the methods used in these studies have differed widely. In some the screening was limited to determination of serum creatinine level and eGFR 15-17 but, with time, the opinion prevailed that, besides GFR, screening should also include a measure of proteinuria or even better microalbuminuria. 18-20 In addition, low eGFR and albuminuria are two basic markers for CKD classification proposed by the Kidney Disease Outcomes Quality Initiative of the National Kidney Foundation13. In the present study dipstick measurement of Nefrologia 2012;32(1):59-66 Ljubica Djukanović et al. Belgrade screening study originals Table 2. Kidney function and urinary findings in the three studied groups Variable 1 2 3 Patients with hypertension (HTA) Patients with type 2 diabetes (DM) >60 yrs without HTA and DM 70.56±16.04a 78.1±16.1 69.5±13.3b 18.3-128.6 41.3-129.9 40.9-123.3 142 (10.8) 54 (26)a 6 (6.5) 2. 60-89.9ml/min/1.73m 848 (64.4) 130 (62.5) 67 (72.0) 3. 30-59ml/min/1.73m2 304 (23.1) 24 (11.5)b 20 (21.5)b 3a. 45-59.9ml/min/1.73m2 262 (20) 19 (9.1)a 17 (18.3) 3b. 30-44.9ml/min/1.73m 42 (3.2) 5 (2.4) 3 (3.1) 22 (1.7) 0 0 Microalbuminuria 279 (21.2) 99 (47.6)a 41 (44.3)c Proteinuria 119 (9.0) 35 (16.8) 9 (10.2) Hematuria 50 (3.8) 10 (4.8) 8 (8.6) eGFR, ml/min/1.73m2 range, ml/min/1.73m 2 Number (%) of patients with eGFR: 1. > 90ml/min/1.73m2 2 2 4. <30ml/min/1.73m2 Number (%) of patients with: b Glycosuria 68 (32.9) Data are means ± SD or numbers (percentages). a p=0.0001, 1 vs.3; b p=0.005, 1 vs.2; c p<0.0001 3 vs.1,2. urine protein and albumin was used together with estimation of GFR by MDRD equation and different combinations of these markers were found in the examined persons. The second methodological question is who should be included in screening for CKD. Universal screening of unselected populations not already known to be at risk has 100% 90% 80% 70% 60% 50% 40% 30% 20% 10% 0% Hypertension Diabetes eGFR >60ml/min/1.73m + neg. urine eGFR >60ml/min/1.73m2 + MAU+H eGFR <60ml/min/1.73m2 + MAU/P 2 >60 years eGFR >60ml/min/1.73m2 + MAU/P eGFR <60ml/min/1.73m2 + neg.urine eGFR <60ml/min/1.73m2 + H MAU/P – MAU and/or proteinuria, H – hematuria Figure 2. Distribution of subjects according to eGFR and urinary findings. Nefrologia 2012;32(1):59-66 not been shown to be cost-effective, so targeted screening for CKD was proposed as more economical than universal screening.21-23 In different guidelines diverse at risk populations have been proposed for screening but patients with hypertension or diabetes were generally accepted as relevant ones.13,24 Therefore, these two at risk groups were included here in addition to subjects older than 60 years without hypertension or diabetes as also proposed as a target population by KDOQI guidelines11. Our inclusion criteria enabled some subjects to have more than one of these three risk factors for CKD. More than a half of the patients with hypertension or diabetes were above 60 years old and majority of patients with diabetes had hypertension. Analysis of the results revealed that subjects with two or three risk factors had almost double the prevalence of reduced eGFR and MAU compared with those with only one risk factor. Reduced eGFR (<60ml/min/1.73m2), a criterion for CKD stage 3 according to KDOQI guidelines,13 was found in 15% to 23% of subjects in the three at risk groups. These values are comparable to those obtained in other studies that targeted different at risk populations.25-27 However, as GFR declines with normal ageing, it was indicated that using KDOQI guidelines classification large numbers of the elderly and females would be classified in CKD stage 3 without other objective evidence of kidney disease.28 In addition, several epidemiological studies showed that the risk of progressive decline in renal function or of cardiovascular events was not equal for all subjects with stage 3 CKD, being low for subjects with eGFR29,30 between 30 to 59ml/min/1.73m2. Therefore, it was proposed to subdivide CKD 3 into stage 3a (eGFR between 63 originals 45–59ml/min/1.73 m2) and stage 3b (eGFR between 30–44ml/min/1.73 m2).14,29,31 This classification was used in the present study and most of the 348 subjects with stage 3 CKD belonged to substage 3a (298; 85.6%). If the subjects with eGFR in this range had neither MAU nor proteinuria, they were considered to be not at risk for renal disease progression.31 Figure 2 showed that in the group of subjects older than 60 years without hypertension and diabetes there were six times more subjects with eGFR below 60ml/min/1.73m2 and normal urinary findings than with pathological urinary findings. In the group with hypertension in which 56.6% of the patients were older than 60 this ratio was 2.3:1. Nevertheless, there are many arguments that the decline in kidney function with age cannot be considered as normal physiology. Therefore, although the subdivision of CKD 3 is useful to focus the attention of health care professionals on patients in substage 3b with worse cardiovascular and CKD outcomes, it does not mean that persons in stage 3a, even with normal urinary findings, could be excluded from the measures for prevention of CKD progression. Also, eGFR, regardless of classification into two substages, should be taken into account in medication dosing, so avoiding drug-induced kidney toxicity. As CKD in its earliest stages is usually asymptomatic, it is often overlooked and most patients, even in at risk populations, were unaware of the CKD diagnosis.32-34 One exclusion criterion in our study was previously known kidney disease and all patients denied any knowledge of kidney disease. Also, their primary care physicians had no data on previous kidney disease, although more than 80% of the patients had checked serum creatinine level and urine analysis in the year preceding the study.35 However, examination of MAU is not available in our health center laboratories, while estimation of GFR has not been introduced in the regular practice of general practitioners, so, despite laboratory control, many CKD patients might remain undetected. Therefore, one of the aims of this screening study was to educate primary care physicians about screening for CKD. The educative meeting that preceded the study, the collaboration of nephrologists and primary care physicians during the study, and especially the results obtained clearly suggested the general practitioners on the necessity of regular screening in at risk populations. As already stressed by other authors, prevention and early detection of CKD cannot be managed by an insufficient number of nephrologists and in nephrology outpatient clinics and it would be best managed in a partnership arrangement between primary and secondary care.9,36 However, it was shown that primary care physicians did not have enough knowledge for this task.37,38 Our results also indicated insufficient attention of primary care physicians to CKD prevention and management. Although, most patients with hypertension used ACEi, average blood pressure in the year preceding the study was above the target value proposed by 64 Ljubica Djukanović et al. Belgrade screening study guidelines. At inclusion in the study 54.5% of patients had systolic blood pressure above 140mmHg and 40.2% had diastolic blood pressure above 90mmHg. A similar state was found concerning control of glycemia in diabetics and 60.6% of these patients had average HbA1c above 7.5%. In addition, the majority of patients, especially those in the hypertension and diabetes groups, were overweight. Therefore, additional education of primary care physicians on risk factors, prevention, early detection and management of CKD is necessary and nephrologists should have the main role in this education. That directed us to organize the presented project and prepare guidelines for prevention, early detection and treatment of CKD patients for primary care physicians10. Nevertheless, guidelines alone are not sufficient and they can be implemented only in close collaboration between primary care physicians and nephrologists. This study has some limitations. First, it was a crosssectional study and CKD markers were measured just once. That might cause false positive and false negative errors. Secondly, both proteinuria and MAU were detected by urine dipstick test with limited accuracy. The use of a single measurement of dipstick proteinuria might lead to overestimation as described for a study population in the United States, where in a repeat measurement only 63% of subjects with proteinuria had a positive result.39 Despite these limitations, the present study carried out in Belgrade is the first study for early detection of CKD in Serbia. Moreover, the study was carried out in collaboration between primary care physicians and nephrologists with the aim to encourage primary care physicians to carry out regular control of CKD markers in patients at risk for CKD and to established better collaboration between primary care physicians and corresponding nephrologists. At the same time similar screening studies are being prepared in several other Serbian towns in order to spread knowledge about the significance and necessity of early detection of CKD as a task of primary care. Conflict of interest The authors have no conflicts of interest to declare. REFERENCES 1. Stengel B, Billon S, Van Dijk PC, Jager KJ, Dekker FW, Simpson K, et al. Trends in the incidence of renal replacement therapy for endstage renal disease in Europe, 1990–1999. Nephrol Dial Transplant 2003;18:1824-33. 2. U. S. Renal data System. USRDS 2001 Annual Data Report: Atlas of End-Stage Renal Disease in the United States, Bethesda, MD, Nefrologia 2012;32(1):59-66 Ljubica Djukanović et al. Belgrade screening study 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, 2001. Levey AS, Atkins R, Coresh J, Cohen EP, Collins AJ, Eckardt KU, et al. Chronic kidney disease as a global public health problem: approaches and initiatives - a position statement from Kidney Disease Improving Global Outcomes. Kidney Int 2007;72:247-59. Kramer A, Stel V, Zoccali C, Heaf J, Ansell D, Grönhagen-Riska C, et al.; ERA-EDTA Registry: An update on renal replacement therapy in Europe: ERA–EDTA Registry data from 1997 to 2006. Nephrol Dial Transplant 2009;24:3557-66. US Renal Data System. USRDS 2008 Annual Data Report: Atlas of End-Stage Renal Disease in the United States, National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 2008. Canadian Institute for Health Information. 2007 Annual Report— Treatment of End-Stage Organ Failure in Canada, 1996–2005, CIHI, Ottawa, 2008. Djukanović Ljubica on behalf of working group on dialysis treatment in Serbia: Epidemiological characteristics of patients on renal replacement therapy in Serbia. [in Serbian]; in Djukanović Lj, ed: Lec enje dijalizom i transplantacijom bubrega u Srbiji u periodu 1999-2009. Monografije naucnih sastanka AMN SLD 2010;1(3):1126. ERA-EDTA Registry: ERA-EDTA Registry Annual Report 2008. Academic Medical Center, Department of Medical Informatics, Amsterdam, The Netherlands, 2010. Available at http://www.eraedta-reg.org/index.jsp Frankel A, Brown E, Wingfield D. Management of chronic kidney disease. Primary and secondary care need to set up a model of combined care. BMJ 2005;330:1039-40. Djukanović Lj, Leczaić V, Dimković N. Reccomendations for prevention and treatment of chronic kidney disease. [in Serbian]; in Djukanović Lj, Leczaić V, Dimković N, eds: Hipertenzija i hronicna bolest bubrega. Monografije nauc nih sastanka AMN SLD 2010;1(1):149-74. Levey AS, Greene T, Kusek JW, Beck GL. A simplified equation to predict glomerular filtration rate from serum creatinine. J Am Soc Nephrol 2000;11(suppl):A0828. World Health Organization (WHO). Obesity: Preventing and Managing the Global Epidemic. Report of a WHO Consultation. World Health Organization: Geneva, Switzerland, 2000. WHO Technical Report Series 894. National Kidney Foundation. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification and stratification. Am J Kidney Dis 2002;39(2 Suppl 1):S1-S266. Early Identification and Management of Chronic Kidney Disease in Adults in Primary and Secondary Care. Available at:http://guidance.nice.org.uk/CG73 Cirillo M, Laurenzi M, Mancini M, Zanchetti A, Lombardi C, de Santo NG. Low glomerular filtration rate in the population. Prevalence, associated disorders, and awareness. Kidney Int 2006;70:800-6. Hallan SI, Dahl K, Oien CM, Grootendorst DC, Aasberg A, et al. Screening strategies for chronic kidney disease in the general population: Follow-up of cross sectional health survey. BMJ 2006;333:1047. Nefrologia 2012;32(1):59-66 originals 17. de Jong PE, van der Velde M, Gansevoort RT, Zoccali C. Screening for chronic kidney disease: where does Europe go? Clin J Am Soc Nephrol 2008;3:616-23. 18. Garg AX, Kiberd A, Clark WF, Haynes RB, Clase CM. Albuminuria and renal insufficiency prevalence guides population screening: Results from the NHANES III. Kidney Int 2002;61:2165-75. 19. Atthobari J, Asselbergs FW, Boersma C, de Vries R, Hillege HL, van Gilst WH, et al. Cost-effectiveness of screening for albuminuria and subsequent treatment with an ACE inhibitor to prevent cardiovascular events: A pharmacoeconomic analysis linked to the PREVEND and the PREVEND IT studies. Clin Ther 2006;28:432-44. 20. de Jong PE, Gansevoort RT. Fact or fiction of the epidemic of chronic kidney disease—let us not squabble about estimated GFR only, but also focus on albuminuria. Nephrol Dial Transplant 2008;23:1092-95. 21. Craig JC, Barratt A, Cumming R, Irwig L, Salkeld G. Feasibility study of the early detection and treatment of renal disease by mass screening. Intern Med J 2002;32:6-14. 22. Boulware LE, Jaar BG, Tarver-Carr ME, Brancati FL, Powe NR. Screening for proteinuria in US adults: A cost-effectiveness analysis. JAMA 2003;290:3101-14. 23. Manns B, Hemmejgarn B, Tonelli M, Au F, Chiasson TC, Dong J, et al. Alberta Kidney Disease Network: Population based screening for chronic kidney disease: cost effectiveness study. BMJ 2010;341:c5869. 24. Joint Specialty Committee of the Royal College of Physicians of London and the British Renal Association. Guidelines for iden¬tification, management and referral of adults with chronic kidney disease. London: Department of Health for England, 2005; 20-3. Available at: http://www.rcplondon.ac.uk/pubs/books/kidney/ 25. Vassalotti JA, Li S, Chen S-C, Collins AJ. Screening populations at increased risk of CKD: The Kidney Early Evaluation Program (KEEP) and the Public Health Problem. Am J Kidney Dis 2009;53(3 Suppl 3):S107-14. 26. Middleton RJ, Foley RN, Hegarty J, Cheung CM, McElduff P, Gibson JM, et al. The unrecognized prevalence of chronic kidney disease in diabetes. Nephrol Dial Transplant 2006;21:88-92. 27. Watanabe Y, Fujii H, Aoki K, Kanazawa Y, Miyakawa T. A crosssectional survey of chronic kidney disease and diabetic kidney disease in Japanese type 2 diabetic patients at four urban diabetes clinics. Intern Med 2009;48:411-4. 28. Glassock RJ, Winearls C. Screening for CKD with eGFR: doubts and dangers. Clin J Am Soc Nephrol 2008;3:1563-8. 29. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events and hospitalization. N Engl J Med 2004;351(13):1296-305. 30. Wen CP, Cheng TY, Tsai MK, Chang YC, Chan HT, Tsai SP, et al. Allcause mortality attributable to chronic kidney disease: a prospective cohort study based on 462 293 adults in Taiwan. Lancet 2008;371:2173-82. 31. Gansevoort RT, de Jong PE. Challenges for the present CKD classification system. Curr Opin Nephrol Hypertens 2010;19:30814. 32. Stevens LA, Fares G, Fleming J, Martin D, Murthy K, Qiu J, et al. Low rates of testing and diagnostic codes usage in a commercial clinical laboratory: evidence for lack of physician awareness of 65 originals chronic kidney disease. J Am Soc Nephrol 2005;16:2439-48. 33. Nickolas TL, Frisch GD, Opotowsky AR, Arons R, Radhakrishnan J. Awareness of kidney disease in the US population: findings from the National Health andNutrition Examination Survey (NHANES) 1999 to 2000. Am J Kidney Dis 2004;44:185-97. 34. Bang H, Vupputuri S, Shoham DA, Klemmer PJ, Falk RJ, Mazumdar M, et al. Screening for occult renal disease (SCORED) simple prediction model for chronic kidney disease. Arch Intern Med 2007;167:374-81. 35. Leczaić V, Dimković N, Perunićić G, et al. Results of Belgrade study for early detection of kidney disease.[Serbian]; in Djukanović Lj, Leczaić V, Dimković N, eds: Hipertenzija i hroniãna bolest bubrega. Monografije naucnih sastanka AMN SLD 2010;1(1):129-48. Ljubica Djukanović et al. Belgrade screening study 36. Lewis R, Dixon J. Rethinking management of chronic diseases. BMJ 2004;328:220-2. 37. Lenz O, Fornoni A. Chronic kidney disease care delivered by US family medicine and internal medicine trainees: results from an online survey. BMC Med 2006;4:30. 38. Crinson I, Gallagher H, Thomas N, de Lusignan S. How ready is general practice to improve quality in chronic kidney disease? A diagnostic analysis. Br J Gen Pract 2010;60(575):403-9. 39. Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis 2003;41:1-12. Sent for Review: 25 Jul. 2011 | Accepted: 3 Oct. 2011 66 Nefrologia 2012;32(1):59-66 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society originals Serum uric acid as a marker of all-cause mortality in an elderly patients cohort Manuel Heras, María J. Fernández-Reyes, Rosa Sánchez, Álvaro Molina, Astrid Rodríguez, Fernando Álvarez-Ude Nephrology Department. General Hospital of Segovia. Segovia, Spain Nefrologia 2012;32(1):67-72 doi:10.3265/Nefrologia.pre2011.Nov.11021 ABSTRACT Introducción: There is growing evidence of the role of serum uric acid (SUA) as a risk factor for cardiovascular and renal disease. We analyze the association between baseline SUA and overall mortality in a cohort of elderly patients followed prospectively for 5 years. Patients and Methods: 80 clinically stable patients, median age 83 years (range 69-97), 31.3% men, 35% diabetic, 83% hypertensives, randomly recruited in Geriatrics and Nephrology consultations between January and April 2006, were followed for 5 years. We measured baseline SUA and serum creatinine and we estimated glomerular filtration rate (GFR) with MDRD abreviated. In patient of Nephrology Department we measured proteinuria in collection urine 24 hours and patients Geriatrics department we measured proteinuria (mg/dl)/creatinine (mg/dl) in urine (first miccion). Predictive variables were: baseline SAU and plasma creatinine; estimated GFR (abbreviated MDRD formula); recorded age, gender, baseline comorbidity (Charlson index), cardiovascular disease individualized, treatment and mortality. Statistical analysis: SPSS15.0. Results: baseline SUA was normally distributed and its median was 5.85 mg/dl. We found not significant differences in levels of SUA by gender, history of diabetes mellitus, hypertension, diuretic use, heart disease, peripheral arterial disease or stroke. Patients with an history of heart failure had significantly higher SUA (7.00±1.74 vs. 5.90±1.71, P=0.031). 41 deaths occured during follow-up (15 men and 26 women): 15 general deterioration, 8 infections, 4 stroke, 4 tumors, 3 cardiovascular disease, 2 complications of fractures and 5 unknown. Patients with SUA higher than the median had significantly lower GFR and higher mortality at 5 years. In Cox´ analysis for overall mortality (independent variables: age, gender, Charlson Index, history of heart failure, SUA, creatinine, proteinuria and GFR:MDRD), only SUA levels (HR:1,35; 1,17-1,56 p=0,000) were independently associated with mortality. Conclusions: In our study, levels of SUA are shown as independent risk factor for mortality in elderly patients. Keywords: Serum uric acid. Mortality. Glomerular filtration rate. Chronic kidney disease. Elderly. Utilidad del ácido úrico como marcador de mortalidad global en una cohorte de ancianos RESUMEN Introducción: Existe evidencia creciente del papel del ácido úrico (AU) como factor de riesgo cardiovascular y renal. En este trabajo analizamos la asociación entre niveles basales de AU y mortalidad global en una cohorte de ancianos seguidos prospectivamente durante 5 años. Pacientes y métodos: 80 pacientes clínicamente estables; mediana de edad, 83 años (rango 6997); 31,3% varones; 35% diabéticos; 83% hipertensos; reclutados aleatoriamente en consultas de Geriatría y Nefrología entre enero y abril de 2006 y seguidos durante 5 años. Medimos basalmente AU y creatinina en plasma y estimamos filtrado glomerular (FG) con fórmula MDRD abreviada. Asimismo, en los pacientes de Nefrología se midió la proteinuria mediante la recogida de orina de 24 horas, y en los vistos en Geriatría se estimó a partir del cociente proteínas (mg/dl)/creatinina (mg/dl) en primera orina de la mañana. Registramos edad, género, comorbilidad basal (Índice de Charlson), patologías cardiovasculares individualizadas, tratamientos y mortalidad. Estadística: SPSS15.0. Resultados: El AU basal presentaba una distribución normal y su mediana era de 5,85 mg/dl. No encontramos diferencias significativas en los niveles de AU según género, antecedentes de diabetes méllitus, hipertensión arterial, uso de diuréticos, cardiopatía isquémica, arteriopatía periférica o ictus. Los pacientes con antecedentes de insuficiencia cardíaca tenían AU significativamente mayor (7,00 ± 1,74 vs. 5,90 ± 1,71, p = 0,031). 41 pacientes (15 varones y 26 mujeres) fallecieron: 15 por deterioro en el estado general; 8 por infecciones; 4 por ictus; 4 por tumores; 3 por causas cardiovasculares; 2 por complicaciones de fracturas y 5 por causas desconocidas. Los pacientes con AU superior a la mediana tenían un FG significativamente menor y una mortalidad a los 5 años más elevada. En el análisis de Cox para mortalidad global (variables independientes: edad, género, Charlson, antecedentes de insuficiencia cardíaca, AU, creatinina, proteinuria y filtrado glomerular-MDRD) sólo los niveles de AU (riesgo relativo: 1,35; 1,17-1,56, p = 0,000) se asociaban de forma independiente a la mortalidad. Conclusiones: en nuestro estudio, los niveles de AU se muestran como factor de riesgo independiente de mortalidad en ancianos. Palabras clave: Ácido úrico. Mortalidad. glomerular. Enfermedad renal crónica. Ancianos. Filtrado INTRODUCTION Correspondence: Manuel Heras Servicio de Nefrología. Hospital General de Segovia. 40002 Segovia. Spain. [email protected] [email protected] Uric acid (UA), a waste product of purine metabolism, is degraded by urate oxidase (uricase) to allantoin, which is freely eliminated in urine. One of the consequences of the 67 Manuel Heras et al. Uric acid and mortality originals absence of uricase in humans is the appearance of higher levels of UA in humans than in other species. It may even reach plasma UA concentrations fifty times higher than in other mammals. This condition, far from being a drawback, has been postulated to be an evolutionary advance related to the protective ability of UA against oxidative damage of free radicals.1,2 Hyperuricaemia is generally defined by UA levels >6.5mg/dl or 7mg/dl in men and >6mg/dl in women.3 Although recent epidemiological studies have focused on gout, which has been increasing in prevalence particularly among elderly individuals, the studies suggest that the incidence of hyperuricaemia has also increased during this time.4 In addition to the role of UA in the onset of rheumatism and gout, it has been postulated for some time that it is a possible determinant of the onset of hypertension (AHT), diabetes mellitus and chronic kidney disease (CKD).2,5 In recent years, there has been growing evidence of a relationship between high levels of UA in blood and renal and cardiovascular disease, endothelial lesions being the proposed pathogenic mechanism.2,6 Prospective epidemiological studies have shown the association between baseline levels of UA and the incidence of CKD (with increasing risk as UA levels increase).7,8 Regarding cardiovascular comorbidity, some studies found an association between UA and increased risk of heart attacks and coronary ischaemic strokes. However, other studies have not confirmed these results in multivariate analyses.2 On the other hand, some studies have shown that UA levels behave as a risk factor for both cardiovascular and overall mortality.9 In this study, we analysed the role of UA as an overall mortality marker in a cohort of elderly patients monitored for a period of 5 years. We also examined the association between baseline UA levels and cardiovascular history, the use of diuretics and renal function (RF). and General Nephrology departments during the period January-April 2006. Patients were in a period of clinical stability when they were selected and were monitored prospectively for 5 years (re-evaluation between JanuaryApril 2011). These patients had a mean age of 82.4±6 years (range 69-97 years) at baseline recruitment. Of these, 68.8% were women, 82.5% had histories of AHT, 35% were diabetic, 19.5% had histories of heart failure (HF), 15% had histories of ischaemic heart disease (IHD) and 68.4% were receiving diuretic therapy. Laboratory analysis A baseline analysis was performed one week before patients attended scheduled visits at the Geriatrics and Nephrology departments. According to the standard procedures of our hospital’s laboratory, we measured creatinine, UA, albumin, cholesterol and triglycerides in venous blood. We also analysed creatinine and proteinuria in 24-hour urine (Nephrology visits) or in first morning urine (Geriatrics visits). Methods This was a prospective observational study. Glomerular filtration rate (GFR) was calculated with the abbreviated MDRD formula.13 Based on the median baseline UA, we established two study groups: Group 1 consisting of 40 patients with UA <5.85mg/dl, and group 2 consisting of 40 patients with UA >5.85mg/dl. We also examined the possible association between high levels of UA (P75) and demographic characteristics, RF and mortality. After five years, we analysed mortality according to study group. Statistics Statistical analysis was performed using the SPSS 15.0 program. Data are expressed as mean and standard deviation, or median and percentiles. Comparison of means was made with the Student’s t-test and comparison of proportions with the chi-squared test. To analyse the simultaneous effect of several variables on mortality, we used a Cox analysis. PATIENTS AND METHODS RESULTS Patients This study was performed by analysing 5-year outcomes for a cohort of patients included in a study of elderly patients with CKD10,11,12 at the General Hospital of Segovia. Patients were recruited randomly while they visited the Geriatrics 68 UA showed a normal distribution. Mean baseline levels of UA were 6.11±1.7mg/dl (range 2.8-11) and their median was 5.85mg/dl (P25=4.90mg/dl, P75=7.07mg/dl). Regarding the use of diuretics, 42.6% used loop diuretics and 29.5% used thiazides. Potassium-sparing diuretics were the least Nefrologia 2012;32(1):67-72 Manuel Heras et al. Uric acid and mortality originals prescribed, at a rate of 11.5%. Table 1 shows the association between uric acid levels and gender, cardiovascular history and use of diuretics. Patients with histories of HF had significantly higher levels of UA (7.00±1.74mg/dl vs 5.90±1.71mg/dl, P=.031). Table 2 shows the comparison between demographic characteristics, RF parameters, comorbidity and mortality at 5 years, according to median UA. Patients with UA higher than the median had significantly lower GFR and higher mortality at 5 years. Twenty patients had UA greater than P75 (7.07mg/dl). Table 3 compares the variables according to this percentile. Table 1. Baseline levels of uric acid (mg/dl) according to gender, cardiovascular history and use of diuretics No Yes P Gender (men/women) 6.51 (1.55) 5.93 (1.78) 0.167 AHT 5.87 (1.81) 6.16 (1.72) 0.56 DM 6.25 (1.85) 5.84 (1.53) 0.314 HF 5.90 (1.71) 7.00 (1.74) 0.031 IHD 5.96 (1.66) 6.93 (1.94) 0.075 Diuretics 6.00 (1.84) 6.17 (1.70) 0.70 Loop diuretics 5.80 (1.53) 6.36 (1.80) 0.20 IHD: Ischemic heart disease; DM: diabetes mellitus; HF: heart failure; AHT: Hypertension. There were no patients lost to follow-up that were not due to death. Overall, 41 patients died during the 5-year follow-up: 15 due to general deterioration, 8 due to infections, 4 due to stroke, 4 due to tumours, 3 due to cardiovascular problems, 2 due to fracture complications and 5 due to unknown reasons. In the multivariate Cox analysis, the only predictor of overall mortality after adjusting for age, gender, Charlson score, history of HF, use of diuretic and creatinine, presence of proteinuria and GFR-MDRD was UA level (mg/dl) (relative risk 1.35; 1.17-1.56; P=.000). Figures 1A and 1B show the Kaplan-Meier mortality curves according to the median uric acid and P75. Both figures show significantly higher mortality in patients with UA greater than P50 and P75. DISCUSSION Overall mortality in the patient group with UA levels higher than the median was significantly higher. Moreover, if we analyse patients with UA levels >P75 (7.07mg/dl), their mortality increases even further, reaching 80%, results that are consistent with other recent studies.9 In this cohort of longer-lived patients, the most frequent cause of mortality was progressive deterioration rather than cardiovascular disease, and baseline UA levels were found to be the only predictors of mortality in the Cox analysis. Hyperuricaemia has been linked to various diseases in humans.4 Gout is a disease that predominantly affects men, with an increase in prevalence in both genders as age increases. In our study, men also had higher levels of UA, although these differences were not significant, which may Table 2. Comparison of variables according to the median uric acid (5.85mg/dl) Group 1: UA <5.85 mg/dl n = 40 Group 2: UA >5.85 mg/dl n = 40 P Baseline uric acid (mg/dl) 4.77 (0.86) 7.45 (1.27) 0.000 Creatinine (mg/dl) 1.15 (0.45) 1.46 (0.51) 0.005 Albumin (g/dl) 4.01 (0.3) 3.92 (0.48) Not significant Cholesterol (mg/dl) 186.56 (31) 187.90 (36) Not significant Triglycerides (mg/dl) 100.56 (36) 114.38 (72) Not significant MDRD (ml/min) 56.94 (15) 45.60 (16) 0.001 Proteinuria (g/24 h) 0.09 (0.38) 0.27 (0.63) Not significant 81.55 (6) 83.42 (6) Not significant 10/30 15/25 Not significant Charlson index 1.62 (1) 1.32 (1.11) Not significant DEATH 5 years 32.5% 70% 0.001 Age (years) Gender (m/f) UA: uric acid; f: female; MDRD: Modification of Diet in Renal Disease; m: male. Nefrologia 2012;32(1):67-72 69 Manuel Heras et al. Uric acid and mortality originals Table 3. Comparison of variables according to P75 of uric acid (7.07mg/dl) Patients with UA <7.07 mg/dl (n = 60) Patients with UA >7.07 mg/dl (n = 20) P Creatinine (mg/dl) 1.16 (0.38) 1.75 (0.51) 0.000 Albumin (g/dl) 4.01 (0.32) 3.92 (0.4) Not significant Cholesterol (mg/dl) 186.80 (33) 188.43 (34) Not significant Triglycerides (mg/dl) 105.44 (58) 113.00 (52) Not significant MDRD (ml/min) 55.27 (14) 39.27 (17) 0.000 Age (years) 81.61 (6) 85.10 (6) 0.037 Gender (m/f) Charlson score 15/45 10/10 0.037 1.75 (1) 1.95 (1) Not significant Diuretics use (%) 69.5% 65% Not significant EXITUS 5 years 41.7% 80% 0.003 UA: uric acid; f: female; MDRD: Modification of Diet in Renal Disease; m: male. CKD has been associated with hyperuricaemia.2 In experimental studies in rats with hyperuricaemia, renal lesions included afferent arteriolopathy, mild interstitial fibrosis, glomerular hypertrophy and/or glomerulosclerosis.15 We also found in our study that elderly patients with greater levels of UA had significantly worse RF. This association may be valid in both directions, that is, the high levels of UA may be explained by renal ischaemia and reduced RF, and/or reduced GFR in patients with higher levels of UA may be due to the direct toxic effects of UA. Diuretics, widely used to treat AHT with the additional benefit of preventing HF episodes,16 increase UA by stimulating the reabsorption of sodium and urate in the proximal tubule.3 Moreover, the increase in UA has also been observed in conditions that are accompanied by hypoxia (obstructive pulmonary disease, congestive HF).17,18 Leyva et al first studied UA concentrations in patients with chronic HF and found an inverse relationship between UA levels and oxygenation, suggesting that damage from oxidative metabolism may play a role in the pathogenesis of HF.19 In the SHEP study, diuretics were demonstrated to reduce cardiovascular mortality in the elderly. However, a recent subanalysis found that cardioprotection was lower in those patients who had high levels of UA.20 increase associated with diuretics. Similarly, patients with history of IHD have higher levels of UA, although in this case it was not statistically significant, possibly due to the low number of patients who had this disease. Lastly, ischaemia determines an increase in xanthine oxidase, which leads to an increase in UA levels.3 Treatment with xanthine oxidase inhibitors, such as allopurinol, has shown to reduce cardiovascular complications after coronary bypass and in patients with dilated cardiomyopathy.21 1.0 Uric acid < 5.85 mg/dl > 5.85 mg/dl 0.8 Cumulated Survival be due to the fact that we treated postmenopausal women in whom estrogenic activity had ceased.14 0.6 0.4 0.2 0.0 In our study, we found that UA levels were similar among those who received diuretics and those who did not. Moreover, elderly patients with histories of HF, who generally require higher doses of diuretics, had significantly greater levels of UA. It is therefore conceivable that the increase in UA levels in elderly patients with histories of HF is showing the effect of a local ischaemia rather than the 70 0 500 1000 1500 2000 Follow-up Days P = 0.002 Figure 1. Kaplan-Meier mortality curve according to the median uric acid (P50). Nefrologia 2012;32(1):67-72 Manuel Heras et al. Uric acid and mortality originals REFERENCES 1.0 P75 Uric acid < 7.07 > 7.07 Cumulated Survival 0.8 0.6 0.4 0.2 0.0 0 500 1000 1500 2000 Follow-up Days P=.007 Figure 2. Kaplan-Meier mortality curve according to P75 of uric acid (P50). Recently, Goicoechea et al found that treatment with allopurinol in patients with chronic renal failure reduced the decline in RF.22 With the data from our study, we cannot confirm that there is a causal relationship between high levels of UA and mortality. A step forward in our study would have been to confirm whether the reduction in UA levels with allopurinol, or more recently with febuxostat (a new inhibitor of the xanthine oxidase),23 contributes to reduced mortality in these patients. An important prognostic factor of morbidity and mortality is the presence of proteinuria.24,25 In our follow-up study of RF in the elderly, the presence of proteinuria at 36 months was an independent factor of mortality.12 This did not occur in our analysis at five years, either proteinuria is included in the model as a qualitative variable (yes/no) or if its numerical value is used. This may be explained by the lack of proteinuria in the baseline sample and by the fact that the patients with higher proteinuria died in the first years of follow-up. In conclusion, our data show that UA is an independent risk factor for overall mortality. Conflicts of interest The authors have no potential conflicts of interest to declare. Nefrologia 2012;32(1):67-72 1. Oda M, Satta Y, Takenaka O, Takahata N. Loss of urate oxidase activity in hominoids and its evolutionary implications. Mol Biol Evol 2002;19(5):640-653. 2. Minguela JI, Hernando A, Gallardo I, Martínez I, García P, Muñoz RI, et al. La hiperuricemia como factor de riesgo cardiovascular y renal. Dial Transp 2011;32(2):57-61. 3. Johnson RJ, Kang D-H, Feig D, Kivlighn S, Kanellis J, Watanabe S, et al. Is there a pathogenetic role for uric acid in hypertension and cardiovascular and renal disease. Hypertension 2003;41:1183-90. 4. So A, Thorens B. Uric Acid transport and disease. J Clin Invest 2010;120(6):1791-9. 5. Perlstein TS, Gumieniak O, Williams GH, Sparrow D, Vokonas PS, Gaziano M, et al. Uric acid and the development of hypertension: the normative aging study. Hypertension 2006;48(6):1031-6. 6. Waring WS, Webb DJ, Maxwell SRJ. Effect of local hyperuricemia on endothelial function in the human forearm vascular bed. Br J Clin Pharmacol 2000;49:511. 7. Obermayr RP, Temml C, Gutjahr G, Knechtelsdorfer M, Oberbauer R, Klauser-Braun R. Elevated uric acid increase the risk for kidney disease. J Am Soc Nephrol 2008;19:2407-13. 8. Ben-Dov IZ, Kark JD. Serum uric acid is a GFR-independent longterm predictor of acute and chronic renal insufficiency: the Jerusalem Lipid Research Clinic cohort study. Nephrol Dial Transplant 2011;26(8):2558-66. 9. Madero M, Sarnak MJ, Wang X, Greene T, Beck GJ, Kusek JW, et al. Uric acid and long term outcomes in CKD. Am J Kidney Dis 2009;53(5):796-803. 10. Heras M, Guerrero MT, Fernández-Reyes MJ, Sánchez R, Muñoz A, Macías MC, et al. Las manifestaciones analíticas asociadas a la insuficiencia renal crónica: ¿a partir de qué grado de filtrado glomerular las detectamos en ancianos? Rev Esp Geriatr Gerontol 2009;44(3):143-45. 11. Heras M, Fernández-Reyes MJ, Guerrero MT, Sánchez R, Muñoz A, Macias MC, et al. Ancianos con enfermedad renal crónica: ¿qué ocurre a los 24 meses de seguimiento? Nefrología 2009;4:343-9. 12. Heras M, Fernández-Reyes MJ, Guerrero MT, Sánchez R, Molina A, Rodríguez A, et al. Seguimiento durante 36 meses de la función renal en el anciano. Dial Transp 2011;32(3):97-101. 13. Levey AS, Greene T, Kusek JW, Beck GJ. Simplified equation to predict glomerular filtration rate from serum creatinine. J Am Soc Nephrol 2000;11:828(A). 14. Galvan AQ, Natali A, Baldi S, Frascerra S, Sanna G, Ciociaro D, et al. Effect of insulin on uric acid excretion in humans. Am J Physiol 1995;268:E1-E5. 15. Nakagawa T, Mazzali M, Kang DH, Kanellis J, Watanabe S, Sánchez-Lozada LG, et al. Hyperuricemia causes glomerular hypertrophy in the rat. Am J Nephrol 2003;23:2-7. 16. Heras M, Fernández-Reyes MJ, Sánchez R, Guerrero MT, Prado F, Alvarez-Ude F. Repercusión sobre la función renal de los fármacos antihipertensivos utilizados en ancianos con hipertensión arterial esencial. Hipertensión 2008;25(5):194-7. 17. Elsayed N, Nakashima J, Postlethwait E. Measurement of uric acid as a marker of oxigen tension in the lung. Arch Biochem Biophys 1993;302:228-32. 71 originals 18. Woolliscroft JO, Colfer H, Fox IH. Hyperuricemia in acute illness: a poor prognostic sign. Am J Med 1982;72:58-62. 19. Leyva F, Anker S, Swan JW, Godsland IF, Wingrove CS, Chua TP, et al. Serum uric acid as an index of impaired oxidative metabolism in chronic heart failure. Eur Heart J 1997;18:858-65. 20. Franse LV, Pahor M, Di Bari M, Shorr RI, Wan JY, Somes G, et al. Serum uric acid, diuretic treatment and risk of cardiovascular events in the Systolic Hypertension in the Elderly Program. J Hypertens 2000;18:1149-54. 21. Tabayashi K, Suzuki Y, Nagamine S, Ito Y, Sekino Y, Mohri H. A clinical trial of allopurinol (Zyloric) for myocardial protection. J Thorac Cardiovasc Surg 1991;101:713-18. Manuel Heras et al. Uric acid and mortality 22. Goicoechea M, García de Vinuesa S, Verdalles V, Ruiz-Caro C, Ampuero J, Rincón A, et al. Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin J Am Soc Nephrol 2010;5:1388-93. 23. Bruce SP. Febuxostat: a selective xanthine oxidase inhibitor for the treatment of hyperuricemia and gout. Ann Pharmacother 2006;40:2187-94. 24. García de Vinuesa S. Factores de progresión de la enfermedad renal crónica. Prevención secundaria. Nefrologia 2008;S3:17-21. 25. Agrawal V, Marinescu V, Agarwal M, Mc Cullough. Cardiovascular implications of proteinuria: an indicator of chronic kidney disease. Nat Rev Cardiol 2009;6(4):301-11. Sent for Review: 21 Jun. 2011 | Accepted: 1 Nov. 2011 72 Nefrologia 2012;32(1):67-72 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society originals Bone mineral density and bone metabolism in hemodialysis patients. Correlation with PTH, 25OHD3 and leptin A. Polymeris1, K. Doumouchtsis1, E. Grapsa2 1 2 Second Division of Endocrinology. Alexandra Hospital. Athens (Greece) Renal Unit. Alexandra Hospital. Athens (Greece) Nefrologia 2012;32(1):73-8 doi:10.3265/Nefrologia.pre2011.Jul.10916 ABSTRACT Background: Bone metabolism disorders in hemodialysed patients (HD) involve several humoral factors, of which PTH plays the central role. Leptin is usually found increased in renal failure and its link with bone metabolism has not been elucidated. We investigated the BMD and bone metabolism in association with serum PTH, 25OHD3 and leptin in HD patients. Methods: We measured bone alkaline phosphatase (bSAP), Cross linked N telopeptide of type 1 collagen (NTx), PTH, 25OHD3 and leptin in 37 HD patients. We also evaluated BMI and BMD in lumbar spine (LS) and in femoral neck (FN) by DXA. Statistical evaluations were based on simple regression analysis. Results: 1) Osteopenia was found in 32,1% in LS and 50% in FN and osteoporosis in 14.3% and 21.4% of our patients, respectively. LS or FN Z score was not related to HD duration. 2) Bone markers, PTH, phosphorus and leptin levels were increased. 3) 25OHD3 was low and was not related to NTx, bSAP or PTH. 4) PTH correlated with bone markers and Z score in LS and FN. 5) Leptin had no correlation with bone markers or Z score (except BMI). Conclusions: In our hemodialysed patients bone metabolism markers were increased in relation with high serum PTH levels. The observed high serum leptin was not associated with bone metabolism. Additionally the duration of hemodialysis did not appear to affect bone density. Keywords: Bone mineral density. Bone metabolism. Hemodialysis. Leptin. PTH. Vitamin D3. Densidad mineral y metabolismo óseo en pacientes en hemodiálisis. Correlación con la hormona paratifoidea, el 25(OH)D3 y la leptina RESUMEN Antecedentes: Los trastornos del metabolismo óseo en pacientes en hemodiálisis (HD) implican varios factores humorales, de los cuales la función central recae sobre la hormona paratiroidea. Cuando hay insuficiencia renal normalmente se detectan niveles elevados de leptina y su relación con el metabolismo óseo está aún por esclarecer. Investigamos la densidad mineral ósea (DMO) y el metabolismo óseo en relación con la hormona paratiroidea sérica, el 25(OH)D3 y la leptina en pacientes en HD. Métodos: Medimos la fosfatasa alcalina ósea (FAO), el telopéptido N, la hormona paratiroidea, el 25(OH)D3 y la leptina en 37 pacientes en HD. Asimismo, evaluamos el IMC y la DMO en la columna lumbar (CL) y en el cuello femoral (CF) mediante DXA. Las evaluaciones estadísticas se basaron en análisis de regresión simples. Entrecruzamiento del telopéptido N del colágeno óseo tipo I. Resultados: 1) De nuestros pacientes, el 32,1% presentaba osteopenia en CL y 50% en CF y el 14,3% y el 21,4% osteoporosis, respectivamente. El puntaje Z en CL o CF no estaba relacionado con la duración de la HD. 2) Los marcadores óseos, la hormona paratiroidea, y los niveles de fósforo y leptina se vieron incrementados. 3) El 25(OH)D3 era bajo y no estaba relacionado con el telopéptido N, la FAO o la hormona paratiroidea. 4) La hormona paratiroidea estaba correlacionada con los marcadores óseos y con el puntaje Z en CL y CF. 5) La leptina no presentaba correlación con los marcadores óseos o con el puntaje Z (con excepción del IMC). Conclusiones: En nuestros pacientes en hemodiálisis, los marcadores del metabolismo óseo se vieron incrementados en relación con los niveles elevados de hormona paratiroidea sérica. La elevada leptina sérica observada no estaba asociada al metabolismo óseo. Además, la duración de la hemodiálisis no pareció afectar a la densidad ósea. Palabras clave: Densidad mineral ósea. Metabolismo óseo. Hemodiálisis. Leptina. Hormona paratiroidea. Vitamina D3. INTRODUCTION Correspondence: Antonis Polymeris Second Division of Endocrinology. Alexandra Hospital. 20 Karditsis str. 15231. Athens. Greece. [email protected] [email protected] Chronic renal failure is almost always associated with alterations in mineral and bone metabolism.1,2 With the starting of haemodialysis (HD), histological signs of secondary hyperparathyroidism can be seen in bones in over 50% of patients.3 In end stage renal disease (ESRD) 73 originals patients, skeletal abnormalities known as Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD) (the so called renal osteodystrophy) include several types of bone tissue lesions, such as the most prevalent high bone turn over disease but also the adynamic bone disease.4,5 Other less frequent bone diseases such as osteomalacia, aluminum-related bone disease, fluorosis, strontium overload or mixed types have also been described3. A. Polymeris et al. BMD in HD. Relation to PTH, D3 and leptin derivative (one alpha) orally. None of the patients received vitamin K or aluminum hydroxide. There was no clinical or biological evidence for other bone disease such as osteomalacia or Paget’s disease. All patients were measured for BMD at lumbar spine and at femoral neck and for body weight and height and their body mass index (BMI) was calculated. Biochemistry It is well known that bone mineral density (BMD) is reduced in patients with chronic renal failure and they are at higher fracture risk6,7. Uremic patients usually exhibit high plasma intact PTH and high serum concentration of biochemical markers of bone metabolism such as bone specific alkaline phosphatase (bSAP) and collagen breakdown products.8 Some studies showed low 25OHD3 levels in uremic patients, which is probably implicated in CKD-MBD.9 Leptin, a hormone produced by fat tissue, decreases appetite and increases basal metabolic rate. Besides that, leptin induces in vitro stem cells differentiation to osteoblasts and reduces osteoclastogenesis, also having an in vivo positive effect on bone mass in mice10-13. Intracerebroventricular administration of leptin in wild or ob/ob leptin deficient mice resulted in bone loss14. In renal failure serum leptin levels are increased, as leptin is cleared by the kidneys15. In ESRD patients, in particular, the blood purification modality appears to affect leptin concentrations16. This fact might contribute to the development of CKD-MBD.17-19 Pre-midweek dialysis blood sampling was collected in the morning from the arteriovenus fistula after a 12h fast. The serum obtained after centrifugation was stored in aliquots at –20 oC until assayed, with measurements made immediately after thawing. Serum calcium, phosphorus, total protein, albumin, urea, creatinine, magnesium and total alkaline phosphatase were determined routinely using an automatic analyzer. Serum bSAP was measured by ELISA (Metra BAP EIA, sensitivity 0.7U/l, intra-assay variation 5.8%, inter-assay variation 5.2%) and serum NTx was also measured by ELISA (Wampole Laboratories, USA, intra-assay variation 4.6%, inter-assay variation 6.9%). Serum bioactive PTH and 25OHD3 were measured by chemiluminescence’s assay (Nichols advantage, functional sensitivity ≤4ng/ml, CV 20% for bioactive PTH and functional sensitivity ≤7ng/ml, CV 20% for 25OHD3). Serum leptin was measured by RIA (Linco Research, sensitivity 0.5ng/ml, intraassay variation 4.6%, inter-assay variation 5%). Bone mineral density The aim of current study is to investigate, the bone mass density and the bone metabolism in hemodialysed patients as estimated by serum markers of bone metabolism (bSAP and NTx) and to correlate with serum Ca, P, PTH, 25OHD3 and leptin. SUBJECTS AND METHODS Patients Thirty seven patients, 18 postmenopausal female and 19 male, on maintenance HD were included in the present study after their informed consent. The study was performed during the period April-May. Females were 43-73 years old, mean age 56.7 years and were on HD for 6-222 months with mean HD duration 68.1 months. Males were 41-79 years old, mean age 58 years and were on HD for 24-207 months with mean HD duration 67.1 months. All the patients were treated by conventional HD 4-5 hours, three times a week. None of the patients had a past history of parathyroidectomy or renal transplantation, of fracture or radiographic evidence of vertebral, rib or hip fracture. At the moment of the evaluation none of the patients, in particular the postmenopausal women, was receiving or had received previous to the study, oestrogen or raloxifene, calcitonin, bisphosphonates, PTH or corticosteroids. Thirteen patients received an active vitamin D 74 Bone mineral density (BMD) of the lumbar spine, total hip, femoral neck and trochanter were measured using Lunar DPX-L densitometer (Lunar, Madison, Wis, USA). All BMD measurements were performed by the same experienced operator. The densitometer was calibrated everyday with a standard phantom specimen. BMD results were obtained in absolute values (g/cm2), in T score and in Z score. T score is the number of standard deviations from the mean of a healthy young adult population (20-40 years old) and is used to determine osteoporosis or osteopenia. Z score is the number of standard deviations from the mean of a healthy age- and gender-matched normal population, which allows the comparison of BMD between patients of different age and gender. Osteoporosis was defined as a BMD T score at any site less than –2,5 and osteopenia as a BMD T score between –1 and –2,5. The reference values were obtained from an Italian normal population, similar to Greek normal population, provided by Lunar. Statistical analysis All results are shown as means±SD, unless otherwise indicated. Correlations between variables were assessed using simple linear regression and p<0.05 was accepted as Nefrologia 2012;32(1):73-8 A. Polymeris et al. BMD in HD. Relation to PTH, D3 and leptin statistically significant. Comparison of categorical variables was performed using chi-square analysis. Bone densinometric data The prevalence of osteoporosis (T score <–2.5) at lumbar spine and femoral neck was 14.3% and 21.4% respectively. The prevalence of osteopenia (T score between –1 and –2.5) at the same sites was 32.1% and 50% respectively. Lumbar spine Z score (m±SD) was –0.09±1.69 and femoral neck Z score was –0.76±1.14. RESULTS Demographic and biochemical data Table 1 depicts the demographic data for the 18 female patients and table 2 for the 19 male patients. Table 3 depicts the biochemical data for all 37 patients. Table 1. Demographic data in 18 female patients Mean±SD Range Age (years) 56.7±10.4 43-73 Duration of HD (months) 68.1±46.7 6-222 BMI (kg/m2) 24.8±4.82 18.9-39.1 SD: standard deviation; HD: hemodialysis; BMI: body mass index. Table 2. Demographic data in 19 male patients Mean±SD originals Range Age (years) 58.4±16.4 41-79 Duration of HD (months) 67.1±50.1 24-207 BMI (kg/m2) 24.1±3.01 19.8-31.7 SD: standard deviation; HD: hemodialysis; BMI: body mass index. Bone markers, PTH, 25OHD3 and leptin Serum levels of bone markers (NTx and bSAP) were very high (table 3). Serum PTH and phosphorus levels were also high as expected. Serum 25OHD3 levels were low and the prevalence of vitamin D deficiency was 89.2% among patients. Only 4 patients had normal 25OHD3 levels. Serum leptin was increased particularly in women (table 3). Correlation of BMD with PTH, leptin, duration of hemodialysis, body weight and BMI Lumbar spine and femoral neck Z score correlated significantly in a negative manner with serum PTH (p <0.025) (figure 1 and 2). Lumbar spine and femoral neck Z score was not related with serum leptin or with the duration of hemodialysis. We found that 10% of lumbar spine Z-score and 22.9% of femoral neck Z score variability is due to serum PTH levels. No correlation was found between body weight or BMI and Z score at any site. Table 3. Biochemical data in all patients Mean±SD Total protein (g/dl) Albumin (g/dl) Urea (mg/dl) Creatinine (mg/ml) Calcium (mg/dl) Phosphorus (mg/dl) Magnesium (mEq/l) Total alk phosphatase (U/l) PTH (pg/ml) 25OHD3 (ng/ml) Leptin (ng/ml) Leptin in females Leptin in males NTx (nMBCE/l) bSAP (U/l) Range Normal values 6.92±0.57 3.93±0.44 173.1±42.1 10.6±2.3 9.4±0.7 6.1±1.3 2.7±0.4 87.2±49.8 100.5±85.1 15.7±13.3 25.85±30.67 43.8±38.4 15.4±13 263.1±235.2 5.7-7.4 2.9-5.3 60-264 5-15.2 7.9-10.7 3.8-9 1.9-3.3 25-250 4-320.6 7.5-57 1.6-169.2 4.1-169.2 1.6-60.2 19.1-958 6.4-8.4 3.5-5 18-48 0.5-1.5 8.5-10.5 2.5-4.6 1.3-2.1 100-290 14-72 10-68 27.6±44.9 8.7-72.1 7.4±3.7 3.8±1.8 (Males) 5.4-24.2 (Females) 6.2-19 15-41 SD: standard deviation. Nefrologia 2012;32(1):73-8 75 A. Polymeris et al. BMD in HD. Relation to PTH, D3 and leptin originals Lumbar spine Z score that 48.5% of serum leptin variability is due to BMI. Leptin levels had no correlation with age or hemodialysis duration. Serum leptin had a positive correlation with serum creatinine (R=0.419; =0.012). R=0.398 p<0.025 DISCUSSION PTH Femoral neck Z score Figure 1. Lumbar spine Z score vs PTH. R=0.396 p<0.025 The data on the effect of hemodialysis in end stage renal disease patients on bone density are limited. To date most of the work has been focused on predialysis, HD and kidney transplanted patients and scarce information is available for patients on peritoneal dialysis, haemofiltration and haemodialfiltration.20-23 The prevalence of osteoporosis in hemodialysed patients is quite variable and depends on several factors including the method used for BMD measurement, the skeletal site and patients’ characteristics. Nonetheless, most of the studies showed reduced BMD in HD patients, which appears to be more pronounced than in peritoneal dialysis patients24. The prevalence of osteoporosis in HD patients has been estimated to be 13-29% at lumbar spine4,5,24 and 16-19% at femoral neck.4 Our findings are in accordance with these studies. Additionally, we found a high prevalence of osteopenia particularly at femoral neck. In CKD-MBD bone loss is site specific and in patients with PTH Figure 2. Femoral neck Z score vs PTH. Serum PTH correlated significantly in a positive manner with serum NTx and bSAP (R=0.715; p<0.001 and R=0.690; p<0.001 respectively) (figure 3 and 4). We found that 50% of serum NTx and bSAP variability is due to PTH levels. No correlation was found between serum PTH and 25OHD3 or leptin levels. NTx Correlation of PTH with bone markers, 25OHD3 and leptin R=0.715 p<0.001 PTH Figure 3. PTH vs NTx. Correlation of 25OHD3 with bone markers and leptin R=0.69 p<0.001 bSAP No correlation was found between 25OHD3, bone markers (NTx and bSAP) or leptin levels. Correlation of leptin with bone markers, BMI, body weight, age and duration of hemodialysis Serum leptin was not correlated with bone markers (NTx and bSAP). As expected, serum leptin levels were positively correlated with BMI (R=0.697; p<0.001) and with body weight (R=0.577; p<0.001) (figure 5 and 6). We also found 76 PTH Figure 4. PTH vs bSAP. Nefrologia 2012;32(1):73-8 A. Polymeris et al. BMD in HD. Relation to PTH, D3 and leptin Leptin R=0.697 p<0.01 BMI Figure 5. Leptin vs BMI. Body weight R=0.577 p<0.001 Leptin Figure 6. Leptin vs body weight. uremic hyperparathyroidism, PTH has a preferential effect on cortical bone.25,26 It has been reported that serum NTx is significantly higher in HD patients than in healthy individuals27 and appears to be the most reliable and useful bone resorption marker in renal osteodystrophy.28 In the present study we found extremely high levels of serum NTx and a significant positive correlation between serum NTx and PTH which is in line with previous published results.27,28 The clearance of bSAP is not performed by the kidney and serum bSAP concentration is therefore not affected by renal function. In our study a number of patients exhibited slightly increased serum bSAP. The significant positive correlation between serum bSAP and NTx with PTH probably suggests that the increased bone turnover is due to secondary hyperparathyroidism. Plasma 25OHD3 levels are often abnormally low in normal population, particularly in the elderly29 and in unselected medical inpatients,30 as well as in HD patients.31 In our cohort of patients there was also a high prevalence of vitamin D deficiency. Ghazali, et al. in their study found that low Nefrologia 2012;32(1):73-8 originals plasma 25OHD3 appears to be a major risk factor for hyperparathyroidism and Looser’s zones independent of calcitriol levels.9 Interestingly, and in contrast, we found no correlation between 25OHD3 and PTH or bone markers. Leptin, an adipocyte-derived hormone, is cleared by the kidney, and thus, plasma leptin levels are elevated in HD patients17. We also found increased serum leptin levels in our HD patients, particularly in women, which is in accordance with previous studies. It has been reported that there is a link between leptin and bone metabolism in vitro and in vivo experiments.10-12 Furthermore some studies in humans have shown a positive relationship between leptin and bone parameters,32-34 while others have not.35-37 In our study simple regression analysis showed no correlation between serum leptin and Z score at lumbar spine and femoral neck. Similarly, no correlation was found between leptin and bone markers (NTx and bSAP). Serum leptin showed a significant positive correlation only with BMI and body weight as expected. In conclusion the prevalence of osteoporosis/osteopenia is increased in HD patients and bone mineral density appears to correlate with high serum levels of bioactive PTH but not with 25OH vitamin D or high serum leptin. Interestingly, the duration of hemodialysis does not appear to affect bone density. The impact of leptin on bone metabolism in HD patients remains to be elucidated with further studies. REFERENCES 1. Hruska KA, Teitelbaum SL. Mechanisms of disease: renal osteodystrophy. N Engl J Med 1995;333(3):166-74. 2. Yamamoto N. Morphological analysis of bone dynamics and metabolic bone disease. Bone Histomorphometry in CKD-MBD (chronic kidney disease mineral bone disorder). Clin Calcium 2011;21(4):589-92. 3. De Vernejoul MC, Kuntz D, Miravet L, Gueris J, Bielakoff J, Ryckewaert A. Bone histomorphometry in hemodialysed patients. Metab Bone Dis Rel Res 1981;3:175-9. 4. Yamamoto N. Morphological analysis of bone dynamics and metabolic bone disease. Bone Histomorphometry: the basic methods and role of bone research and clinical significance. Clin Calcium 2011;21(4):529-33. 5. Sherrard D, Hercz G, Pei Y. The spectrum of bone disease in end stage renal failure-an evolving disorder. Kidney Int 1993;43(2):43642. 6. Taal M, Masud T, Green D, Cassidy M. Risk factors for reduced bone density in haemodialysis patients. Nephrol Dial Transplant 1999;14:1922-8. 7. Atsumi K, Kushida K, Yamazaki K, Shimizu S, Ohmura A, Inoue T. Risk factors for vertebral fractures in renal osteodystrophy. Am J Kidney Dis 1999;33(2):287-93. 8. Urena P, De Vernejoul MC. Circulating markers of bone remodeling in uremic patients. Kidney Int 1999;55:2141-56. 9. Ghazali A, Fardellone P, Pruna A, Atik A, Achard JM, Oprisiu R, et 77 originals A. Polymeris et al. BMD in HD. Relation to PTH, D3 and leptin al. Is low plasma 25-(OH)vitamin D a major risk factor for hyperparathyroidism and Looser’s zones independent of calcitriol? Kidney Int 1999;55(6):2169-77. 10. Thomas T, Gori F, Khosla S. Leptin acts on human marrow stromal cells to enhance differentiation to osteoblasts and to inhibit differentiation to adipocytes. Endocrinology 1999;140(4):1630-8. 11. Holloway WR, Collier FM, Aitken CJ. Leptin inhibits osteoclast generation. J Bone Miner Res 2002;17:200-9. 12. Tsuji K, Maeda T, Kawane T, Matsunuma A, Horiuchi N. Leptin stimulates fibroblast growth factor 23 expression in bone and supresses renal 1alpha 25-dihydroxyvitamin D3 synthesis in leptin deficient mice. J Bone Miner Res 2010;25(8):1711-23. 13. Steppan CM, Crawford DT, Chidsey-Fink KL, Ke H, Swick AG. Leptin is a potent stimulator of bone growth in ob/ob mice. Regul Pept 2000;92(1-3):73-8. 14. Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, et al. Leptin inhibits bone formation through a hypothalamic relay: a central role of bone mass. Cell 2000;100(2):197-207. 15. Song J, Li H, Zhang XD. Influence of diferent bpood purification treatment on the serum leptin and neuropeptide Y levels in patients with chronic renal failure. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2010;26(11):1116-8. 16. Taskapan MC, Taskapan H, Sahin I, Keskin L, Atmaca H, Ozyalin F. Serum leptin, resistin and lipid levels in patients with end stage renal failure with regard to dialysis modality. Ren Fail 2007;29(2):147-54. 17. Mantzoros CS. Leptin in renal failure. J Ren Nutr 1999;9(3):122-5. 18. Mallamaci F, Tripepi G, Zoccali C. Leptin in end stage renal disease (ESRD): a link between fat mass, bone and the cardiovascular system. J Nephrol 2005;18(4):464-8. Review. 19. Ghazali A, Grados F, Oprisiu R, Bunea D, Moriniere P, El Esper N, et al. Bone mineral density directly correlates with elevated serum leptin in haemodialysis patients. Nephrol Dial Transplant 2003;18:1882-90. 20. Zayour D, Daouk M, Medawar W, Salamoun M, El-Hajj Fuleihan G. Predictors of bone mineral density in patients on hemodialysis. Transplant Proc 2004;36(5):1297-301. 21. Urena P, Bernard-Poenaru O, Ostertag A. Bone mineral density, biochemical markers and skeletal fractures in haemodialysis patients. Nephrol Dial Transplant 2003;18:2325-31. 22. Doumouchtsis KK, Kostakis AI, Doumouchtsis SK, Tziamalis MP, Stathakis CP, Diamanti-Kandarakis E, et al. Associations between osteoprotegerin and femoral neck BMD in haemodialysis patients. J Bone Miner Metab 2008;26(1):66-72. Epub 2008 Jan 10. 23. Negri A, Álvarez-Quiroga M, Bravo M, Fradinger E, Jacob de Marino A. Estimation of the prevalence of low turnover renal osteodystrophy using biochemical markers in a peritoneal dialysis population. Nefrologia 2001;21(4):392-4. Fontaine M, Albert A, Dubois R, Saint-Remy A, Rorive G. Fracture and bone mineral density in hemodialysis patients. Clin Nephrol 2000;54:218-26. Yamaguchi T, Kanno E, Tsubota J, Shiomi T, Nakai M, Hattori S. Retrospective study on the usefulness of radius and lumbar bone density in the separation of hemodialysis patients with fractures from those without fractures. Bone 1996;19:549-55. Stein MS, Packham DK, Ebeling PR, Wark JD, Becker GJ. Prevalence and risk factors for osteopenia in dialysis patients. Am J Kidney Dis 1996;28:515-22. Montagnani A, Gonnelli S, Cepollaro C, Mangeri M, Martini S, Franci MB, et al. A new serum assay to measure N-terminal fragment of telopeptide of type 1 collagen in patients with renal osteodystrophy. Eur J Intern Med 2003;14:172-7. Nakashima A, Yorioka N, Mizutani T, Yamagata Z, Ueno T, Takasugi N. Serum cross-linked N-terminal telopeptide of type 1 collagen for evaluation of renal osteodystrophy in hemodialysis patients. Nephron Clin Pract 2005;99:c78-c85. Van der Wielen RP, Lowik MR, Van den Berg H, De Groot L, Haller J. Serum vitamin D concentrations in elderly in Europe. Lancet 1995;346:207-10. Thomas MK, Lloyd Jones DM, Thadam AJ, Shaw AC, Fikelstein R. Hypovitaminosis D in medical inpatients. N Engl J Med 1998;338:777-83. Bayard F, Bey P, Ton That M, Louvet J. Plasma 25OH cholecalciferol in chronic renal failure. Eur J Clin Invest 1973;3:447-50. Pasco JA, Henry MJ, Kotowicz MA, Collier GR, Ball MG. Serum leptin levels are associated with bone mass in nonobese women. J Clin Endocrinol Metab 2001;86(5):1884-7. Yamauchi M, Sugimoto T, Yamaguchi T, Naganuma S, Akiyama Y, Nuki Y, et al. Plasma leptin concentrations are associated with bone mineral density and the presence of vertebral fractures in postmenopausal women. Clin Endocrinol (Oxf) 2001;55(3):341-7. Blain H, Vuillemin A, Guillemin F, Durant R, Hanesse B, De Talance N, et al. Serum leptin levels is a predictor of bone mineral density in postmenopausal women. J Clin Endocrinol Metab 2002;879(3):1030-5. Rauch F, Blum WF, Klein K, Allolio B, Schonau E. Does leptin have an effect on bone in adult women? Calcif Tissue Int 1998;63:453-5. Odabasi E, Ozata M, Turan M, Bingol N, Yonem A, Cakir B, et al. Plasma leptin concentrations in post menopausal women with osteoporosis. Eur J Endocrinol 2000;142(2):170-3. Thomas T, Burguera B, Melton LJ 3rd, Atkinson EJ, O’Fallon WM, Riggs BL, Khosla S. Role of serum leptin, insulin and estrogen levels as potential mediators of the relationship between fat mass and bone mineral density in men versus women. Bone 2001;29(2):114-20. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. Sent for review: 28 Mar. 2011 | Accepted: 7 Jul. 2011 78 Nefrologia 2012;32(1):73-8 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society originals The lack of income is associated with reduced survival in chronic haemodialysis Sergio Marinovich1, Carlos Lavorato1, Guillermo Rosa-Diez1, Liliana Bisigniano2, Víctor Fernández2, Daniela Hansen-Krogh3 Comité de Estadísticas y Registros. Sociedad Argentina de Nefrología. Ciudad Autónoma de Buenos Aires (Argentina) Comisión Científico-Técnica. Instituto Central Único Coordinador de Ablación e Implante. Ciudad Autónoma de Buenos Aires (Argentina) 3 Departamento de Informática. Instituto Central Único Coordinador de Ablación e Implante. Ciudad Autónoma de Buenos Aires (Argentina) 1 2 Nefrologia 2012;32(1):79-88 doi:10.3265/Nefrologia.pre2011.Nov.11110 ABSTRACT La falta de ingresos económicos se asocia a menor supervivencia en hemodiálisis crónica Introduction: Poor socioeconomic status in the patient population is one of the causes of the lack of primary and secondary prevention of chronic kidney disease and negatively affects the survival of patients on chronic haemodialysis (HD). Objective: To confirm whether the low or absent income of the incident population on HD is a factor of poor prognosis. Methods: We used the incident HD population of the Argentine Registry of Chronic Dialysis. Follow-up lasted 12 months, performing an intention to treat analysis. We applied the Cox model to assess the association between income and survival of patients after adjusting for age, sex, diabetes, comorbidities, initial laboratory results, and first vascular access. Results: We analysed 13466 adult patients (age at onset: 60.4±15.6 years; 57.2% were male, and 39.2% diabetic) who were assigned to 2 groups: 1) «no income» group, 5661 patients (age at onset: 60.3±15.4 years; 53.1% were male and 41.4% diabetic), 2) «with income» group, 7805 patients (age at onset, 60.5±15-8] years; 60.1% were male and 37.5% diabetic). The «no income» group had a hazard ratio of 1.19 (95% confidence interval [CI]: 1.11-1.28) in the univariate analysis, 1.23 (95% CI: 1.14-1.32 ) considering age and gender, 1.22 (95% CI: 1.13-1.31) by adding diabetes mellitus, 1.26 (95% CI: 1.181.36) by adding comorbidities, 1.25 (95% CI: 1.16- 1.35) by adding the initial laboratory results, and 1.24 (95% CI: 1.151.33) if temporary vascular access is included. All models resulted in a significance of P=.000. Conclusions: Low or no income of patients at the time of entry into HD is an independent risk factor for immediate lower survival. RESUMEN Introducción: Las pobres condiciones socioeconómicas de la población es uno de los causales de la falta de prevención primaria y secundaria de la enfermedad renal crónica e influiría negativamente en la sobrevida de los pacientes en hemodiálisis (HD) crónica. Objetivo: Confirmar si el bajo o nulo ingreso económico de la población incidente en HD es un factor de mal pronóstico vital. Métodos: Utilizamos la población incidente en HD del Registro Argentino de Diálisis Crónica. El seguimiento fue de 12 meses, realizándose un análisis por intención de tratar. Se aplicó el Modelo de Cox para evaluar la asociación entre ingresos económicos y la sobrevida de los pacientes ajustando por edad, sexo, diabetes, comorbilidades, laboratorio inicial y primer acceso vascular. Resultados: Analizamos 13.466 pacientes adultos (edad al inicio: 60,4 [±15,6] años, 57,2% varones, 39,2% diabéticos) que fueron asignados a 2 grupos: 1) «Sin ingresos», 5.661 pacientes (edad al inicio: 60,3 [±15,4] años, 53,1% varones, 41,4% diabéticos); 2) «Con ingresos», 7.805 pacientes (edad al inicio: 60,5 [±15,8] años, 60,1% varones, 37,5% diabéticos). «Sin ingresos» mostró un hazard ratio de 1,19 (intervalo de confianza [IC] 95%: 1,11-1,28) en el univariado; de 1,23 (IC 95%: 1,14-1,32) considerando edad y género; de 1,22 (IC 95%: 1,13-1,31) agregando diabetes mellitus; de 1,26 (IC 95%: 1,18-1,36) agregando comorbilidades; de 1,25 (IC 95%: 1,16-1,35) adicionando laboratorio inicial y de 1,24 (IC 95%: 1,15-1,33) si se incluye acceso vascular transitorio. En todos los modelos la significación resultó en una p = 0,000. Conclusiones: Los bajos o nulos ingresos económicos del paciente en el momento del ingreso a HD es un factor de riesgo independiente de menor sobrevida inmediata. Keywords: Haemodialysis. Income level. Socioeconomics status. Survival. Palabras clave: Hemodiálisis. Nivel de ingresos. Estado socioeconómico. Sobrevida. Correspondence: Sergio Marinovich Comité de Estadísticas y Registros. Sociedad Argentina de Nefrología Superí, 370. 2000 Rosario. Argentina. [email protected] [email protected] INTRODUCTION The total population in Argentina reached 39 745 613 inhabitants in the year 2008, an approximate 1% population increase since 2004.1 The incidence rate of patients on 79 originals chronic dialysis (CD) increased by 4.38% during this time period, from 137.5 to 143.1 patients per million population (pmp), a 1.1% increase each year. The prevalence rate of CD increased by an even greater percentage (3.2% annual), increasing from 550.3pmp in 2004 to 623.4pmp in 2008.2,3 Chronic kidney disease (CKD) causes very high rates of morbidity and mortality, proportional to the CKD stage, with 30 times higher rates of cardiovascular mortality in stage V patients. This depends, among other factors, on the availability of access to renal replacement therapy and to a greater extent on access to primary and secondary kidney disease prevention measures in early stages of the disease. Sergio Marinovich et al. Income and survival in haemodialysis survival of 5360 new incident patients on chronic haemodialysis, giving a score of 1 for the final score obtained, where a higher score indicates a lower survival rate.5 Additionally, Abraham et al in India and Caskey et al in England-Wales found an association between poor socioeconomic conditions and a higher mortality rate on dialysis. In this last study, the authors observed a lower probability of transplant in the group with the greatest economic need.6,7 However, Eisenstein et al, in a study from the USA based on estimated income by housing area and race, found no significant differences in mortality rates while on haemodialysis among the different income level groups (low, middle, and high).8 It can be said that there are no restrictions on the availability of chronic dialysis treatment in Argentina. Any citizen that reaches a stage of terminal renal failure (TRF) with the need for dialysis is accepted by both public and private health centres. Even so, the low adjusted incidence rates of patients entering CD in some Argentine provinces lead us to strongly suspect that the health systems in these areas suffer some level of deficit in communication with chronic kidney disease patients.2,3 Consequently, it is quite probable that the totality of the Argentine population is not being contacted in time using the correct methods in order to carry out primary and secondary prevention measures for kidney disease. The aim of this study was to analyse whether a low or absent income in the incident population on chronic haemodialysis is a factor for a poor prognosis, adjusting for the variables that have been shown to influence 1-year survival. In addition to this inequality in the detection of kidney disease, over one third of the incident population on CD lives in poverty.2,3 Major epidemiological studies carried out at the national level have shown that poverty indicators are observed in between 30% and 33% of the general population. The study carried out by the Catholic University of Argentina (Universidad Catolica Argentina) also shows that 14.6% of homes have a severe housing deficit. As of 2009, 31.6% of homes had no seweage system, 9.8% were without running water, and 8.8% of households were overcrowded (a strong indicator in the “unsatisfied basic needs index”); additionally, 15.9% of homes have incomes that do not satisfy their basic food needs (10.8% moderate and 5.1% severe).4 Selection Life in situations where basic needs are not met, more commonly known as poverty, has not been examined thoroughly as a risk factor for mortality in the population of patients on CD using indexes or adequate multivariate studies, since, in order to understand its true effect, the socioeconomic variable must also be adjusted for other variables known to affect mortality in these patients. The few studies that have mentioned the poor conditions of CD patients suggest that these conditions have negative effects on survival, as well as on the probability of having access to a kidney transplant. By using a multivariate analysis to design a new prognostic index, we were able to demonstrate that insufficient income for patients and their family members was a variable of negative prognosis for the 1-year 80 METHODS Observational, retrospective, longitudinal and predictive analytical study. We used the data obtained through the National Registry of Chronic Dialysis. In Argentina, it is mandatory to register patients when they enter or exit a CD programme. When the patient is admitted into a programme, a questionnaire called the first registry of a chronic dialysis patient (primer ingreso de paciente a dialisis cronica, DRI) which, signed under oath by the CD Centre Director, must be submitted to the central office of the Registry under the Central Coordinating Institute for Ablation and Implants (Instituto Central Único Coordinador de Ablación e Implante, INCUCAI). The questionnaire includes demographic variables, pre-existing diseases, initial laboratory results, and socioeconomic and occupational variables from when the patient first started chronic dialysis treatment. This form must be completed by the attending nephrologists and social workers.9 Insufficient or absent income is very closely checked through the evaluations made by the social workers both through a review of paperwork and on-site evaluation of the living conditions at the patient’s home in order to corroborate or disprove the information provided. Our study included patients 18 years of age or older that started for the first time a chronic haemodialysis (HD) programme in any of the haemodialysis centres (n=462) in Argentina (24 provinces) between 1 April 2004 and 31 December 2008. The observation period came to a close on 31 December 2009, in order to guarantee a theoretical Nefrologia 2012;32(1):79-88 Sergio Marinovich et al. Income and survival in haemodialysis follow-up period of one year. We excluded all patients that did not have complete data for all variables considered in the study. The patients were censored in the case of recovered renal function, and event was defined as the death of a patient. As such, we performed an intention to treat analysis, with follow-up until patient death, whether or not the patient underwent a transplant. The start time for the follow-up period was day 1, and so all patients were included, even those with less than 90 days of follow-up. The population was divided into two groups: 1) “No income,” including patients that did not have any income declared, from neither the patients themselves or those who shared their home, at the start of treatment. 2) “With income,” including patients with declared income at the start of treatment. STATISTICAL METHOD originals background of myocardial infarction”, “presence of arterial hypertension”, “presence of cardiac arrhythmia”, “presence of congestive heart failure”, “presence of chronic pulmonary disease”, “presence or background of cerebrovascular disease (including dementia, hemiplegia)”, “presence of peripheral vascular disease (including amputated patients)”, “solid cancer without metastasis in the last 5 years (excludes basal/squamous skin cancer)”, “solid cancer with metastasis in the last 5 years”, “acute and chronic leukaemia”, and “lymphoma (includes myeloma according to the Charlson criteria)” (15 covariates). Model 4. In addition to the Model 3 variables, we added the following variables from the initial laboratory results: “positive HIVAb”, “positive HBsAg”, “positive HBVAb”, “initial albumin <3.5g/dl”, “initial haematocrit”, and “initial glomerular filtration rate” (glomerular filtration rate according to the abbreviated Levey formula MDRD-7)10 (21 covariates). Continuous variables were expressed as mean and standard deviation, and categorical variables were expressed in terms of frequency and proportion. For the comparison of numerical variables from both groups, we used Student’s ttests for parametric variables and Wilcoxon tests for nonparametric variables; we used Pearson’s chi-square test for qualitative variables, and considered a P-value <.05 to be statistically significant. Model 5. In addition to the Model 4 variables, we added “starting haemodialysis with a temporary vascular access (non-tunnelled catheter)” (22 covariates). In the survival analysis, we used the Kaplan-Meier (KM) method. We used the log-rank test (Mantel-Cox) for a simple comparison between the two populations. RESULTS We applied five different Cox proportional hazards models to test for differences between the two groups, adjusting for covariates that were considered to be predictive a priori based on previous studies.3,5 We determined hazard ratio (HR) or Exp (B) values for the independent variable “income” (dichotomous: no income and with income) after adjusting for the effect of other independent variables in the equation. The variable “income” was present in all models. In all cases, variables were considered as pre-existing conditions or conditions at the start of the therapy. We excluded those independent variables that were highly correlated with each other. The proportional hazard was calculated based on log (-log) survival. Model 1. “Age at start of treatment” and “Male” (3 covariates). Model 2. To the previous variables we added: “Diabetes mellitus as a cause of TRF” (4 covariates). Model 3. In addition to the previously mentioned variables, we added the following comorbidities as dichotomous variables (yes/no): “Presence of persistent angina or Nefrologia 2012;32(1):79-88 We performed all statistical analyses using SPSS version 15.0 statistical software for Windows (SPSS Inc., Chicago, IL). We evaluated a total of 13,466 patients. Table 1 summarises the characteristics of the study population. Upon starting HD, 56.7% of our patients were 60 years of age or older. Diabetes was present as a cause of TRF in 35.9% of the total, although 39.2% of the incident population was diabetic. The patients that reported no income within their household were 42.0% of the total. Overall mortality at 1 year was 22.3%, and 1-year survival was 77.7%. Only 1.8% of patients had to be censored from the study, with 245 recovering renal function. Table 2 displays general patient variables, comorbidities, laboratory results, type of initial vascular access, and the socioeconomic conditions for both groups and the corresponding statistical comparisons between them. The “no income” group had a significantly higher proportion of males, foreigners, people residing outside of Buenos Aires, and a similar mean age as compared to the other group. Upon analysis of the comorbidities present, although the “no income” group had a significantly higher percentage of diabetic patients, we found only one variable (a priori with a worse prognosis) that yielded a significantly higher percentage for this group: the presence of peripheral vascular disease. On the other hand, we observed a significantly lower percentage of angina and previous heart 81 Sergio Marinovich et al. Income and survival in haemodialysis originals Table 1. Basic patient characteristics Characteristics Values No. of patients 13466 Male 7698 (57.2%) Mean age (years) 60.4 (±15.6)a Age groups <50 years 3090 _>50<60 years 2710 _>60<70 years 3526 _>70<80 years 3075 _>80<90 years 1022 _>90 years 43 Causes of TRF Diabetic nephropathy 4839 (35.9%) Nephroangiosclerosis 2919 (21.7%) Unknown 2429 (18.0%) Glomerulonephritis 974 (7.2%) Obstructive nephropathy 695 (5.2%) Polycystic kidney disease 627 (4.7%) Chronic tubulointerstitial nephritis 191 (1.4%) Lupus nephritis 180 (1.3%) Multiple myeloma 94 (0.7%) Amyloidosis 47 (0.3%) Haemolytic uremic syndrome 44 (0.35) Other 427 (3.2%) Diabetes as a cause of TRF or lack of income 5272 (39.2%) No income 5661 (42.0%) With income 7805 (58.0%) Deaths 3002 (22.3%) Censored Completed the follow-up period 245 (1.8%) 10 219 (75.9%) attack, arrhythmia, chronic pulmonary disease, cancer, and tobacco use in the “no income” group. In terms of laboratory results, the “no income” patients started treatment with significantly worse conditions for almost all variables: lower glomerular filtration rate, lower albuminaemia, lower haematocrit, greater percentage of HBsAg positive, etc. Additionally, the use of a temporary non-tunnelled catheter was significantly more common in this group as the first vascular access point established. The variables associated with socioeconomic status were also significantly worse for the “no income” group: lower 82 level of education, greater instability in their living situation, and a larger number of people in each home (ratio of inhabitants/rooms), among others. The mortality rate after one year was significantly greater in the “no income” group: 24.2% vs 20.9%. We observed a very significant difference (P=.000) between the two groups in the number of patients that underwent transplants. In the “no income” group, 0.25% (14/5661) of patients underwent transplants, and in the “with income” group, 1.18% (92/7805). This difference was the basis for performing an intention to treat analysis, considering all transplant recipients with a maximum follow-up period of 12 months. The KM survival curves for the overall population and by group are displayed in Figure 1: total survival after one year was 77.71%. Survival in the “with income” group was significantly higher than in the “no income” group: 79.12% vs 75.76%, and the log-rank test (Mantel-Cox) resulted in a chi-square value of 22.49 (P=.000). To confirm these findings, we used a univariate Cox regression model that yielded an Exp (B) or hazard ratio (HR) of 1.19 for the “no income” group (95% confidence interval [CI]: 1.11-1.28). Considering other covariates in the equation, we will see whether the result is the same. Table 3 displays the HR for the variables included in each of the five multivariate regression models used, with special emphasis on the variable “no income.” Figure 2 shows the Cox predictive curves for models 3 and 5 for a typical individual (theoretical predictive survival) after adjusting for all covariates for the categorical variable of “with or without income.” The HR for the “no income” group did not change significantly in any of the five multivariate models. It did improve compared to the univariate model, although this difference did not reach statistical significance. The presence of comorbidities in model 3 produced the highest HR value (1.26), which decreased when the initial laboratory analysis values were added in model 4, and even more so when adjusting for vascular access in model 5. However, the differences are small, which implies that whether with or without adjustment, the lack of income for patients on HD is an immediate factor for a poor prognosis, regardless of the presence of other factors. Additionally, we performed a sixth Cox multiple regression model, which considered 4 variables: “no education or only incomplete primary school, “unstable living situation”, “male sex”, and “age at the start of treatment”. The two first variables did not enter into models 1 and 5 because of their close correlation with “no income”; for the same reason, “no income” was not evaluated in this sixth model. “Unstable living situation” had an HR of 1.38 (95% CI: 1.21-1.58; Nefrologia 2012;32(1):79-88 Sergio Marinovich et al. Income and survival in haemodialysis originals Table 2. General variables, comorbidities, initial laboratory results, and socioeconomic conditions of each group Variables No income With income P (n=5661) (n=7805) 60.32 (±15.4) 60.53 (±15.8) 0.440 Male (%) 53.1 60.1 0.000 Foreign-born (%) 6.7 5.7 0.017 Does not reside in the city of Buenos Aires (%) 96.6 89.0 0.000 General Age at start (years) Comorbidities (%) Previous myocardial infarction or persistent angina 9.1 12.1 0.000 Arterial hypertension 86.4 84.8 0.306 Cardiac arrhythmia 9.0 11.5 0.000 Congestive heart failure 22.8 22.9 0.930 Chronic pulmonary disease 6.4 7.7 0.005 Diabetes Mellitus 41.4 37.5 0.000 Cerebrovascular disease 7.7 7.4 0.545 Peripheral vascular disease 24.9 22.6 0.002 Solid cancer without metastasis in the last 5 years 3.3 5.3 0.000 Solid cancer with metastasis in the last 5 years 0.18 0.56 0.000 Acute or chronic leukaemia 0.07 0.12 0.410 Lymphoma (includes multiple myeloma) 0.79 1.09 0.085 Tobacco use in the last 5 years 16.0 17.8 0.007 Creatinemia (mg/dl) 7.81 (±4.20) 7.54 (±3.85) 0.000 Glomerular filtration rate (ml/m) 8.48 (±4.17) 8.90 (±4.32) 0.000 0.001 Initial laboratory results and vascular access Glomerular filtration rate > _15ml/m (%) Albuminaemia (g/dl) Albuminaemia <3.5g/dl (%) Haematocrit 7.1 8.6 3.35 (±0.62) 3.43 (±0.60) 0.000 54.1 48.8 0.000 26.49 (±5.26) 27.14 (±5.40) 0.000 Haematocrit <30 % (%) 71.9 69.1 0.000 Positive HIVAb (%) 0.35 0.36 0.958 Positive HBsAg (%) 0.79 0.42 0.005 Positive HCVAb (%) 1.97 1.67 0.202 Started haemodialysis with a temporary catheter (%) 64.2 60.2 0.000 No education or incomplete primary school (%) 34.1 18.9 0.000 Precarious household (not made with concrete or strong materials) (%) 11.5 5.7 0.000 Socioeconomic Number of rooms per home 2.65 (±1.19) 2.86 (±1.26) 0.000 Number of inhabitants per home 3.40 (±2.36) 3.32 (±2.27) 0.038 Ratio of inhabitants/rooms 1.40 (±0.87) 1.27 (±0.78) 0.000 91.3 96.1 0.000 Bathrooms installed in home (%) Water pipes (%) 92.0 96.5 0.000 First-year mortality (%) 24.2 20.9 0.000 Transplant recipients within the first year (%) 0.25 1.18 0.000 Recovery of renal function (%) 1.82 1.82 1.000 Mean values for numerical variables with corresponding standard deviation (±); %: percentage of patients in each dichotomous variable. P=.000). “No education or only incomplete primary school” had an HR of 1.09 (95% CI: 1.00-1.18; P=.046). This is simply another manner of evaluating poverty-indigence and its Nefrologia 2012;32(1):79-88 repercussions on immediate survival on HD: the absence of an adequate home significantly increased the relative risk of death by 38%, after adjusting for age, sex, and level of education. 83 Sergio Marinovich et al. Income and survival in haemodialysis originals 1.00 1.00 0.95 Total population Cumulative survival Cumulative survival 0.95 0.90 0.85 0.80 With income No income 0.90 0.85 0.80 Log rank (Mantel-Cox) Chi-square 22.49 (P = .000) Months on haemodialysis 0.75 0 13.466 3 12.063 6 11.267 9 10.678 Months on haemodialysis 0.75 12 0 10.219 With income7.805 No income 5.661 3 7.058 5005 6 6.628 4.639 9 6.305 4.373 12 6.033 4.186 Figure 1. Kaplan-Meier survival curves. Left: total population. Right: by group: “with income” and “no income.” The numbers below each curve show the number of patients that continued haemodialysis treatment each year. . DISCUSSION A home is considered to be poor if the “per capita” income is insufficient to cover the basic needs for food and other amenities (total basic needs) of its inhabitants, including clothing, education, transportation, and health. Additionally, if the home does not have sufficient income to provide adequate sustenance to its members (basic dietary needs), the inhabitants are considered to be indigent or extremely poor. We cannot consider the declarations of patient income to be absolutely correct; however, this declaration was certified by on-site evaluations in plain view of the living situations in the majority of cases where patients declared that they did not have any income in their households. We have observed in this study that the differences in education and households are notable between those that declare income and those that do not. This variable, therefore, is strongly associated with indigence or poverty. It is quite probable that patients that do not declare income do receive some type of economic subsidy after starting renal replacement therapy. However, this cannot be determined here for lack of data on the subject. Even so, we could observe in our study that the relative risk of death in the first year of hemodialysis treatment was significantly higher (19% to 26%) in patients that declared no income as compared to those that did declare an income. Diabetes mellitus is a poverty-linked disease; as Table 2 shows, this condition was significantly more prevalent in the “no income” group. Confirming the results from our study, Caskey et al in England and Wales observed that diabetes was a cause of TRF at a much higher rate in the population 84 living in areas of greater social deprivation.7 Also, Lorenzo et al performed an evaluation of the high prevalence of diabetes in the Canary Islands (Spain), and found that, among other causes, poverty and social inequality were causative forces for reduced use of health resources, lower rates of compliance with treatment, and inadequate hygienediet habits, leading to increased prevalence of diabetes mellitus in predisposed individuals.11 The initial laboratory results and a greater frequency of non-tunnelled temporary catheters as the first vascular access point provided showed that “no income” patients had worse conditions than “with income” patients, all of which is probably related to delayed contact with the health system. The location of the patient’s residence is also an influential factor: the city of Buenos Aires (not including the outlying urban areas) has been considered in other studies2,3 as the “gold standard” for renal health in Argentina, since it has the best indicators in the entire country: more advanced age upon starting a CD programme (which implies a better treatment of kidney disease and consequent delay in reaching stage 5 TRF), lower incidence rate of CD (due to better prevention methods), a higher rate of kidney transplant, and a better adjusted survival rate of patients on CD. Buenos Aires has the best health infrastructure in Argentina, especially for poor and indigent patients. In our study, we have found that this city is one of the four districts that have the lowest rates of “no income” patients in the entire country, 18.2%, significantly lower than the national mean of 42%. Observing a map of Argentina (Figure 3), we can see major inequality between the different provinces in income in the homes of incident patients on HD. The majority of the northNefrologia 2012;32(1):79-88 Sergio Marinovich et al. Income and survival in haemodialysis originals Table 3. Multivariate Cox proportional hazards models Variables Model 1 Model 2 Model 3 Model 4 Model 5 Age at start (per each year) 1.04 (1.04-1.05) 1.04 (1.04-1.05) 1.04 (1.04-1.04) 1.04 (1.04-1.05) 1.04 (1.04-1.04) Male 1.05 (0.97-1.13) 1.07 (0.99-1.15) 1.02 (0.94-1.09) 1.03 (0.95-1.11) 1.03 (0.95-1.11) No income 1.23 (1.14-1.32) 1.22 (1.13-1.31) 1.26 (1.18-1.36) 1.25 (1.16-1.35) 1.24 (1.15-1.33) P=.000 P=.000 P=.000 P=.000 P=.000 * 1.36 (1.27-1.46) 1.31 (1.21-1.42) 1.24 (1.14-1.35) 1.22 (1.12-1.32) Diabetes Mellitus Previous myocardial infarction/angina * * 1.04 (0.93-1.16) 1.07 (0.96-1.19) 1.06 (0.95-1.17) Arterial hypertension * * 0.70 (0.63-0.77) 0.72 (0.64-0.79) 0.75 (0.67-0.83) Cardiac arrhythmia * * 1.16 (1.05-1.29) 1.19 (1.07-1.32) 1.19 (1.08-1.32) Congestive heart failure * * 1.30 (1.19-1.41) 1.27 (1.17-1.38) 1.22 (1.13-1.33) Chronic pulmonary disease * * 1.18 (1.04-1.33) 1.18 (1.05-1.34) 1.16 (1.02-1.31) Cerebrovascular disease * * 1.32 (1.18-1.49) 1.34 (1.20-1.51) 1.34 (1.19-1.51) Peripheral vascular disease * * 1.30 (1.19-1.42) 1.28 (1.17-1.39) 1.31 (1.20-1.43) Solid cancer without metastasis * * 2.07 (1.81-2.35) 2.11 (1.85-2.41) 2.12 (1.86-2.42) Solid cancer with metastasis * * 1.83 (1.20-2.79) 2.03 (1.33-3.10) 1.92 (1.26-2.94) Acute or chronic leukaemia * * 1.89 (0.85-4.23) 1.95 (0.87-4.37) 2.01 (0.90-4.48) Lymphoma * * 3.32 (2.61-4.23) 3.15 (2.48-4.01) 2.94 (2.31-3.74) Positive HIVAb * * * 2.35 (1.43-3.85) 2.19 (1.33-3.59) Positive HbsAg * * * 0.78 (0.46-1.32) 0.77 (0.46-1.30) Positive HCVAb * * * 1.34 (1.04-1.72) 1.35 (1.05-1.73) Albuminaemia less than 3.5 g/dl * * * 1.60 (1.48-1.72) 1.45 (1.34-1.57) Haematocrit (for each unit % more) * * * 0.99 (0.99-1.00) 1.00 (0.99-1.00) Glomerular filtration rate (per each ml/m) * * * 1.00 (1.00-1.01) 1.01 (1.00-1.02) Initial temporary vascular access * * * * 2.12 (1.95-2.31) Values expressed as HR (95% confidence interval); the variables/values that resulted significant in the models are in bold. *Variable not included in the model western provinces (Tucuman, Salta, and Jujuy), all of the north-eastern provinces (Formosa, Chaco, Corrientes, and Misiones), three of the central provinces (La Pampa, Buenos Aires, and Cordoba), and all of the Cuyo provinces (Mendoza, San Juan, and San Luis) have percentages greater than 40%. All other provinces have lower percentages, with the lowest observed in the southern provinces of Patagonia (Chubut, Santa Cruz, and Tierra del Fuego) and the city of Buenos Aires. Some 42% of the Argentinean total corresponds to the incident population on HD between 2004 and 2008. However, it is heartening to know that the percentage of patients with no income declared decreased over time from 61% of incident patients in 2004, to 52% in 2005, 46% in 2006, 35% in 2007, and finally 22% in 2008. “Lack of income” was demonstrated to be a predictive factor for low survival after one year in patients on HD, although the HR obtained, in spite of its significance (P=.000), appears to be quite low, and we would have expected to observe a value higher than 1.22-1.26. As an explanation for this “relatively low” HR value, we could hypothesise that the lack of economic resources is a reliable reflection of a Nefrologia 2012;32(1):79-88 lifestyle in which the basic needs are not met, and that probably the HR was not higher because, once attended by the health system, the patient moves from being very vulnerable to less vulnerable. Care is provided as soon as he/she starts haemodialysis: social workers are sent to the home, the poor living conditions are recognised, a better household and more food are acquired through the county, provincial, or national government aid systems, and some sort of economic subsidy is provided. That is to say, after starting renal replacement therapy, the patient is often the recipient of several actions that are not considered in our study, and that eventually change the status of the patient from one of very high risk to one of moderate risk in the “no income” population. Another finding from our study was the fact that the “no income” population had a significantly lower rate of access to kidney transplants than the other group during the first year on HD: 0.25% vs 1.18%. Both values are low, but the value for poor patients is much lower. The causes of this phenomenon are still unknown, but hypothetically, poor patients must go through much more bureaucratic processes (with a greater loss of time) in order to be included on the kidney waiting list. 85 Sergio Marinovich et al. Income and survival in haemodialysis 1.00 1.00 0.95 0.95 With income No income 0.90 Cumulative survival Cumulative survival originals 0.85 0.80 With income No income 0.90 0.85 0.80 Months on haemodialysis 0.75 Months on haemodialysis 0.75 0 3 6 9 12 0 3 6 9 12 Figure 2. Left: Cox predictive curves for Model 3 using the dichotomous variable of with or without income. Right: predictive curves for model 5, also for the dichotomous covariate of with or without income The quality of health care provided to renal patients, in particular renal replacement therapy, whether through dialysis or transplant, has been the subject of a large number of medical publications. Many authors have dealt with the subject, examining all of the different factors that influence the survival of these patients. However, very few studies have made the attempt to include and evaluate the incidence of socioeconomic factors (for the patient and family members) in the treatment results. In a literature review we observed that some of these studies have certain local and methodological factors that impede the possibility of generalising these results across other populations. The various methods used to measure poverty and low socioeconomic status, for instance, and the different responses of health systems to high dependency patients, such as those with renal failure, makes comparisons difficult. In the USA, a strong relationship was shown between a low socioeconomic status and a high mortality rate,12,13,14 whereas other researchers in the same country could not find these results.8 Abraham et al in southern India examined 558 CD patients, finding that mortality was higher at lower socioeconomic levels (especially among diabetics), as well as a lower level of access to kidney transplants; but the extremely poor access to treatment, the fact that these patients receive treatment at different institutions, and the young age of the prevalent patients does not allow us to make comparisons.6 The study by Caskey et al showed that low-income patients are referred for treatment late in the progression of the disease, have a lower probability of undergoing peritoneal dialysis, undergoing a kidney transplant, and reaching target haemoglobin and phosphorous values after one year of treatment, and have a 86 higher mortality rate under the age of 65 years. This lower survival rate remained significant even after adjusting for age, sex, and the underlying cause of the renal failure, but was corrected when adjusting for comorbidities. The authors inferred that these factors should be responsible for the difference. However, in this study, social deprivation was based solely on the characteristics of the area or neighbourhood where the patient was living, which is a completely indirect method with a high margin of error.7 Our study, which was based on the entire incident population in Argentina during a given period and used a survey taken upon starting HD, with the final result of survival after one year, has certain aspects that have not been explored previously. Our results reinforce the idea that, in addition to the absence of income, an unstable household and low level of education are both statistically significant independent risk factors. Our study did have some limitations: 1) Absence of races and ethnicities. Since colonial times, Argentina has been the site of substantial racial mixing, which was enhanced by the three large waves of immigrants that arrived here between 1880 and 1950. As such, it is difficult to establish patterns of race and ethnicity. The selfrecognised indigenous population was only 1.56% of the total population in 2004-2005.15 There are no official data for people of African heritage (Afro-Argentine), but this ethnicity is thought to make up only 0.03% of the total population. 2) The study is not reproducible in societies where the level of poverty and indigence is very low. It is, however, in other Latin American countries where access to renal replacement therapy is also universal and where Nefrologia 2012;32(1):79-88 Sergio Marinovich et al. Income and survival in haemodialysis Tucuman Misiones Entre Rios City of Buenos Aires Ne uq ue n Cordoba Rio Negro Relative frequencies (%) Tierra del Fuego Equal to or more than 50 45-49 40-44 35-39 30-34 Less than 30 Figure 3. Percentage of incident patients on CHD in 20022004 that declared no income (patients and house members). Distribution by Argentine province the patterns of poverty are similar to Argentina: in particular, Uruguay, Chile, and Brazil.16 It is quite probable that the indigent and poor people of Argentina have a very late contact with the health system, which could be due to a lack of information, the need to deal with their basic needs at all times, the lack of time to spend in long lines at public hospitals, or the lack of health centres and public hospitals close to their place of residence. For some or all of these reasons, these patients do not have access to a healthy life. The vulnerability of this population is extreme. This is not just an issue of renal health, or even public health: this is a social problem that has its roots in the highest circles of social health policy, and will not be solved or even improved until it is approached from this perspective. It will improve when the social, cultural, economic, and health status of the population improves, and when the level of inequality is reduced. High poverty indices were once the exclusive domain of developing countries, but in the economic crisis of the last few Nefrologia 2012;32(1):79-88 originals years, large portions of the population in developed countries have begun to enter these conditions or are about to. As such, these countries may do well to anticipate these changes by developing containment policies directed at the population with renal failure and a low socioeconomic level. In conclusion, “lack of income” was shown to be a predictive factor for low survival after one year on HD, after adjusting for age, sex, diabetes, comorbidities, albuminaemia, glomerular filtration rate, haematocrit, and the initial type of vascular access. We propose the consideration of this variable or another similar measure that represents low or absent income in the incident HD patient, adjusted for other factors, in order to adequately predict short-term survival. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. REFERENCES 1. Instituto Nacional de Estadística y Censos (INDEC). Proyecciones provinciales de población por sexo y grupos de edad 2001-2015. Serie Análisis Demográficos, vol. 31. Ministerio de Economía y Producción de la República Argentina-Secretaría de Política Económica, 2005. Available at: http://www.indec.gov.ar/nuevaweb/cuadros/2/proyecciones_provinciales_vol31.pdf (Accesed July 9, 2011). 2. Marinovich S, Lavorato C, Celia E, Bisignano L, Soratti M, Hansen Krogh D, et al. Registro Argentino de Diálisis Crónica SAN-INCUCAI 2008. Informe 2010. Nefrología Argentina 9, Suplemento 1 (parte 1). P.7-62. 2011. Available at: http://www.san.org.ar/docs/registros/dc/2008/REGISTRO_ARGENTINO_DC_2008_VERSION_COMPLETA.pdf (Accesed July 9, 2011). 3. Marinovich S, Lavorato C, Celia E, Bisignano L, Soratti M, Hansen Krogh D, et al. Registro Argentino de Diálisis Crónica 2008. Informe 2010. Nefrología Argentina 9, Suplemento 1 (parte 2). P.71-127. 2011. Available at http://www.san.org.ar/docs/registros/dc/2008/REGISTRO_ARGENTINO_DC_2008_VERSION_COMPLETA.pdf (Accesed July 9, 2011). 4. Barómetro de la Deuda Social Argentina. Universidad Católica Argentina, Número 6. Año 2010. Available at http://www.uca.edu.ar/index.php/site/index/es/universidad/investigacion/programa-observatorio-de-la-deuda-social-argentina/publicaciones/informes/nro-6—-201 0/ (Accesed July 9, 2011). 5. Marinovich S, Lavorato C, Moriñigo C, Celia E, Bisignano L, Soratti M, et al. A new prognostic index for one-year survival in incident hemodialysis patients. Int J Artif Organs 2010;33(10):689-99. 6. Abraham G, Jayaseelan T, Matthew M, Padma P, Saravanan AK, Lesley N, et al. Resource settings have a major influence on the outcome of maintenance hemodialysis patients in South India. Hemodial Int 2010;14(2):211-7. 87 originals 7. Caskey FJ, Roderick P, Steenkamp R, Nitsch D, Thomas K, Ansell D, et al. Social deprivation and survival on renal replacement therapy in England and Wales. Kidney Int 2006;70(12):2134-40. 8. Eisenstein EL, Sun JL, Anstrom KJ, Stafford JA, Szczech LA, Muhlbaier LH, et al. Do income level and race influence survival in patients receiving hemodialysis? Am J Med 2009;122(2):170-80. 9. SINTRA. Formulario 02-DRI. Available at: http://sintra.incucai.gov.ar/ (Accesed November 16, 2011). 10. Levey AS, Bosch JP, Breyer-Lewis J, Greene T, Rogers N, Roth A. A more accurate meted to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Ann Intern Med 1999;130:461-70. 11. Lorenzo V, Boronat M. La enfermedad renal asociada con diabetes en Islas Canarias: un problema de salud pública, de elevado sufrimiento humano y alto coste económico. Nefrologia 2010;30(4):381-4. Sergio Marinovich et al. Income and survival in haemodialysis 12. Port F, Wolfe R, Levin N. Income and survival in chronic dialysis patients. ASAIO Trans 1990;36:M154-M157. 13. Garg P, Diener-West M, Rowe NR. Income-based disparities in outcomes for patients with chronic kidney disease. Semin Nephrol 2001;21:377-85. 14. Sandhu GS, Khattak M, Rout P, Williams M, Gautam S, Baird B, et al. Social adaptability index: application and outcomes in a dialysis population. Nephrol Dial Transplant 2011;26(8):266774. 15. Instituto Nacional de Estadística y Censos (INDEC). Encuesta complementaria de pueblos indígenas 2004. Available at: http://www.indec.mecon.ar/ (Accesed July 9, 2011). 16. Lugon JR, Strogoff de Matos JP. Disparities in end-stage renal disease care in South America. Clin Nephrol 2010;74 Suppl 1:S66-71. Sent for review: 3 Ago. 2011 | Accepted: 1 Nov. 2011 88 Nefrologia 2012;32(1):79-88 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society originals Effect of Intranasal DDAVP in Prevention of Hypotension during Hemodialysis Seyed S. Beladi-Mousavi1, Marzieh Beladi-Mousavi2, Fatemeh Hayati1, Mehdi Talebzadeh1 1 2 Department of Internal Medicine, Faculty of Medicine. Jundishapur University of Medical Sciences. Ahvaz (Iran) Department of Chemistry, Islamic Azad University, Omidiyeh Branch, Omidiyeh (Iran) Nefrologia 2012;32(1):89-93 doi:10.3265/Nefrologia.pre2011.Nov.10967 ABSTRACT Efecto de la DDVAP intranasal en la prevención de la hipotensión durante la hemodiálisis Introduction: The development of intradialytic hypotension during hemodialysis (HD) in which fluid removal is the primary goal, contributes to the excessive morbidity that is associated with the dialysis procedure. Materials and Methods: In a double blinded clinical trial, we compared the possible effect of intranasal DDAVP with intranasal distilled water as a placebo in prevention of intradialytic hypotension (IDH) in patients with known symptomatic IDH. In the first month of the study, nasal spray of distill water were administrated 30 minutes before all HD session (Placebo Group, Group 1) and then after a 30-day washout period we were used intranasal DDAVP 30 minutes before HD session (Vasopressin Group, Group 2). Blood pressure was measured just before HD, two hours later and after termination of HD. A hypotensive episode was defined as a decline of systolic blood pressure of more than 10mm Hg. Results: In overall Seventeen patients (nine men, eight women; mean age, 47.5 years) with known symptomatic IDH were enrolled in the study. The kind of dialysis membranes, mean of blood flow rate, dialyzate flow rate and ultrafiltration rate were the same in both groups. Each group has 204 HD session (17 * 12). Hypotensive episode occurred 18 times (8.82%) in vasopressin group compared with 125 times (61.27%) in placebo group and there was a significant association between them (p=0.0001). In addition mean arterial blood pressure in vasopressin group was 80.77 and in placebo group was 73.92 and also there was a significant association (p=0.0001). The mean Kt/v in group 1 and 2 were 1.29 and 1.28 without any differences between them (p=0.896). Conclusion: These results indicate that Compared with placebo, Vasopressin is significantly associated with a decreased incidence of intradialytic hypotension episodes during hemodialysis. Introducción: La aparición de hipotensión intradialítica durante la hemodiálisis (HD) en la que el objetivo principal es la eliminación de fluidos, contribuye a una morbilidad excesiva que se asocia con la diálisis. Materiales y métodos: Mediante un ensayo clínico doble ciego, comparamos los posibles efectos de la DDAVP intranasal con los del agua destilada intranasal como placebo en la prevención de la hipotensión intradialítica (HID) en pacientes con HID sintomática diagnosticada. Durante el primer mes del estudio, la pulverización nasal de agua destilada se realizaba 30 minutos antes de todas las sesiones de HD (grupo de placebo, grupo 1) y luego, tras un periodo de reposo de 30 días, utilizamos DDVAP intranasal 30 minutos antes de las sesiones de HD (grupo vasopresina, grupo 2). La presión arterial se medía justo antes de la HD, dos horas después y una vez finalizada la HD. Se definió como episodio de hipotensión la caída de la presión arterial sistólica del más de 10 mmHg. Resultados: Se incluyó en el estudio un total de 17 pacientes (nueve hombres y ocho mujeres de 47,5 años de edad media) con HID sintomática diagnosticada. En ambos grupos, el tipo de membranas de diálisis, la media del flujo sanguíneo, la tasa del flujo dializado y la tasa de ultrafiltración eran los mismos. Ambos grupos se sometieron a 204 sesiones de HD (17 x 12). Los episodios de hipotensión sucedieron en 18 ocasiones (8,82%) en el grupo de vasopresina en comparación con las 125 ocasiones (61,27%) del grupo de placebo y hubo una relación significativa entre ellos (p=0,0001). Además, la presión arterial media en el grupo de vasopresina era de 80.77 y en el grupo de placebo era de 73,92 e igualmente se observó una asociación significativa (p=0,0001). La media Kt/v en el grupo 1 y el 2 fue de 1,29 y 1,28 sin diferencias entre ellos (p=0,896). Conclusión: Estos resultados indican que, en comparación con el placebo, la vasopresina está relacionada de forma significativa con una menor incidencia de los episodios de hipotensión intradialítica durante la hemodiálisis. Keywords: DDAVP. Ultrafiltration Intradialytic Hypotension. Palabras clave: DDAVP. Ultrafiltración. Hemodiálisis. Hipotensión intradialítica. Hemodialysis. Correspondence: Seyed S. Beladi-Mousavi Department of Internal Medicine, Faculty of Medicine, Jundishapur University of Medical Sciences, Ahvaz, Iran [email protected] [email protected] RESUMEN INTRODUCTION Although considerable technical improvements have gained since the introduction of hemodialysis in the early 1950’s 89 originals Seyed S. Beladi-Mousavi et al. DDAVP in Prevention of Hypotension during HD and life expectancy of patients with End stage renal disease (ESRD) has improved, there are some complications with different underlying mechanisms that commonly occur during hemodialysis (HD) including intradialytic hypotension (IDH), cramps, itching, nausea and vomiting, chest and back pain, headache, and fever and chills.1-4 IDH is an important side effect of HD and continues to be a leading problem, especially in the elderly and cardiovascularly compromised patients and it has a negative impact on health-related quality of life.5-7 In some patients, the development of IDH necessitates decrease of the blood flow rate in HD apparatus and in some times, even discontinuation of HD and therefore it is an important cause of under dialysis. On the other hand, in the patients that they are need to ultrafiltration during HD, the development of hypotention episodes contribute to discontinuation of ultrafiltration and in some times, even necessitate intravenous fluid replacement before they are able to leave the dialysis unit and therefore, this problem can cause volume overload and some other significant complication.7 Unfortunately the incidence of this problem is very high especially among patients that they are received ultrafiltration during dialysis. It is occurred in significant percent of ESRD patients during or immediately following HD and its incidence ranges from 15 to 50 percent of dialysis sessions.7,8 Although a number of Studies have been evaluated to decrease the incidence of IDH, however because of small number of comparative studies and conflicting results, there are no generally accepted guidelines for prevention of hypotension during hemodialysis.9-14 The aim of this study is evaluating the possible effect of intranasal DDAVP for prevention of IDH episodes during hemodialysis. MATERIAL AND METHODS In a cross sectional clinical trial from May 2010 to September 2010, the present double blinded study was performed on ESRD patients who underwent hemodialysis treatments at Imam hospital, Ahvaz, Iran. The ESRD was defined as permanent and irreversible loss of renal function due to any causes with creatinine clearance of less than 10-15ml/min per 1.73m2 requiring renal replacement therapy. A standardized questionnaire was used to collect general information such as age, gender, the record of previous diseases and drugs, vital signs, causes of ESRD, date of onset of HD and length of time receiving HD services. 90 HD patients with known symptomatic episode of IDH in at least 30% of HD session in the 30 days preceding enrollment were included and those with the following characteristics were excluded from the study. Patients who had used antihypertensive drugs in recent two weeks, those that they didn’t need to ultrafiltration during HD, Patients who had used other preventive measure for prevention of hypotention during HD such as cold dialysate, midodrine and others, those with a history of MI in 6 weeks ago, anemic patients with hemoglobin level less than 10 gr/dl, and Patients who were suffered from gastrointestinal bleeding during HD. The study was explained to the subjects and all participants provided written informed consent. The study has approved by the Research Center of Ahvaz Joundishapur University of Medical Sciences. After selection of patients, in the first month of the study, all participants were received nasal spray of distill water (two puffs) 30 minutes before all HD session (Placebo group, Group 1) and then after a 30-day washout period we were used intranasal DDAVP (two puffs) 30 minutes before all HD session (Vasopressin group, Group, 2). Blood pressure was measured and recorded by a trained neurse, just before the needles for dialysis access were placed, 2 hour after starting and at the end of each HD session. Mean arterial blood pressure (MABP) was also calculated as the diastolic pressure plus one-third of the pulse pressure. For measurement of blood pressure, the patients were seated for at least 5 minutes and arm supported at heart level. We were used a manual aneroid sphygmomanometer with an appropriate cuff size so that the cuff bladder encircles at least 80% of the arm. An intradialytic hypotensive episode was defined as a fall in systolic blood pressure of at least 10 mm Hg two hour after starting and or after termination of HD compared to pre dialysis. Statistical analysis: For data analysis, we were used the statistical package for social sciences (SPSS) version 15 software. Chi-square tests or Fishers exact test were performed to evaluate the distribution of variables. Statistical significance was assessed at a probability level of < 0.05 in all analysis. Hemodialysis Methods Hemodialysis was performed for 9-12h, three times a week, using Fresenius machines, semi-synthetic (cellulose diacetate), or synthetic (polysulfone) dialyzer membranes, and bicarbonate- buffered dialysate (sodium 135mmol/l, potassium 2mmol/l, calcium 1.5mmol/l, magnesium 0.5 mmol/l and bicarbonate 35mmol/l). Blood flow rate was maintained from 250 to 400 mL/min, and the dialysate flow Nefrologia 2012;32(1):89-93 Seyed S. Beladi-Mousavi et al. DDAVP in Prevention of Hypotension during HD rate at 500ml/min. Dialysate temperature was 36.58C throughout the study period. Dry weight and rate of ultrafiltration was determined individually by the patient’s attending nephrologist on clinical grounds. KT/V KT/V was also calculated for the first and the end HD session in two groups. For evaluation of KT/V, blood sampling for blood urea nitrogen (BUN) was done immediately before HD session and for postdialysis BUN, our practice was to slow the blood pump to 100ml/min and then obtain the blood sample 15 seconds later. RESULTS One hundred twenty eight ESRD patients were on HD in Imam Hospital, Ahvaz, Iran. From them, one hundred eleven patients were not assessable because they had no hypotensive episodes and or because they had other exclusion criteria; therefore the study was performed on seventeen HD patients (nine men, eight women) with mean age of 47.5 years (range of 22 yr to 65 yr). The cause of ESRD in the patients on the study was diabetes mellitus in eight patients (four men and four women) and others were non diabetic (Hypertension, 5; Unknown, 3 and ADPKD, 1). In overall 408 HD performed in the period of the study; 204 times (17 X 12) in the placebo group (Group 1) and 204 times (17 X 12) in the vasopressin group (Group 2). The kind of hemodialysis machines, dialyzer membranes, dialysate were the same in the both groups. The mean rate of blood flow rate, dialysate flow rate and ultrafiltration rate among vasopressin and placebo group in each HD session were 300ml/mim and 290ml/min, 500ml/min and 500ml/min and 2.3 liters and 2.2 liters respectively without a significant difference between them. Hypotensive episode during HD occurred 18 times (8.82%) in vasopressin group compared with 125 times (61.27%) in placebo group and therefore the rate of IDH was significantly lower in vasopressin group (p=0.001). The mean of systolic and diastolic blood pressure after termination of HD session were 111.854 and 65.228 in vasopressin group and 102.671 and 59.550 in placebo group and there were a significant association between them (p=0.025 in mean of systolic and p=0.033 in mean of diastolic blood pressure). In addition, the mean of arterial blood pressure after termination of HD session in vasopressin group was 80.77 and in placebo group was 73.92 and also there was a significant difference between them (p=0.0001). The mean of Kt/v in group 1 and 2 were 1.297±0.217 and 1.290± 0.252 without any association between them (p=0.896). Nefrologia 2012;32(1):89-93 originals DISCUSSION The pathogenesis of hemodialysis hypotension is thought to be multifactorial, but generally results from inadequate cardiovascular compensatory mechanisms and impairs autonomic response to the aggressive reduction of circulating blood volume during ultrafiltration.15,16 In addition there are very strong arguments for an important role of several vasoactive substances such as adenosine (cardiodepressive and endogenous vasodilator) and nitric oxide (endogenous vasodilator) which may be synthesized or released during dialysis in pathogenesis of IDH.17,18 Although vasopressin is widely recognized for its role in the regulation of sodium balance and plasma osmolality, it is also a well-recognized vasoconstrictor and the role of vasopressin insufficiency as an important cause of IDH has also demonstrated in recent years by several observations.19-23 In the first time, the possible role of vasopressin insufficiency as a cause of hemodynamic instability during HD, showed by Friess et al in 1994. They measured plasma AVP level in 23 patients with recurrent dialysis hypotension and showed that AVP concentration only increased in six patients with nausea and hypotension and in the remaining 17 patients without nausea AVP level did not increased.19 In the setting of hypotension as an example in patients with septic shock, the release of vasopressin is usually increased and in together with other vasoconstrictors cause systemic vascular resistance and elevates blood pressure.23 It is therefore hypothesized that the inappropriately low vasopressin concentrations due to decreased endogenous AVP synthesis and or secretion may explain recurrent dialysis hypotension in the previous study. Sato et al and Cignareli et al in two separate study in nondialysis diabetic patients with severe diabetic neuropathy demonstrated that AVP concentrations do not appropriately increase in the setting of orthostatic hypotension.24,25 Therefore the results of two studies are supported the important effect of vasopressin in maintenance of blood pressure. In addition because of diabetes mellitus is the most common co morbidities associated with ESRD, the results of these studies also suggested that vasopressin insufficiency may be a part of the underlying mechanism of IDH in diabetic patients. Mira Rho et al also demonstrated vasopressin insufficiency as a possible mechanism of IDH in ESRD patients and supported the findings of Friess et al.20 They performed an observational pilot study on 20 chronic hemodialysis patients and observed that AVP concentration did not increase as would normally be expected in patients with symptomatic IDH in response to severe hypotension and therefore they have suggested that intravenous vasopressin 91 originals Seyed S. Beladi-Mousavi et al. DDAVP in Prevention of Hypotension during HD and perhaps intranasal vasopressin administration may improve hemodynamic stability in patients with symptomatic IDH. Although in our study we were not measured concentration of AVP in HD patients with IDH, however the data from this clinical trial are also supported the findings of the studies of Friess et al and Mira Rho et al. According to the our study, the use of two puffs of intranasal DDAVP 30 minutes before HD session was significantly associated with a decreased incidence of hypotensive episodes among patients that they are received ultrafiltration during dialysis. hypotension episodes among patients that they are received ultrafiltration hemodialysis. Although the results of our study are interesting but the small number of patients enrolled in the study is a limit factor and therefore further research with larger number of patients is needed to determine the effect of vasopressin administration for prevention of hypotension during HD. Acknowledgements Other than our study, there are few clinical trials that they have evaluated the possible effect of vasopressin in prevention of IDH episodes during hemodialysis. As an example, in a study Van der Zee et al measured plasma vasopressin concentration during HD and found that plasma vasopressin levels did not increase during ultrafiltration dialysis. Then they examined 22 ESRD patients in a randomized, double-blinded and placebo-controlled trial and showed that blood pressure was more stable in the patients receiving constant infusion of a non-pressor dose of vasopressin during hemodialysis and the incidence of symptomatic hypotensive episode was significantly lower in comparison to the placebo group. Finally, they concluded that administration of vasopressin improves cardiovascular stability and facilitates fluid removal during hemodialysis.21 This paper is issued from thesis of Dr. Mehdi Talebzadeh and financial support was provided by Ahvaz Joundishapur University of Medical Sciences. We would like to express our appreciation to the division head and the staff and of course ESRD patients in HD center of Imam Hospital in the province of Khuzestan, Ahvaz, Iran, for their help. Jills et al have also showed efficacy of vasopressin in prevention of hypotension during hemodialysis. In this double-blind crossover study, they were used intranasal lysine vasopressin in 6 patients with refractory hemodialysis-induced hypotension and have reported that Systolic, diastolic, and mean arterial blood pressures were more stable with use of vasopressin.22 1. Skroeder NR, Jacobson SH, Lins LE, Kjellstrand CM. Acute symptoms during and between hemodialysis: the relative role of speed, duration, and biocompatibility of dialysis. Artif Organs 1994;18:880. 2. Van der Sande FM, Kooman JP, Leunissen KM. Intradialytic hypotension—new concepts on an old problem. Nephrol Dial Transplant 2000;15:1746. 3. Milinkovic M, Zidverc-Trajkovic J, Sternic N, Trbojevic-Stankovic J, Maric I, Milic M, et al. Hemodialysis headache. Clin Nephrol 2009;71:158. 4. Zager PG, Nikolic J, Brown RH, Campbell MA, Hunt WC, Peterson D, et al. “U” curve association of blood pressure and mortality in hemodialysis patients. Kidney Int 1998;4:561-9. 5. Shoji T, Tsubakihara Y, Fujii M, Imai E. Hemodialysis-associated hypotension as an independent risk factor for two-year mortality in hemodialysis patients. Kidney Int 2004;66:1212-20. 6. Dasselaar JJ, Huisman RM, de Jong PE, Franssen CF. Measurement of relative blood volume changes during haemodialysis: merits and limitations. Nephrol Dial Transplant 2005;20(10):2043-9. 7. Kooman J, Basci A, Pizzarelli F, Canaud B, Haage P, Fouque D, et al. EBPG guideline on haemodynamic instability. Nephrol Dial Transplant 2007;22 Suppl 2:ii22-44. 8. Dheenan S, Henrich WL. Preventing dialysis hypotension: a comparison of usual protective maneuvers. Kidney Int 2001;59(3):117581. 9. Daugirdas JT. Preventing and managing hypotension. Semin Dial 1994;7:276-83. 10. Knoll GA, Grabowski JA, Dervin GF, O’Rourke K. A randomized, CONCLUSION Intradialytic hypotension (IDH) continues to be a leading problem in patients with ESRD and it has an important negative effect on health-related quality of life. Unfortunately, because of small number of comparative studies, there are no generally accepted guidelines for prevention of this problem. Some studies have showed that AVP concentration do not increase as would normally be expected in patients with symptomatic IDH. Therefore it is hypothesized that the inappropriately low vasopressin concentrations is a significant part of the underlying mechanism of IDH and perhaps intravenous vasopressin and or intranasal vasopressin administration may prevent hypotension during HD. According to the present clinical trial, the use of two puffs of intranasal DDAVP 30 minutes before HD session Compared with placebo is significantly associated with a decreased incidence of intradialytic 92 Conflict of interest The authors declare they have no potential conflicts of interest related to the contents of this article. REFERENCES Nefrologia 2012;32(1):89-93 Seyed S. Beladi-Mousavi et al. DDAVP in Prevention of Hypotension during HD 11. 12. 13. 14. 15. 16. 17. 18. controlled trial of albumin versus saline for the treatment of intradialytic hypotension. J Am Soc Nephrol 2004;15(2):487-92. Yu AW, Ing TS, Zabaneh RI, Jensen UB, Tryggvason K. Effect of dialysate temperature on central hemodynamics and urea kinetics. Kidney Int 1995;48:327-43. Prakash S, Garg AX, Heidenheim AP, House AA. Midodrine appears to be safe and effective for dialysis-induced hypotension: a systematic review. Nephrol Dial Transplant 2004;19:2553-8. Moret K, Aalten J, Wall Bake W, Gerlag P, Beerenhout C, van der Sande F, et al. The effect of sodium profiling and feedback technologies on plasma conductivity and ionic mass balance: a study in hypotension-prone dialysis patients. Nephrol Dial Transplant 2006;21:138-44. Donauer J. Hemodialysis-induced hypotension: impact of technologic advances. Semin Dial 2004;17:333-5. Daugirdas JT. Pathophysiology of dialysis hypotension: an update. Am J Kidney Dis 2001;38(4 suppl 4):S11-7. Leunissen KM, Kooman JP, van Kuijk W, van der Sande F, Luik AJ, van Hooff JP. Preventing haemodynamic instability in patients at risk for intra-dialytic hypotension. Nephrol Dial Transplant 1996;11 Suppl 2:11-5. Armengol NE CAA, Bono Illa M, Calls Ginesta J, Gaya Bertran J, Rivera Fillat DR. Vasoactive hormones in uraemic patients with chronic hypotension. Nephrol Dial Transplant 1997;12:321-4. Shoji T, Tsubakihara Y, Fujii M, Imai E. Hemodialysis-associated hypotension as an independent risk factor for two-year mortality in he- originals modialysis patients. Kidney Int 2004;66:1212-20. 19. Friess U, Rascher W, Ritz E, Gross P. Failure of arginine-vasopressin and other pressor hormones to increase in severe recurrent dialysis hypotension. Nephrol Dial Transplant 1995;10:1421-7. 20. Rho M, Perazella MA, Parikh CR, Peixoto AJ, Brewster UC. Serum Vasopressin Response in Patients with Intradialytic Hypotension: A Pilot Study. Clin J Am Soc Nephrol 2008;3(3):729-35. 21. Van der Zee S, Thompson A, Zimmerman R, Lin J, Huan Y, Braskett M, et al. Vasopressin administration facilitates fluid removal during hemodialysis. Kidney Int 2007;71:318-24. 22. Lindberg JS, Copley JB, Melton K, Wade CE, Abrams J, Goode D. Lysine Vasopressin in the Treatment of Refractory Hemodialysis-Induced Hypotension. Am J Nephrol 1990;10:269-75. 23. Holmes CL, Patel BM, Russell JA, Walley KR. Physiology of vasopressin relevant to management of septic shock. Chest 2001;120:989-1002. 24. Sato K, Kimura T, Ota K, Shoji M, Ohta M, Yamamoto T, et al. Changes in plasma vasopressin levels and cardiovascular function due to postural changes in diabetic neuropathy. Tohoku J Exp Med 1995;177:49-60. 25. Cignarelli M, De Pergola G, Paternostro A, Corso M, Cospite MR, Centaro GM, et al. Arginine-vasopressin response to supine-erect posture change: an index for evaluation of the integrity of the afferent component of baroregulatory system in diabetic neuropathy. Diabete Metab 1986;12:28-33. Sent for review:11 May. 2011 | Accepted: 1 Nov. 2011 Nefrologia 2012;32(1):89-93 93 originals http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society Echocardiographic impact of hydration status in dialysis patients Isabel Juan-García1, María J. Puchades1, Rafael Sanjuán2, Isidro Torregrosa1, Miguel Á. Solís1, Miguel González1, Marisa Blasco2, Antonio Martínez1, Alfonso Miguel1 1 2 Servicio de Nefrología. Hospital Clínico Universitario de Valencia. Spain Servicio de Medicina Intensiva. Hospital Clínico Universitario de Valencia. Spain Nefrologia 2012;32(1):94-102 doi:10.3265/Nefrologia.pre2011.Nov.10867 ABSTRACT Introduction: Cardiovascular disease is the main cause of death in Chronic Kidney Disease patients. Left ventricular hypertrophy is the most common manifestation and it is linked to arterial hypertension and overhydration. The goal of this paper is to stratify dialyzed patients according to hydration status and to make an evaluation about the possible echocardiography alterations of the different groups. Methods: a transversal study was carried out with 117 patients: 65 were on hemodialysis and 52 on peritoneal dialysis. We performed the following tests: multifrequency bioimpedance with the BCM-Body Composition Freesenius’ Monitor system, transthoracic echocardiography, and blood tests. If ECW/TBW (extracellular water vs total body water) normalization ratio for age and gender was > 2.5% SD, the patient was considered overhydrated. Results: HD patients are significantly overhydrated before HD (67.1%) compared to DP patients (46.1%), and almost half of the overhydrated population presents arterial hypertension. However, after an HD session, a better control of the hydration status is reached (26.1%). DP patients frequently present high arterial pressure and/or are under antihypertensive treatment (DP 76.9% vs HD 49.2%). Left ventricular hypertrophy is much more common in HD overhydrated patients, eccentric LVH being more prevalent. Overhydrated patients present significantly high values of LAVI, ILVM, OH/ECW. Conclusions: Bioimpedance technique allows for the detection of a large number of overhydrated patients. Echocardiographic alterations in dialyzed patients show a high correlation between the hydration stage by ECW/TBW normalized ratio for age and gender and the LAVI and ILVM. Keywords: Bioimpedance spectroscopy. Left arterial volumen index. Left ventricular hypertrophy. Volume status. Correspondence: Isabel Juan García Servicio de Nefrología. Hospital Clínico Universitario de Valencia. Avda. Blasco Ibáñez 17. Valencia. Spain. [email protected] 94 Repercusión ecocardiográfica del estado de hidratación en los pacientes en diálisis RESUMEN Introducción: La enfermedad cardiovascular es la principal causa de muerte en los pacientes con enfermedad renal crónica. La hipertrofia ventricular izquierda (HVI) es la manifestación más frecuente y está relacionada con la hipertensión arterial y la hiperhidratación. El objetivo del presente trabajo es estratificar a los pacientes en diálisis según el estado de hidratación y valorar las posibles alteraciones ecocardiográficas en los distintos grupos. Métodos: Realizamos un estudio transversal de 117 pacientes, 65 en hemodiálisis (HD) y 52 en diálisis peritoneal (DP). Las exploraciones realizadas fueron: bioimpedancia multifrecuencia con el sistema BCM-Body Composition Monitor de Freesenius, ecocardiografía transtorácica y analítica de sangre. Definimos hiperhidratación cuando el cociente volumen extracelular-volumen corporal total (ECW/TBW) normalizado para edad y sexo es > 2,5% de la desviación estándar. Resultados: Los pacientes en HD están pre-HD (67,1%) más hiperhidratados de forma significativa que los de DP (46,1%), presentando casi la mitad de la población hiperhidratada hipertensión arterial; tras la sesión de HD se consigue un mejor control del estado de hidratación (26,1%). Los pacientes en DP presentan con más frecuencia cifras de tensión arterial alta y/o llevan tratamiento antihipertensivo (DP 76,9 vs. HD 49,2%). La HVI es más frecuente en los pacientes en HD e hiperhidratados, siendo la más prevalente la HVI excéntrica. Los pacientes hiperhidratados presentan cifras superiores, de forma significativa, del IVAI (volumen de aurícula izquierda indexada por superficie corporal, la IMVI (masa ventricular izquierda indexada) y el cociente sobrehidratación-agua extracelular. Conclusiones: La bioimpedancia es una técnica que nos permite detectar un gran número de pacientes hiperhidratados. Al estudiar las alteraciones ecocardiográficas en los pacientes en diálisis encontramos una alta correlación entre el estado de hidratación por ECW/TBW normalizado para edad y sexo, y el IVAI e IMVI. Palabras clave: Bioimpedancia espectroscópica. Volumen de aurícula izquierda indexada. Hipertrofia de ventrículo izquierdo. Estado de hidratación. INTRODUCTION Cardiovascular disease is the main cause of death in patients with chronic kidney disease (CKD).1 This is usually due to Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients the presence of traditional risk factors, such as diabetes mellitus, hypertension (AHT), dyslipidaemia and advanced age,2 and to the actual kidney disease (overhydration, uraemic cardiomyopathy and vascular damage, i.e. atherosclerosis, vascular calcification and arterial stiffness). The main manifestations of cardiovascular disease in these patients are arterial vascular disease and cardiomyopathy.3 The high prevalence of cardiomyopathy is due to AHT and atherosclerosis, which create excess pressure and lead to the development of concentric left ventricular hypertrophy (LVH). Anaemia, fluid overload and arteriovenous fistulae create volume overload, which leads to left ventricular dilation with eccentric LVH.4,5,6 There are various methods for assessing hydration status. Assessment of the inferior vena cava and biochemical parameters, such as B-type natriuretic peptide, are useful methods for assessing intravascular hydration status,7 while bioimpedance spectroscopy (BIS) assesses body and extracellular hydration states.8 The latter technique is simple, inexpensive, reproducible, non-invasive and easy to apply, and is based on the human body’s resistance to alternating electrical currents. It assesses not only hydration status but also intracellular and extracellular water, the extracellular and intracellular ratio, the total water volume, as well as nutritional parameters. The aim of this study was to stratify patients on dialysis according to hydration status, and to assess possible echocardiographic abnormalities in the various groups according to dialysis technique. METHOD Patients We performed a cross-sectional observational study on 117 clinically stable patients on the dialysis programme of the University Clinical Hospital of Valencia between 2008 and 2010. The study included 65 patients (41 males) from the Haemodialysis Unit and 52 patients (28 males) from the Peritoneal Dialysis Unit. Those patients who had arrhythmia, severe valvular heart disease, amputation of any limb, pacemakers or metal prostheses that would interfere with bioimpedance were excluded. All patients signed the informed consent and the hospital ethics committee approved the study protocol. Measurements Examinations of patients on haemodialysis (HD) were performed during the middle of the week, while patients on peritoneal dialysis were examined before the first replacement of the morning with an empty peritoneum. Nefrologia 2012;32(1):94-102 originals To assess hydration status, we used multifrequency bioimpedance with the Fresenius BCM-Body Composition Monitor, which measures 50 different frequencies from 5kHz to 1MHz. This technique has been validated by dilution techniques that are considered the gold standard,9 dual X-ray absorptiometry and plethysmography,10 among others.11,12 For the measurement, we used two pairs of electrodes: one at a distal position, an injector and a sensor, placed dorsally on the hand (third metacarpophalangeal and carpal joints, respectively) and another on the foot (third metatarsophalangeal and tibiotarsal joints). The reference was the right hemisoma; in HD, the reference was the hemisoma free of vascular accesses. Prior to the procedure, patients were accurately measured and weighed. Examination was performed in the HD group before and after the session. In both HD and peritoneal dialysis (PD), the patient was placed in supine decubitus for 15-20 minutes before the examination in order to help distribute excess fluid and avoid the presence of oedema that could distort results. After the HD session, we also waited the same amount of time to allow balancing among the various compartments (intravascular-extravascular-cellular). Among the various parameters obtained by BIS, we chose the extracellular water-total body water ratio (ECW/TBW) to assess hydration status and the overhydration-extracellular water ratio (OH/ECW) for the mortality risk. Patients were classified by hydration status by means of ECW/TBW normalised for age and gender using the method described by Lindley et al, i.e. the difference between the theoretical ratio under normal conditions and that obtained by bioimpedance. If the difference is >2.5%, the patient is hyperhydrated (HHD), if between >2.5% and <2.5% the patient is normohydrated (NHD), and dehydrated if <2.5%.13 The OH/ECW is a ratio that currently is defined as a significant and independent predictor of mortality in patients on dialysis when it is greater than 15%.14 We defined AHT in those patients who had blood pressure (BP) readings >140/80mm Hg and/or were taking antihypertensive drugs. On the examination day, a blood test was performed to determine haemoglobin (mg/dl), calcium (mg/l), phosphorus (mg/l), parathormone (pg/ml), total proteins (g/dl) and albumin (g/dl) in each patient. We performed transthoracic echocardiography with a multifrequency transducer and a tissue Doppler program (Aloka) before the HD session, and after emptying the peritoneal cavity in PD. Readings and measurements were made following the recommendations of the American Society of Echocardiography (ASE).11,15 Left ventricular mass (LVM) was calculated using the modified ASE 95 originals Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients formula,16,17 which is the most widely used method since it has shown values closely associated with autopsy findings: LVM=0.8x [1.04x (LVTDD + TDTS + TDTPW) 3+ (LVTDD)3] +0.6 (LVTDD: LV telediastolic diameter; TDTS: telediastolic thickness of the septum; TDTPW: telediastolic thickness of the posterior wall). LVM was indexed by body surface (ILVM). Based on the studies of Devereux et al, the cut-off point for the diagnosis of LVH using the ILVM was ≥125g/m2, with no difference by gender (Figure 1).16 r > 0.45 ILVM (g/m2) < 125 ILVM (g/m2) > 125 Remodeling Concentric hypertrophy Normal Eccentric LVH To classify the type of LVH, we calculated the relative thickness (RT) with the formula: RT=2xPW/LVTDD; we considered a normal RT when <0.45.16 The assessment of the left atrium was performed by measuring the left atrium volume by the indexed Simpson’s disc summation method, where the volume of the left atrium is the individual summation of all discs in the series. We used this method for its speed and for correlating well with any method for determining left atrial volume.18,19 r < 0.45 ILVM: indexed left ventricular mass Figure 1. Patterns of left ventricular hypertrophy Data were processed using the SPSS 15 program. Results are expressed as mean, standard deviation for data with normal distribution; and as median, interquartile range (IQR) and confidence interval (CI) for data that did not have normal distribution. We performed the comparison of means with the Mann-Whitney and Student’s-t test according to the distribution of the variables. Linear regression was performed with Pearson’s p. To compare means between the two groups, we performed ANOVA for one factor with Bonferroni and Dunnett’s C post-hoc tests, according to the homogeneity of variances. Values of P<.05 were considered statistically significant. We performed a multivariate analysis, using eccentric LVH as the dependent variable and the other study variables as independent variables. RESULTS We examined 117 patients, 65 patients in the HD group and 52 in the PD group. The relevant demographic and clinical characteristics and cardiovascular risk factors are shown in Table 1. It is noteworthy that patients who were on HD had been longer on the technique and had lower residual renal function (RRF) than patients on PD. As for laboratory tests, it is noteworthy that patients on HD had higher readings of total proteins and albumin in blood. The presence of AHT was greater in PD patients, although HD had poorer control of readings. The use of antihypertensive drugs and their combination was greater in patients on PD patients (59.6% vs 30.8%). The most frequently used drugs in both groups were calcium antagonists and angiotensin-converting enzyme inhibitors (ACEI)/angiotensin II receptor antagonists (ARAII). Diuretics were used more frequently in PD. 96 We initially examined the relationship in each dialysis group between the hydration status and BP, by means of normalised ECW/TBW ratios. Patients on HD before and after performing the session presented hydration states and BP readings that are shown in Figure 2 and Figure 3. The distribution of patients on PD is shown in Figure 4. In the HD group, 37.5% presented systolic blood pressure (SBP) readings >140mm Hg, with 25% being HHD and 12.5% NHD. Some 62.5% were normotensive, with 42.1% being HHD and 18.8% NHD. In the PD group, 24.9% had SBP >140mm Hg, 11.5% were HHD and 11.5% were NHD. Some 74.9% were normotensive, with 34.6% being HHD and 32.6% NHD. We observed a decline in the number of HHD patients in the HD group after the session (67.1% pre-HD compared to 26.1% post-HD), as well as a decline in blood pressure readings (SBP>140mm Hg - 37.5% to 32.6%). If we consider the presence of hypertension (>140/80mm Hg or drug treatment) and the hydration status in each dialysis group, we observe AHT in 45.45% of the HD group, of which 30.3% were HHD. After the HD session, we observed a decrease in AHT patients (post-HD 43.4%) and a decrease in the number of HHD patients (68.1% vs 27.3%). In PD patients, 69.2% had HTA, of which 28.8% were HHD. Therefore, if we consider the readings for BP and drug treatment, we find greater AHT in PD patients and greater HHD states in PD patients than in readings after HD. The presence of HHD according to ECW/TBW was more frequent in HD than in PD before the dialysis session (67.1% pre-HD compared to 46.1% PD), but after the HD Nefrologia 2012;32(1):94-102 Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients originals Table 1. Characteristics of patients on peritoneal haemodialysis and dialysis HD Group PD Group (n=65) (n=52) 60 ±13,8 59 ±17,8 NS 41/24 28/24 NS 51.8 (3-259) 33.5 (3-93) 0.04 RRF (% pac) 43.1 65.38 NS Diuresis (cc) 1000 (421.5-749.6) 1625 (482-981.6) NS Hypertension (%) 46.2 69.2 0.04 BP>140/80mm Hg (%) 37.5 24.9 NS Antihypertensive treatment (%) 30.8 59.6 0.003 Diuretics (%) 15,2 21,2 NS ß-blockers (%) 12,1 17,3 NS Calcium antagonists (%) 18,2 32,7 NS ACEI/ARAII (%) 18,2 46,2 0,007 α-blockers (%) 18,2 19,2 NS Drug combination (%) 18.2 48.1 NS Calcium (mg/l) 9.2±0.64 9.5±0.63 P<.05 Phosphorus (mg/l) 4.9±1.48 4.7±1.37 NS 283.83 (20.8-1.485) 214.8 (18-763.7) NS Total protein (g/dl) 6.8±0.54 6.2±0.59 P<.001 Albumin (g/dl) 3.9±0.31 3.5±0.37 P<.001 Haemoglobin (mg/dl) 12.24±1.7 11.8±1.68 NS Age (years) Gender (M/F) Time on technique (months) PTH (pg/ml) SD ARA II: angiotensin II receptor antagonists; PD: peritoneal dialysis; SD: standard deviation; HD: haemodialysis; ACEI: angiotensin-converting enzyme inhibitors; RRF: residual renal function; NS: not significant; PTH: parathormone; BP: blood pressure. session a better control of the hydration status is achieved (post-HD 26.1%). When analysing each dialysis group according to hydration status, the subgroup of HHD patients in both techniques had been longer on the technique (HHD HD 60.9 months vs NHD HD 36.5 months; HHD PD 36.3 months vs NHD PD 34.8 months), with only those on HD showing a significant difference. The presence of RRF was similar in both techniques, although it was lower in the HHD group (RRF HHD HD 34.9% vs NHD HD 55%; HHD PD 37.5% vs NHD PD 42.9%). According to the data obtained using BCM, patients on HD had significantly greater overhydration (OH) and extracellular/intracellular (E/I) water than those on PD (Table 2). The second phase of the study consisted of assessing each dialysis group, according to the hydration status as Nefrologia 2012;32(1):94-102 measured by ECW/TBW, the echocardiographic characteristics and the correlation among the data obtained by bioimpedance (Table 3). Left atrial volume indexed to body surface (LAVI) and indexed left ventricular mass (ILVM) were significantly greater in patients in the HD group and in the HHD subgroup of both techniques. The OH/ECW ratio was significantly greater in HD patients than in PD patients (>15%, 14.06% in HD vs 3.8% in PD), as well as in HHD patients in each type of dialysis. If we assess the geometry of the left ventricle in our study population, we observe the presence of LVH in 27.6% of the HD group and 13.4% in PD; the most prevalent in both groups being the eccentric one. According to the hydration status, the presence of LVH was greater in the HHD group (HD 27.9% vs PD 20.8%) than in the NHD group (25% vs 9.5%), with eccentric LVH being the most prevalent (Tables 4 and 5). In the univariate study of eccentric LVH with all 97 Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients originals 220 220 12.5% 251% 200 BP (mm Hg) BP (mm Hg) 160 140 120 18.7% 1.5% 100 42.1% 140 120 4.9% 44.2% 18% 80 -4 -2 0 2 4 6 8 60 -6 10 Pre HD ECW/TBW (%) 24.5% 11.5% 11.5% 200 180 160 140 120 100 7.6% 34.6% 32.6% 80 60 -6 -4 -2 0 2 -2 0 2 4 6 8 10 ECW/TBW: extracellular water-total body water ratio; HD: haemodialysis; BP: blood pressure. Figure 2. Distribution of patients on haemodialysis before the session according to blood pressure readings and the difference of the ECW/TBW ratio. 220 -4 Post HD ECW/TBW (%) ECW/TBW: extracellular water-total body water ratio; HD: haemodialysis; BP: blood pressure. BP (mm Hg) 160 100 80 4 6 8 10 PD ECW/TBW (%) PD: peritoneal dialysis; ECW/TBW: extracellular water-total body water ratio; BP: blood pressure. Figure 4. Distribution of patients on peritoneal dialysis according to blood pressure readings and the difference of the ECW/TBW ratio variations, the only one of significance was eccentric LVH with hydration status (normalised ECW/TBW) with a significance of P=.03 (CI 1.1-11.5). DISCUSSION Bioimpedance is a technique that allows us to detect a large number of HHD patients. It is an easily applicable method and may be considered the gold standard in determining 98 8.1% 180 180 60 24.5% 200 Figure 3. Distribution of patients on haemodialysis after the session according to blood pressure readings and the difference of the ECW/TBW ratio. hydration states. Various methods have been used to assess hydration status, but they are unreliable since numerous factors may interfere. The assessment of the hydration status by means of the BP may be distorted by antihypertensive therapy, AHT not dependent on volume, and the presence of heart disease;20 the inferior vena cava shows the intravascular volume by diastolic dysfunction;21 and RRF and the presence of heart diseases,22 among others, may interfere with biochemical markers, such as BNP and pro-BNP. We found in the literature that various parameters have been used to define hydration status by bioimpedance: OH/ECW, ECW/TBW, E/I and OH. Lopot et al determined the optimal dry weight in HD patients using the deviation between ECW/TBW obtained by bioimpedance and that obtained in a control group, according to age and sex. This is based on changes produced in the cell mass (with increasing age, the cell mass and therefore the intracellular water decreases) and the hydration status (with increasing age and/or female gender, the intracellular water decreases).23 Lindley et al determined the dry weight in PD using the difference between ECW/TBW obtained by BIS compared to ECW/TBW of a control group according to patient age and gender (defining HHD as those who had +2.5% standard deviation).13 We assessed the hydration status of our patients based on these principles.9,24 In the HD group, we found significantly more HHD patients before HD (67.1%) than in the PD group (46.1%), and almost half had AHT. After the HD session, better control of hydration status is achieved (26.1%). Devolder I et al assessed hydration status by means of OH/ECW (overhydrated OH/ECW>15%),23 Plum et al by Nefrologia 2012;32(1):94-102 Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients originals Table 2. Assessment of the heart and the overhydration-extracellular water ratio according to the state of hydration in each dialysis group LAVI (ml/m ) 2 ILVM (g/m2) HD Group HDHHD HDNHD HD Group PDHHD PDNHD 35.6±13.2 37.5±12.9 31.2±11.6 28.5±10.3 31.6±10.9 25.6±10 (32.3-38.9)α (33.5-41.6)β (25.7-36.6) (25.5-31.6) (26.5-36.8) (20.8-30.4) 108.8±30.3 116.5±28.4 94±26.1 96.7±25.1 105.5±26.4 85.1±20.7 (101.2-116.4)α (107.5-125.4)β (81.7-106.2)γ (104.2-89.28) (93.1-117.9) (75.1-95.1) 14.06 100 0 3.8 100 0 OH/ECW (> 15%) ECW/TBW 48.7±3.05 50.2±2.3 46.2±1.5 47±4.6 50.7±3.1 44.6±2.8 (48.03-49.5)α (49.5-50.9)β (45.4-46.9)γ (45.7-48.3) (49.4-52.1) (43.4-45.9) (a) P<.05 between HD and PD; (b) P<.05 between HDHHD and PDHHD; (g) P<.05 between HDNHD and PDNHD. PD: peritoneal dialysis; ECW/TBW: extracellular water-total body water ratio; HD: haemodialysis; HHD: hyperhydrated; ILVM: indexed left ventricular mass; LAVI: left atrial volume indexed to body surface; NHD: normohydrated; OH/ECW: overhydration/extracellular water ratio. Table 3. Parameters obtained by bioimpedance in peritoneal haemodialysis and dialysis groups HD Group PD Group P Pre-HD Post-HD OH (l) 1.13±1.36 (0.7-1.4) –0.27±1.39 (–0.63-0.08) 0.28±1.3 (–0.09-0.66) 0.000 0.001 TBW (l) 32.9±7.7 (31-34.8) 31.7±7.5 (29.8-33.6) 32.7±6 (31-34.4) 0.000 0.86 ECW (l) 16±3.9 (15.11-16.9) 14.6±3.3 (13.7-15.4) 15.2±2.5 (14.5-15.9) 0.000 0.2 ICW (l) 16.8±4.3 (15.8-17.9) 17.1±4.4 (15.9-18.2) 17.4±4.1 (16.2-18.5) 0.059 0.48 E/I 0.96±0.11 (0.93-0.99) 0.86±0.1 (0.83-0.89) 0.9±0.17 (0.85-0.95) 0.000 0.03 32.6±9.8 33.4±9.7 33.2±11.9 0.08 0.7 (30.1-35) (31-35.9) (29.8-36.5) 28.9±9.2 (26.6-31.2) 27±8.6 (24.8-29.2) 26.7±10.2 (23.9-29.6) 0.09 0.2 Muscle mass (kg) Fat mass (kg) Pre-Post-HD-P PD: peritoneal dialysis; ECW: extracellular water; E/I: ECW/ICW ratio; HD: haemodialysis; ICW: intracellular water; OH: overhydration water; TBW: total body water. ECW/TBW25 and Passauer et al by OH (litres of overhydration >1.1 I).11 They found that hydration status was harder to control in patients on PD than in post-HD patients, which is consistent with our data. Table 4. Left ventricular geometry according to dialysis type HD Group (%) DP Group (%) Normal 46.15 61.5 Concentric remodelling 26.1 25 Eccentric hypertrophy 21.5 9.6 Concentric hypertrophy 6.1 3.8 PD: peritoneal dialysis; HD: haemodialysis. Nefrologia 2012;32(1):94-102 The bodily distribution of excess fluid in patients on dialysis varies according to the type of technique. Therefore, the impact of hydration status on BP and at the cardiac level must vary. In the literature, excess fluid in patients on PD is found peripherally in the subcutaneous tissue, while in HD patients it is located in the bloodstream.25 In HD, there is an increase in body weight in the interdialytic period (48 hours), due to the consumption of liquids and food, and a decrease during the session (4 hours) by the ultrafiltration of the accumulated water volume and by the convective and diffusive transport of sodium. Abrupt changes occur at the intravascular level, very slowly from the interstitial space by means of refilling. Interdialytic weight gain is greater in patients who do not have RRF. The PD technique is usually performed continuously, and the balance between the various compartments is produced constantly. Ultrafiltration is produced from the interstitial space. 99 Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients originals Table 5. Left ventricular geometry according to hydration state HHDHD (%) NHDHD (%) DHDHD (%) HHDPD (%) NHDPD(%) DHDPD(%) Normal 46.5 45 ---- 54.1 66.6 71.5 Concentric remodelling 25.5 30 ---- 25 23.8 28.5 Concentric hypertrophy 9.3 0 ---- 8.3 0 0 Eccentric hypertrophy 18.6 25 ---- 12.5 9.5 0 DHD: dehydrated; PD: peritoneal dialysis; HD: haemodialysis; HHD: hyperhydrated; NHD: normohydrated. We must remember that bioimpedance assesses the intraextracellular hydration status, and not hydration at the intravascular level. The increased state of HHD in HD before the session is due to weight gain in the interdialytic period, which is located at the extracellular and intravascular level, resulting in a greater E/I ratio. After the HD session, the hydration status is reduced by the ultrafiltration of the intravascular space, and it takes some time for the various compartments to reach a balance. In PD, we observed a significantly lower E/I ratio than in HD (E/I pre-HD 0.96 vs PD 0.9) due to a lower extracellular water (ECW pre-HD 16 1 vs PD 15.2 1) because ultrafiltration in this technique is performed constantly. Therefore, when we perform bioimpedance in patients on HD, especially after the session, balance has not yet been reached among the various compartments. Probably because of this, hydration parameters in PD compared to post-HD may be distorted. dry weight, continuous changes in the peritoneal membrane, etc.) Patients in HD had greater hydration states before the dialysis session, but decreased after the session, with subsequent increases up to the next session, reflecting these changes in the blood pressure. Patients on HD usually had less antihypertensive treatment because during the HD session a reduction in intravascular volume is produced and this must be adapted. If the patient is undergoing antihypertensive treatment, the compensation and/or vascular adaptation mechanisms are blocked and present continuous vasodilation, which results in hypotension, poor tolerance, and an inability to reach the target dry weight. At the same time, vascular refilling is produced from the interstitial liquid at different speeds, depending on the patient’s characteristics (elderly, diabetic, and patients with ventricular dysfunction or pulmonary hypertension present very slow refilling). In the literature, the prevalence of AHT in patients on HD is around 60% in the interdialytic period,26 while for PD it is greater (80%).27 Our prevalence of AHT was lower. The presence of high BP readings and/or antihypertensive treatment was greater in PD patients (PD 76.9% vs HD 49.2%), but they had better blood pressure control (BP<140mm Hg, 37.5% HD vs 24.9% PD), probably due to the greater use of antihypertensive drugs. The most widely used drugs in both groups were calcium antagonists and ACEI/ARAII. The prevalence of LVH is 75% among patients on dialysis,30,31,32 and develops in initial stages of CKD. Numerous studies have shown its increase in parallel with the reduction of GFR.33 AHT and hyperhydration have an impact on the heart with the development of LVH, among other risk factors. We found studies, such as the Wang AY et al study, where RRF had considerable influence and 70% of patients with no RRF presented ventricular modelling disorders.34 The development of LVH varies according to the type of overload. Fluid or volume overload is related to the onset of eccentric LVH, while pressure overload and AHT is related to concentric LVH. We found a correlation between eccentric LVH and the hydration state. Foley et al suggest that regression of LVH does not occur after starting dialysis and that it is irreversible, due to high patient mortality.35,36 However, last year we found published studies that demonstrated that LVH regression is possible after years of dialysis. Several therapeutic strategies may work: control of anaemia, control of hydration status, use of antihypertensive drugs in normotensive and hypertensive patients, daily or night-time use of HD, prevention and treatment of hyperphosphataemia, vitamin D administration, and multifactorial intervention.37,38,39 We found regression not only in patients on HD, but also in patients on PD.40,41 Diuretics are used more in PD due to the greater prevalence of RRF, and this allows us to increase the diuresis volume and adjust hydration status. The increased presence of RRF in PD is due to PD being a continuous technique, with constant ultrafiltration that precludes abrupt changes in volume, which helps protect the kidney. Moreover, patients on HD usually have poorer renal function due to the greater use of nephrotoxic agents (aminoglycosides, contrast media, non steroidal anti-inflammatory drugs), the bioincompatibility of filter membranes and/or increased frequency of hypotension episodes.28,29 We found an inverse correlation between time on dialysis and presence of RRF in our patients, due to the continuous development of kidney disease over time, which results in a reduced RRF, although in most studies the mean time on the technique is lower than ours. The greater frequency of AHT in PD patients may be due to the greater hydration status (less accuracy in the adjustment of 100 In our study, the prevalence of LVH was less (53.5% in HD vs 38.4% in PD). The lower presence of cardiac disorders may be due to acceptable control of hydration status and BP, a high presence of RRF, as well as good control over Nefrologia 2012;32(1):94-102 Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients haemoglobin readings, calcium-phosphorus metabolism, and the use of ACEI and ARA II. The difference found between both dialysis techniques may be explained by the greater percentage of patients with no RRF in HD,31 the lower use of ACEI/ARAII, and greater extracellular water ratio. LAVI shows the average of increased filling pressures,42 the functional situation of the heart and overload, and thus, the hydration status. Patients on HD have a significantly greater LAVI than those on PD, and HHD patients have greater intravascular volume and therefore greater cardiac overload, leading to increased LVH. Lastly, we analysed two emerging markers for the stratification and monitoring of cardiovascular risk in patients with CKD, LAVI and OH/ECW.8,14,17,42 Wizemann et al14 defined the OH/ECW ratio >15 as a cardiovascular risk factor, based on the study performed by Wabel et al in which NHD patients presented an OH/ECW of 6.8%-15%.20 LAVI is a chronic marker of diastolic function that shows the average of increased filling pressures,42 and is considered the best index for assessing filling pressures and the functional situation of the heart, since it is linked to the severity and duration of diastolic dysfunction of the left ventricle.18,19 In our study, both indices are significantly higher in patients on HD, as well as in HHD patients in both groups. Therefore, being HDD according to the normalised ECW/TBW ratio >2.5% is a high cardiovascular risk. 4. 5. 6. 7. 8. 9. 10. 11. We can conclude that bioimpedance detects a greater number of HDD patients on both dialysis techniques. When studying echocardiographic disorders in dialysis patients, we found a high correlation between hydration status and LAVI and ILVM. Additional prospective studies are warranted to assess the hydration states of the dialysis population and to determine whether there is regression in cardiac remodelling with the control of the hydration status by means of bioimpedance. 12. 13. Conflict of interest 14. The authors declare they have no potential conflicts of interest related to the contents of this article. 15. REFERENCES 16. 1. Locatelli F, Marcelli D, Conte F, D’Amico M, Del Vecchio L, Limido A, et al. Cardiovascular disease in chronic renal failure: the challenge continues. Nephrol Dial Transplant 2000;15 (Suppl.5):69-80. 2. Tonelli M, Wiebe N, Culleton B, House A, Rabbat C, Fok M, et al. Chronic kidney disease and mortality risk: a systematic review. J Am Soc Nephrol 2006;17:2034-47. 3. Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL, et al.; American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Kidney disease as a risk factor for development Nefrologia 2012;32(1):94-102 17. 18. 19. originals of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation 2003;108(17):2154-69. Parfrey PS, Foley RN. The clinical epidemiology of cardiac disease in chronic renal failure. J Am Soc Nephrol 1999;10:1606-15. Silverberg DS, Wexler D, Iaina A, SteinbruchS, Wollman Y, Schwartz D. Anemia, chronic renal disease and congestive heart failure—the cardio renal anemia syndrome: the nedd for cooperation between cardilogists and nefephrologists. Int Urol Nephrol 2006;38:295-310. Kuhlmann MK, Zhu F, Seibert E, Levin NW. Bioimpedance, dry weight and blood pressure control: new metrhods and consequences. Curr Opin Nephrol Hypertens 2005;14:543-9. Bargnoux AS, Kloudche K, Fareh J, Barazer I, Villard-Saussine S, Dupuy AM, et al. Prohormone brain natriuretic peptide (proBNP), BNP and Nterminal-proBNP circulating levels in chronic hemodialysis patients. Correlation with ventricular function, fluid removal and effect of hemodiafiltration. Clin Chem Lab Med 2008;46:1019-24. Matthie J, Zarowitz B, De Lorenzo A, Andreoli A, Katzarski K, Pan G, et al. Analytic assessment of the various bioimpedance methods used to estimate body water. J Appl Physiol 1998;84:1801-16. Moissl UM, Wabel P, Chamney PW, Bosaeus I, Levin NW, Bosy-Westphal A, et al. Body fluid volume determination via body composition spectroscopy in health and disease. Physiol Meas 2006;27:921-33. Moissl U, Bosaeus I, Lemmey A, Hovgesen S, Wabel P, Chamney PW, et al. Validation of a 3C model for determination of body fat mass. J Am Soc Nephrol 2007;18:257A. Passauer J, Petrov H, Schleser A, Leicht J, Pucalka K. Evaluation of clinical dry weight assessment in haemodialysis patients using bioimpedance specctroscopy: a cross-sectional study. Nephrol Dial Transplant 2010;25:545-51. Wizemann V, Rode C, Wabel P. Whole-body spectroscopy (BCM) in the assessment of normovolemia in hemodialysis patients. Contrib Nephrol 2008;161:115-8. Lindley E, Devine Y, Hall L, Cullen M, Cuthbert S, Woodrow G, et al. A Ward-based procedure for assessment of fluid status in peritoneal dialysis patients using bioimpedance spectroscopy. Perit Dial Int 2005;25 Suppl 3:S46-S48. Wizemann V, Wabel P, Chamney P, Zaluska W, Moissl U, Rode Ch, et al. The mortality risk of overhydration in haemodialysis patients. Nephrol Dial Transplant 2009;24(5):1574-9. Devereux RB, Lutas EM, Casale PN, Kligfield P, Eisenberg RR, Hammond IW, et al. Standardization of M-mode echocardiographic left ventricular anatomic measurements. J Am Coll Cardiol 1984;4(6):1222-30. Devereux RB, Alonso DR, Lutas EM, Gottlieb GJ, Camp E, Sachs I, et al. Echocardiographic assessment of Left Ventriular Hypertrophy: Comparison to Necropsy Findigs. Am J Cardiol 1986;57(6):450-8. Quiñoles MA, Otto CM, Stoddard M, Waggoner A, Zoghbi WA. Recommendations for quantification of Doppler echocardiography: a report from the Doppler Quantification Task Force of the nomenclature and Standards Committee of the American Society of Echocardiography. J Am Soc Echocardiogr 2002;15(2):167-84. Tripepi G, Benedetto FA, Mallamaci F, Tripepi R, Malatino L, Zoccali C. Left atrial volumen in end-stage renal disease: a prospective cohort study. J Hypertens 2006;24:1173-80. Tripepi G, Benedetto FA, Mallamaci F, Tripepi R, Malatino L, Zoccali C. 101 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. Left atrial volumen monitoring and cardiovascular risk in patients with end-stage renal disease: a prospective cohort stuydy. J Am Soc Nephrol 2007;18:1316-22. Wabel P, Moissl U, Chamney P, Jirka T, Machek P, Ponce P, et all. Towards improvend cardiovascular manegement: the necessity of combining blood pressure and fluid overload. Nephrol Dial Transplant 2008;9:2965-71. Leunissen KM, Kouw P, Kooman JP. New techniques to determine fluid status in hemodialyzed patients. Kidney In 1993;41:S50-S56. Bargnoux AS, Klouche K, Fareh J, Barazer I, Villard-Saussine S, Dupuy AM, et al. Prohormone brain natriuretic peptide (proBNP), BNP and Nterminal-proBNP circulating levels inchronic hemodialysis patients. Correlation with ventricular function, fluid removal and effect of hemodiafiltration. Clin Chem Lab Med 2008;46:1019-24. Devolder I, Verleysen A, Vijt D, Vanholder R, Van Biesen W. Body composition, hydration, and related parameters in hemodialysis versus peritoneal dialysis patients. Perit Dial Int 2010;30(2):208-14. Lopot F, Nejedly B, Novotna H, Mackova M, Sulkova S. Age-related extracellular to total body water volume ratio (ECV/TBW)- Can it be used for «dry weight» determination in dialysis patients? Application of ultifrequency bioimpedance measurement. Int J Artif Organs 2002;25:762-9. Plum J, Schoenicke G, Kleophas W, Kulas W, Steffens F, Azem A, et al. Comparison of body fluid distribution between chronic haemodialysis and peritoneal dialysis patients as assessed by biophysical and biochemical methods. Nephrol Dial Transplant 2001;16:2378-85. Kooman JP, Gladziwa U, Böcker G, Wijnen AG, van Bortel K, Luik AJ, et al. Blood pressure during the interdialytic period in hemodialysis patients; estimación of representative pressure values. Nephrol Dial Transplant 1992;7:917-23. Menon MK, Naimark DM, Bargman J, Vas SI, Oreopoulos DG. Long-term blood pressure control in a cohort of PD patients and its association with residual renal function. Nephrol Dial Transplant 2001;16:2207-13. Lysaght MJ, Vonesh EF, Gotch F, Ibels L, Keen M, Lindholm B, et al. The influence of dialysis traatment modality on the decline of remaining renal function. ASAIO Trans 1991;37:598-604. Gámez C, Teruel JL, Ortuño J. Evolución de la función renal residual en enfermos tratados con hemodiálisis. Nefrologia 1992;12:125-9. Foley RN, Parfrey PS, Harnett JD, Kent GM, Martin CJ, Murray DC, et al. Clinical and echocardiographic disease in patients starting end-stage renal disease therapy. Kidney Int 1995;47(1):186-92. 31. López Gómez JM, Verde E, Pérez García R. Blood pressure left ventricular hipertrophy and long term prognosis in hemodialysis patients. Kidney Int Suppl 1998;68:S92-8. 32. Levin D, Singer J, Thompson CR. Prevalent lsft hypertrophy in the predialysis populations: identiffying opportunities for intervention. Am J Kidney Dis 1996;27:347-54. 33. Bossola M, Tazza L, Vulpio C, Luciani G. Is regression of left ventricular hypertrophy in maintenance hemodialysis patients possible? Semin Dial 2008;21(5):422-30. 34. Wang AY, Wangg M, Woo J, Law MC, Chow KM, Li PY, et al. A novel association between resudual renal function and left ventricular hypertrophy in peritoneal dialysis patients. Kidney Int 2002;62:639-47. 35. Foley RN, Parfrey PS, Kent GM, Harnett JD, Murray DC, Barre PE. Long-term evolution of cardiomiopathy in dialysis patients. Kidney Int 1998;54:1720-5. 36. Foley RN, Parfrey PS, Morgan J, Barré PE, Campbell P, Cartier P, et al. Effect of hemoglobin levels in hemodialysis patients with asymptomatic cardiomyopathy. Kidney Int 2000;58(3):1325-35. 37. Ardeleanu S, Panaghiu L, Prisada O, Sasca u R, Voroneanu L, Hogas, S, et al. up study. Int Urol Nephrol. [Epub ahead of print] 38. London GM, Pannier B, Guerin AP, Blacher J, Marchais SJ, Darne B, et al. Alterations of left ventricular hypertrophy in and survival of patients receiving hemodialysis: follow-up of an interventional study. J Am Soc Nephrol 2001;12(12):2759-67. 39. Levin A, Thompson CR, Ethier J, Carlisle EJ, Tobe S, Mendelssohn D, et al. Left ventricular mass index increase in early renal disease: impact of decline in hemoglobin. Am J Kidney Dis 1999;34(1):125-34. 40. Wang AY, Wang M, Lam CW, Chan IH, Lui SF, Sanderson JE. Heart Failure in Long-Term Peritoneal Dialysis Patients: A 4-Year Prospective Analysis. Clin J Am Soc Nephrol 2011;6(4):805-12. 41. Io H, Ro Y, Sekiguchi Y, Shimaoka T, Inuma J, Hotta Y, et al. Cardiac function and structure in longitudinal analysis of echocardiography in peritoneal dialysis patients. Perit Dial Int 2010;30(3):353-61. 42. Tsang TS, Barnes ME, Gersh BJ, Bailey KR, Seward JB. Left atrial volume as a morphophysiologic expression of left ventricular diastolic dysfunction and relation to cardiovascular risk burden. Am J Cardiol 2002;90:1284-9. c 20. Isabel Juan-García et al. Echocardiographic impact of hydration status in dialysis patients c originals Sent for Review: 23 Feb. 2011 | Accepted: 1 Nov. 2011 102 Nefrologia 2012;32(1):94-102 http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society short originals Vascular accesses in haemodialysis: a challenge to be met Gloria Antón-Pérez1, Patricia Pérez-Borges1, Francisco Alonso-Almán2, Nicanor Vega-Díaz1 1 2 Servicio de Nefrología. Hospital Universitario de Gran Canaria Dr. Negrín. Las Palmas de Gran Canaria (Spain) Unidad Satélite de Hemodiálisis. Centro Satélite de Hemodiálisis RTS-Baxter. Las Palmas de Gran Canaria (Spain) Nefrologia 2012;32(1):103-7 doi10.3265/Nefrologia.pre2011.Oct.11027 Accesos vasculares en hemodiálisis: un reto por conseguir ABSTRACT Background: Chronic kidney disease is a leading problem in public health due to its high incidence, prevalence and high morbidity and mortality, especially for those who require renal replacement therapy (RRT). As has already been described by other authors, the vascular access is one of the factors determining morbidity and mortality of patients in haemodialysis as well as their complications, which incur a high cost. Objectives: To know the real situation of our clinical practice, compare it with data from other studies, and to measure the degree of compliance by these patients with the recommendations of haemodialysis (HD) Clinical Practice Guidelines regarding vascular access . Also, to assess survival according to the type of vascular access used, adjusting for comorbidity factors. Patients and Methods: We studied the vascular access of our prevalent patients on haemodialysis by October 2009 (n=299, 62% men). Of these, 64% underwent HD through an autologous arteriovenous fistula (AVF), 3% were carrying synthetic grafts, and 33% had a central venous catheter (CVC). These percentages do not comply with the recommendations of the S.E.N. and KDOQI clinical guidelines. In order to know the real situation of our clinical practice, we compared our data with other studies, and measured the degree of compliance with the recommendations of the guidelines. The incident patients on HD were studied from January 2004 to October 2009 (n=422). We analysed basal nephropathy, associated diseases, and the type of vascular access at the start of HD. Results: A total of 30% had an AVF, 1% had synthetic grafts, and 69% had CVC. The calculated relative risk (RR) of death associated with the use of CVC at the start of HD was 3.68 (95% CI: 2.93-6.35) adjusted for other factors of comorbidity (age, diabetes mellitus, ischaemic heart disease, peripheral arterial disease). Conclusions: The high mortality associated at the beginning of HD with CVC (RR: 3.68), independently of other factors, make the decrease in the use of this vascular access an objective of first order. Presently, we have not been able to meet the objectives from the different Clinical Guidelines with respect to the prevalence and incidence of the vascular accesses for HD. Keywords: Central venous catheter. Arteriovenous fistula. Haemodialysis. Vascular access Correspondence: Gloria Antón-Pérez Servicio de Nefrología. Hospital Universitario de Gran Canaria Dr. Negrín. Las Palmas de Gran Canaria. Spain. [email protected] RESUMEN Introducción: La enfermedad renal crónica representa un problema de salud pública por su elevada incidencia, su prevalencia, su alta morbimortalidad, sobre todo en aquellos que precisan de tratamiento renal sustitutivo. Uno de los factores que determinan la morbimortalidad de los pacientes en hemodiálisis (HD) es el acceso vascular del que disponen, y las complicaciones asociadas a los problemas de acceso vascular suponen una importante carga en nuestro trabajo diario, así como un elevado coste. Objetivos: Conocer la situación real de nuestra práctica clínica, compararla con otros estudios y medir el grado de cumplimiento de las recomendaciones de las Guías de Práctica Clínica en HD en lo relativo al acceso vascular de pacientes incidentes y prevalentes. Estudiar la supervivencia de los pacientes incidentes en función de su acceso vascular, ajustada a otros factores comórbidos. Pacientes y métodos: Se estudiaron los pacientes incidentes en HD desde enero de 2004 a octubre de 2009 (n = 422). Se analizaron: acceso vascular al inicio de HD, nefropatía de base, servicios de procedencia y enfermedades asociadas. Estudiamos el acceso vascular de nuestros pacientes prevalentes a fecha de octubre de 2009 (n = 299). Comparamos la supervivencia de los pacientes incidentes en función de su acceso vascular, ajustándolo a otros factores comórbidos. Resultados: El 67% de los pacientes prevalentes (62% hombres) portaban acceso vascular definitivo, y el 33%, un catéter venoso central (CVC). Del total de 422 pacientes incidentes, 42% provenían de la consulta por enfermedad renal crónica avanzada. El 54% eran diabéticos, el 92% hipertensos, el 28% presentaban cardiopatía isquémica filiada y un 13% arteriopatía periférica. Un 30% de los pacientes iniciaron HD a través de fístula arteriovenosa, un 1% portaban injerto sintético de PTFE (politetrafluoretileno) y un 69% CVC. El riesgo relativo de muerte asociado al uso de CVC al inicio de HD fue de 3,68 (intervalo de confianza: 95%, 2,93-6,35), ajustándolo a otros factores de comorbilidad (edad, diabetes mellitus, cardiopatía isquémica, arteriopatía periférica). Conclusiones: La alta mortalidad asociada al inicio de HD con CVC (riesgo relativo: 3,68), independientemente de otros factores, hacen de la reducción del uso de este acceso vascular un objetivo de primer orden. En nuestro medio no hemos podido conseguir los objetivos reseñados en las diferentes Guías en lo referente a la prevalencia e incidencia de los accesos vasculares para HD. Palabras clave: Catéter venoso central. Fístula arteriovenosa. Hemodiálisis. Acceso vascular INTRODUCTION Chronic kidney disease (CKD) is a public health problem due to its elevated incidence, prevalence, morbidity and mortality rates, and because it is considered as an 103 Gloria Antón-Pérez et al. Vascular accesses in haemodialysis short originals independent cardiovascular risk factor, especially in patients that require renal replacement therapy (RRT). According to the registry maintained by the Spanish Society of Nephrology (S.E.N.), 36 388 patients underwent RRT in Spain in 2007, over 46% of them on haemodialysis (HD). The incidence of patients on RRT is 125 patients per million population (pmp), a value that increases to 400pmp in the elderly age group (>70 years).1 One of the factors that determines the morbidity and mortality rates in HD patients is the type of vascular access used. The recognised clinical practice guidelines, the European Best Practice Guidelines (EBPG),2 the Kidney Disease Outcomes Quality Initiative (KDOQI),3 and the guidelines from the S.E.N.,4 which are currently under review, establish as quality indicators in HD that the percentage of incident patients with a permanent vascular access point (arteriovenous fistula [AVF]) must be 50% (KDOQI) vs 80% (S.E.N.). Also, the percentage of prevalent patients with AVF in HD units must reach 80% (S.E.N), and the number of prevalent patients with a tunnelled central venous catheter (CVC) must be less than 10% (S.E.N. and KDOQI). Several different studies have analysed the conditions in which HD patients are treated, including vascular accesses. The DOPPS study,5,6 in its various phases, describes a progressive increase in the use of CVC in incident patients on HD. By 2007, based on a representative sample of Spanish patients participating in this study, there was a greater relative risk (RR) of death (RR: 1.2) associated with CVC than with AVF.7 The ANSWER study8 in 2006 also reported high percentages of CVC use in incident HD patients (41%), similar to those described in 2006 by the Spanish Group for CKD9 in a sample of 1504 incident patients on RRT from 35 Spanish hospitals. More recently, in 2009, Gruss et al published a prospective study including 260 incident patients on HD and reported up to 47% of patients with CVC. They also observed high mortality rates (hazard ratio [HR]: 1.86) associated with this type of vascular access, which increased with the duration of use.10 The aim of our study was to evaluate the current situation in our clinical practice as regards the use of CVC in HD units, and the high mortality associated with this technique, comparing it to the published results from our field of medicine. MATERIAL AND METHOD We examined the vascular accesses in 299 prevalent patients on HD (62% males) at our unit of the Dr. Negrín University Hospital, Gran Canaria (HUGCDN) in October 2009, as well as the vascular accesses for the 422 incident patients on HD (66% males) between January 2004 and October 2009. We analysed the underlying nephropathy and associated diseases in each case. We also compared the results with the objectives set out by the clinical guidelines and with the results published in the mentioned studies. In the sample of incident patients, we did not include those with acute renal failure without previous CKD that started emergency HD and then recovered renal function. We analysed where the incident patients were referred from and how this affected the vascular access point at the start of HD, as well as the percentage of deaths in incident patients. We also calculated the RR of death associated with the use of CVC at the start of HD, and adjusted values for other factors of comorbidity. We used SPSS software, version 17, for statistical analyses. We performed a descriptive analysis of the continuous and non-continuous variables. In order to compare for differences between the means of continuous variables, we used Student’s t-tests. We compared survival curves for incident patients on HD with CVC or AVF using KaplanMeier analyses and log-rank tests. We used a Cox regression analysis to evaluate whether other factors were influencing mortality in incident HD patients. RESULTS Prevalent patients Of the total 299 prevalent patients in October 2009, 67% were undergoing dialysis through a permanent vascular access, and 33% through a CVC (Table 1). Incident patients Of the 422 patients that started RRT on HD, 93% were incident RRT patients, 6% came from peritoneal dialysis (PD), and 1% were patients with non-functioning kidney transplants. The mean age of this patient group was 62 years. Table 1. Vascular accesses used in prevalent patients. Comparison with guideline recommendations Vascular access S.E.N. recommendatioNs (%) KDOQI recommendations (%) DOPPS (%) Dr. Negrín hospital (%) 64 AV fistula >80 >40 81 Synthetic graft <10 ------- 10 3 CVC <10 <10 8 33 Vascular accesses in prevalent patients at our haemodialysis unit (%) compared to the recommendations established in the guidelines. AV: arteriovenous; CVC: central venous catheter 104 Nefrologia 2012;32(1):103-7 Gloria Antón-Pérez et al. Vascular accesses in haemodialysis When we analysed our sample by year, the annual incidence rate was fairly homogeneous, ranging between 62 patients in 2007 and 82 in 2008. In this group, 53.8% were diabetic, 91.7% had arterial hypertension, 28.2% had ischaemic heart disease of a known origin, and 12.6% had peripheral arterial disease, based on the data from the Canary Islands Renal Patient Registry.11 As regards where the patients came from, 42% were derived from advanced chronic kidney disease (ACKD) visits, 15% from other nephrological visits, 1% were transplant recipients with graft loss/dysfunction, 6% were from PD, 15% were from hospital emergency departments, and 21% were from other hospital departments. The patients started HD with a permanent vascular access in 31% of cases (AVF/PTFE [polytetrafluoroethylene] graft); and 77% of them (24% of the total) came from ACKD visits. Patients started HD with a CVC in 69% of cases, 74% of which were derived from other nephrological visits or other departments (Table 2). We compared the vascular accesses in incident patients from our unit with the recommendations from the guidelines and results from previously published studies (Table 3). We compared survival curves between the group of incident HD patients with CVC and the group with AVF (Figure 1). The RR of death was 3.68 times greater in patients that started HD using a CVC (95% confidence interval [CI]: 2.93-6.35). Using the Cox regression analysis (Table 4), we examined which other factors may influence the mortality of our patients, and found a significant association with age (HR: 1.02) and the presence of diabetes mellitus (HR: 1.5), with an HR value associated with starting HD with a CVC of 2.85. We found no significant differences between the mean age of incident patients starting HD with a CVC and those starting with an AVF (62.7 years vs 61.2 years, respectively). When comparing our results from October 2009 with those already analysed from our department in December 2005 and November 2007, we found no significant differences in the use of AVF and/or CVC in prevalent patients, although the use of CVC has progressively increased by 27%, 29%, and 33%, respectively, and the use of AVF has increased by 72%, 71%, and 67%. short originals DISCUSSION There is a wealth of evidence in the literature regarding the mortality of patients on HD with a series of potentially modifiable factors, such as planning the entrance into HD, where patients are referred from, adequate control of phosphorous-calcium metabolism, anaemia and nutrition, and of course, the type of vascular access used.5-8,10,12 The studies published in our field have produced varying results in the level of compliance with the recommendations set forth by the clinical practice guidelines in terms of the type of vascular access to be used in HD, but they certainly are far from perfect.5-10 Are these recommendations realistic? Many authors have analysed the possible causes of the disparities observed:13 - The current real profile of an incident HD patient: elderly, with multiple comorbidities. - The characteristics of the various reference centres, the existence of ACKD visits, the formation of multidisciplinary teams (nephrologists, vascular surgeons, and interventional radiologists). - The equipment for monitoring vascular accesses in HD units. - Protocols for early action, given the complications in permanent vascular accesses. In our daily clinical practice, despite the fact that 42% of our patients are referred from ACKD visits, which would imply the ability to choose the correct technique, scheduled entrance, and a better control of other factors (anaemia, nutrition, etc.) and other preparations,12 up to 18% of these patients start HD with a CVC. In this group of patients, we registered at least one case of attempting a permanent vascular access (AVF) that failed. The incident population on HD in our sample had a mean age similar to that reported in other studies (62 years), although the incidence of diabetic nephropathy in our study was much higher (42% vs 21.5%),10,11 as is also shown in the most recent registries of patients on RRT published by the S.E.N. in 2009.14 One could interpret that these comorbidities are causing the elevated use of CVC on HD, but other authors have shown Table 2. - Vascular accesses in incident patients according to departments of origin Vascular access AV fistula Synthetic graft CVC ACKD (%) 23 1 18 NEPH (%) 3 0 12 Other (%) 4 0 39 Type of vascular access (%) according to the origin of incident patients on HD at our unit between January 2004 and October 2009. AV: arteriovenous; CVC: central venous catheter; ACKD: advanced chronic kidney disease visits; NEPH: Other nephrological visits. Nefrologia 2012;32(1):103-7 105 Gloria Antón-Pérez et al. Vascular accesses in haemodialysis short originals Table 3. Vascular accesses in incident patients. Comparison with guideline recommendations and previous studies Vascular access S.E.N. recommendations (%) KDOQI recommendations (%) DOPPS (%) ANSWER (%) Spanish Group CKD (%) HUGCDN(%) AV fistula 80 50 71 54 40 30 Synthetic graft <10 ------- 5 -------- 1 1 CVC <10 >10 24 30 43 69 Comparison of the level of compliance with S.E.N. and KDOQI guideline recommendations regarding vascular accesses in incident patients on HD (%) for the different studies cited and our own findings. AV: arteriovenous; CVC: central venous catheter; HUGCDN: Dr. Negrín University Hospital, Gran Canaria that, even in the non-diabetic population with no associated cardiovascular comorbidities and with nephrological monitoring previous to HD, the percentage of incident patients starting with a CVC is high (31.4%).15 Also, the difference in the mortality rates from patients with a CVC and those with an AVF is dependent on time, and is maintained even after adjusting for other comorbidity factors.10 In our study, we showed that elderly and diabetic patients die at a greater rate, but the factor most highly associated with mortality was starting HD with a CVC. A decrease in the use of this type of vascular access should be a primary objective, necessitating the formation of multidisciplinary teams (nephrologists, vascular surgeons, and interventional radiologists) and coordination for the monitoring of patients with advanced chronic kidney disease. Additionally, HD units should be equipped with the means to adequately monitor the permanent vascular accesses used and to guarantee emergency treatment of these, avoiding the need for CVC. However, in our clinical experience, although we should optimise the monitoring of vascular accesses and use protocols for early action in the case of complications, we do have the impression that there is a sub-group of patients with CVC following several failed attempts at establishing a permanent access point, probably due to their precarious vascular system. This all leads us to reflect on the indication for this technique to be used in select populations: is the survival of these patients (diabetics, elderly, with compromised vascular system analysed using a venous map) greater when following a conservative treatment for CKD? CONCLUSIONS Throughout the years encompassed by our study, we have not been able to achieve the objectives set out by the different guidelines in terms of the prevalence and incidence 1.0 Cumulative survival 0.8 Table 4. Factors related to mortality. Cox regression. 0.6 0.2 0.0 0.00 20.00 40.00 Time 60.00 AVHD1_R CATHETER AVF CATHETER-censored AVF-censored Kaplan-Meier survival curves. Survival of patients with an arteriovenous fistula vs those with a central venous catheter Figure 1. Survival curves 106 P OR (95% CI) Age upon starting HD 0.014 1.020 (1.004-1.037) Male 0.292 1.225 (0.840-1.786) DM 0.050 1.504 (0.999-2.264) AHT 0.439 0.705 (0.291-1.709) IC 0.445 1.153 (0.800-1.663) PAP 0.620 1.126 (0.705-1.7961) VAHD 0.000 2.825 (1.744-4.576) 0.4 DM: Diabetes Mellitus. AHT: Arterial hypertension. IHD: ischaemic heart disease. PAD: peripheral arterial disease. VAHD: vascular access at the start of HD (CVC). Nefrologia 2012;32(1):103-7 Gloria Antón-Pérez et al. Vascular accesses in haemodialysis short originals of vascular accesses for HD. The high mortality associated with a start of HD using a CVC (RR: 3.68), independently of other factors, makes the decrease in the use of this type of vascular access a primary objective, taking into account the resources used and potential costs. Probably, the high prevalence of diabetic nephropathy in our field negatively influences the achievement of these objectives. could improve these results, as appears to be the case in isolated experiences. The implementation in our hospitals of monographic ACKD visits and the creation of multi-disciplinary teams (nephrologists, vascular surgeons, and vascular radiologists) Conflicts of interest Acknowledgements: We would like to thank Dr José Carlos Rodríguez Pérez for his efforts in reviewing and correcting this manuscript. Authors declare no potential conflicts of interest. REFERENCES 1. Registro Español de Enfermos Renales 2007. Congreso Nacional de Nefrología, San Sebastián, 2008. [Available at www. senefro.org]. 2. Tordoir J, Canaud B, Haage P, Konner K, Basci A, Fouque D, et al. EBPG on vascular Access. Nephrol Dial Transplant 2007; 22(Suppl 2): 88–117. 3. National Kidney Foundation. KDOQI Clinical Practice Guidelines for Hemodialysis Adequacy, 2000. Am J Kidney Dis 2001;37:S7-S64. 4. Guías SEN de Centros de Hemodiálisis. Nefrología 2006;6(Supl. 8). 5. Young E, Goodkin DA, Mapes DL, Port FK, Keen ML, Chen K, et al. The Dialysis Outcomes and Practice Patterns Study (DOPPS). An international hemodialysis study. Kidney Int 2000;57(Suppl. 74):S74-81. 6. Pisoni RL, Arrington CJ, Albert JM, Ethier J, Kimata N, Krishnan M, et al. Facility hemodialysis vascular access use and mortality in countries participating in DOPPS: an instrumental variable analysis. Am J Kidney Dis 2009;53(3):475-91. 7. Piera L, Cruz JM, Braga-Gresham JL, Eichleay MA, Pisoni RL, Port FK. Estimación, según el estudio DOPPS, de los años de vida de pacientes atribuibles a las prácticas de hemodiálisis modificables en España DOPPS Study. Arbor Research Collaborative for Health. Nefrología 2007;27(4):496-504. 8. Pérez-García R, Martín-Malo A, Fort J, Cuevas X, Lladós F, Lozano J, et al., and on behalf of all Investigators from the ANSWER study. Baseline characteristics of an incident haemodialysis population in Spain: results from ANSWER—a multicentre, prospective, observa- tional cohort study. Nephrol Dial Transplant 2009;24(2):578-88. 9. Marrón B, Ortiz A, de Sequera P, Martín-Reyes G, de Arriba G, Lamas JM, Martínez Ocaña JC, Arrieta J, Martínez F; Spanish Group for CKD. Impact of end-stage renal disease care in planned dialysis start and type of renal replacement therapy--a Spanish multicentre experience. Nephrol Dial Transplant. 2006 Jul;21 (Suppl 2):ii51-5. 10. Gruss E, Portolés J, Tato A, Hernández T, López-Sánchez P, Velayos P, et al. Repercusiones clínicas y económicas del uso de catéteres tunelizados de Hemodiálisis en un Area Sanitaria. Nefrología 2009;29:123-9. 11. RERCAN. Registro de las enfermedades renales de Canarias. Sociedad Canaria de Nefrología (Informe de la Sociedad), 2008. 12. Marrón B, Martínez Ocaña JC, Salgueira M, Barril G, Lamas JM, Martín M, et al. Analysis of patient flow into dialysis: role of education in choice of dialysis modality. Perit Dial Int 2005;25(Suppl. 3):S56-59. 13. Roca Tey R. El Acceso Vascular para Hemodiálisis. La asignatura pendiente. Nefrología 2010;30:280-7. 14. Registro Español de Enfermos Renales 2009. Congreso Nacional de Nefrología, Granada, 2010. [Available at: www. senefro.org]. 15. Arcos E, Comas J, Deulofeu R y la Comisión de seguimiento del Registre de Malalts Renals de Catalunya. Datos del Registre de Malalts Renals de Catalunya. Grupos de trabajo: accesos vasculares. [Available at: www.senefro.org]. Send for review: 27 Jun. 2011 | Accepted: 22 Oct. 2011 Nefrologia 2012;32(1):103-7 107 short reviews http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society Changes in body composition parameters in patients on haemodialysis and peritoneal dialysis M. Cristina Di-Gioia, Paloma Gallar, Isabel Rodríguez, Nuria Laso, Ramiro Callejas, Olimpia Ortega, Juan C. Herrero, Ana Vigil Servicio de Nefrología. Hospital Severo Ochoa. Leganés, Madrid (Spain) Nefrologia 2012;32(1):108-13 doi:10.3265/Nefrologia.pre2011.Oct.10938 ABSTRACT Cambios en los parámetros de composición corporal en pacientes en hemodiálisis y diálisis peritoneal RESUMEN Introduction: Proper hydration is one of the major aims in haemodialysis (HD) and peritoneal dialysis (PD). Bioimpedance spectroscopy appears to be a promising method for the evaluation and follow up of the hydration status in dialysis patients (P). Objectives: We compared body composition between stable patients on HD and PD after six months. Patients and method: An observational study with 62 P on HD and 19 P on PD was performed. Clinical, biochemical and bioimpedance parameters were analysed. Results: In the comparative study, PD P were younger (50±10 vs 57±14 years, P=.031). The Charlson Comorbidity Index (4.8±3 vs 7.5±3, P<.001), time on dialysis (16.9±18.01 vs 51.88±68.79 months, P=.020) and C-Reactive Protein [3 (3-9.3) vs 5.25 (1-76.4)] were lower. Total protein levels (7.46±0.44 vs 7.04±0.55 g/dl, P=.005) and transferrin levels (205±41 vs 185±29 mg/dl, P=.024) were higher. BIS: Intracellular water (19.67±3.61 vs 16.51±3.36 litres, P=.010), lean tissue mass (LTM) (37.20±8.65 vs 32.57±8.72 kg, P=.029), total cellular mass (TCM) (20.53±5.65 vs 17.56±5.91 kg, P=.033), and bioelectrical impedance phase angle (Phi 50) (5.81±0.86 vs 4.74±0.98, P=.000) were higher than in HD P. Overhydration: 22% in HD y 10% in PD, in conditions referred to in methods. Six months later, PD P increased in weight (73.75±12.27 vs 75.22±11.87 kg, P=.027), total fat (FAT) (26.88±10 vs 30.02±10 kg, P=.011) and relative fat (Rel FAT) (35.75±9.87 vs 39.34±9.12, P=.010); and decreased in ICW (18.56±3.45 vs 17.65±3.69 l, P=.009), LTM (36.95±8.88 vs 34±9.70 kg, P=.008) and relative LTM (Rel LTM) (50.85±12.33 vs 45.40±11.95%, P=.012). In the multivariate analysis, weight variation (∆) was related to ∆ FAT (P<.001). We found a correlation between fat increase and lean tissue mass decrease. Six months later, in HD P, we observed a reduction in ECW (15.11±2.45 vs 14.00±2.45, P=.001), without changes in other parameters. Conclusions: Bioelectrical impedance analysis facilitates the assessment of changes in body composition so as to correct dry weight and to introduce changes in treatment schedule. Introducción: La normohidratación es uno de los mayores objetivos en hemodiálisis (HD) y diálisis peritoneal (DP). La bioimpedancia por espectroscopia (BIS) se postula como el método más prometedor para la evaluación y seguimiento del estado de hidratación en pacientes en diálisis. Objetivo: Comparar la composición corporal de pacientes prevalentes en HD y DP en un intervalo de seis meses. Pacientes y métodos: Estudio observacional de 62 pacientes en HD y 19 en DP comparando los parámetros clínicos, bioquímicos y de bioimpedancia. Resultados: En el estudio comparativo, los pacientes en DP fueron más jóvenes (50 ± 10 vs. 57 ± 14 años, p = 0,031). El índice de comorbilidad de Charlson (4,8 ± 3 vs. 7,5 ± 3, p < 0,001), tiempo en diálisis (16,9 ± 18,01 vs. 51,88 ± 68,79 meses, p = 0,020) y proteína C reactiva [3 (3-9,3) vs. 5,25 (1-76,4)] fueron menores. Los niveles de proteínas totales (7,46 ± 0,44 vs. 7,04 ± 0,55 g/dl, p = 0,005) y transferrina (205 ± 41 vs. 185 ± 29 mg/dl, p = 0,024) fueron más elevados. BIS: agua intracelular (AIC) (19,67 ± 3,61 vs. 16,51 ± 3,36 litros, p = 0,010), masa muscular total (MM) (37,20 ± 8,65 vs. 32,57 ± 8,72 kg, p = 0,029), masa celular total (MCT) (20,53 ± 5,65 vs. 17,56 ± 5,91 kg, p = 0,033) y ángulo de fase (Phi 50) (5,81 ± 0,86 vs. 4,74 ± 0,98, p = 0,000) fueron más elevados que en HD. Sobrehidratados 22% en HD y 10% en DP, en las condiciones referidas en métodos. A los seis meses en DP observamos aumento de peso (73,75 ± 12,27 vs. 75,22 ± 11,87 kg, p = 0,027), grasa total (MG) (26,88 ± 10 vs. 30,02 ± 10 kg, p = 0,011) y relativa (MG %) (35,75 ± 9,87 vs. 39,34 ± 9,12%, p = 0,010); disminución de AIC (18,56 ± 3,45 vs. 17,65 ± 3,69 l, p = 0,009), MM (36,95 ± 8,88 vs. 34 ± 9,70 kg, p = 0,008) y MM relativa (MM %) (50,85 ± 12,33 vs. 45,40 ± 11,95%, p = 0,012). En el análisis multivariante, la variación (∆) de peso guarda relación con el ∆ de grasa (p < 0,001). Encontramos correlación entre el incremento de grasa y el decremento de masa muscular (p = 0,01). A los seis meses en HD no se observaron cambios en estos parámetros, salvo una reducción en el agua extracelular (15,11 ± 2,45 vs. 14,00 ± 2,45, p = 0,001). Conclusiones: BIS permite valorar los cambios en la composición corporal y ayuda a establecer el peso seco e introducir cambios en las pautas de tratamiento. Keywords: Body composition. Overhydration. Haemodialysis. Peritoneal dialysis. Bioimpedance spectroscopy Palabras clave: Composición corporal. Sobrehidratación. Hemodiálisis. Diálisis peritoneal. Bioimpedancia por espectroscopia. Correspondence: M. Cristina Di Gioia Servicio de Nefrología. Hospital Severo Ochoa. Leganés, Madrid. (Spain). [email protected] [email protected] 108 INTRODUCTION Achieving a normal hydration state is one of the primary objectives in haemodialysis (HD) and peritoneal dialysis M. Cristina Di Gioia et al. Body composition in HD and PD (PD) treatments. The concept of dry weight is essential to integrated dialysis therapy.1 The abnormal state of overhydration has been related to arterial hypertension, signs and symptoms of pulmonary and peripheral oedema, heart failure, left ventricular hypertrophy, and other adverse cardiovascular effects.2 The increase in left ventricular mass is correlated with worse cardiovascular evolution in PD patients,3 and it has also been described that hydration state is an important independent predictor for mortality in chronic HD patients.4 It appears essential that dialysis providers have a good strategy for maintaining the euvolemic state of their patients. However, the evaluation of normovolemia is difficult, since there is no method that has been established for use in daily clinical practice. The clinical evaluation of dry weight is the most commonly used method, but this leads to frequent conditions of sub-clinical over-hydration and subhydration, which can cause increased morbidity rates.5 Among the various tests that can be used to measure dry weight, chest x-ray aids in the clinical management of patients, but does not comply with the aims of being rapid and non-invasive; the diameter of the inferior vena cava and its respiratory variations are good measures of preload,6 but they are also influenced by cardiovascular factors such as diastolic dysfunction, pulmonary hypertension, and chronic obstructive pulmonary disease.7 Biochemical markers such as ANP (atrial natriuretic peptide) have a prognostic value and may indirectly reflect overhydration due to its effect on left ventricular mass,5 but these levels appear to depend more on the primary situation of the ventricle. It is difficult to establish the proper concentration in dialysis patients, and values frequently remain elevated in patients considered to be properly hydrated.8 The standard methods for measuring body water such as deuterium and sodium bromide for extracellular water are laborious and are not commonly used in clinical practice. 9 The new bioelectrical impedance analysis techniques are being used for the evaluation and follow-up of hydration state. In a study evaluating the detection limits for different methods used for determining hydration states in dialysis patients, bioimpedance spectroscopy has demonstrated high sensitivity, emerging as the most promising method for a practical treatment of dialysis patients.7 This technique uses the variation in the electrical frequency applied and distinguishes between extracellular water and total body water. The variation in frequency applying currents that range between 5kHz and 1000kHz facilitates the determination of extracellular water (ECW) and total body water (TBW); intracellular water (ICW) is extrapolated by analysing at different frequencies.8,10 It has been well established that the changes produced over time in body weight among peritoneal dialysis patients are due to changes both in body water content and lean tissue/fat Nefrologia 2012;32(1):108-13 short reviews mass. Multifrequency bioelectrical impedance analysis offers the possibility of evaluating body composition and hydration state.11 The body composition monitor (BCM, Fresenius Medical Care) has been validated for use in clinical practice to determine hydration state.10 As measured by BCM, a relative overhydration greater than 15% has been shown to be associated with increased cardiovascular mortality in haemodialysis patients.4 Recently, overhydration has also been correlated with inflammation, malnutrition, and atherosclerosis in PD patients.12 The objective of our study is to compare the body composition of prevalent HD and PD patients in a crosssectional analysis, and to evaluate the changes in these values by performing two studies 6 months apart in both techniques, HD and PD, in a dialysis unit with special attention to maintaining dry weight. METHODS Patients We performed a cross-sectional study of prevalent patients on HD (n=62) and PD (n=19) monitored at the same centre for a period that included two measurements of bioelectrical impedance with an interval of 6 months. The number of patients at the start of the study was 65 on HD and 19 on PD. After 6 months, there were 49 on HD and 14 on PD. All patients were older than 18 years. We excluded those patients with contraindications for bioelectrical impedance: an implanted electronic device, any type of metallic implants, amputation, pregnancy, and lactating women. All patients signed an informed consent that was approved by the ethics committee. Of the 62 patients on HD, 21 used online haemodiafiltration and 41 were on conventional HD. Of the 19 patients on PD, 10 were on automated PD (APD) and 9 were on continuous ambulatory PD (CAPD); icodextrin was administered in 49% of patients on both techniques, and 23 patients (27%) were diabetic. Measurements In the initial cross-sectional study, we compared the following clinical parameters for the two techniques: age, sex, Charlson comorbidity index,13 time on dialysis, weight, body mass index (BMI), and systolic and diastolic blood pressure (BP). 109 short reviews M. Cristina Di Gioia et al. Body composition in HD and PD Laboratory analyses: we measured C-reactive protein (CRP) using immunoturbidimetry, and creatinine, total protein, albumin, transferrin, and haemoglobin were measured using certified methods in the biochemistry department of the Severo Ochoa Hospital. The erythropoietin resistance index was defined as the weekly doses of erythropoietin (U/kg predialysis/dose) divided by haemoglobin (Hb) g/dl. had higher values of total protein (TP) (7.46±0.44g/dl vs 7.04±0.55g/dl, P=.005) and transferrin (205±41mg/dl vs 185±29mg/dl, P=.024) than HD patients. Patients on PD had a residual renal function (RRF) of 5.33±3.89ml/min, diuresis at 1115±758ml/day, and ultrafiltration at 887±445ml/day. The ultrafiltration volume per session of HD patients on the day of bioelectrical impedance analysis was 2372±921ml. We performed the predialysis bioelectrical impedance analysis immediately before the second session of the week, and in PD patients, coinciding with the peritoneal equilibration test and with the peritoneum full. We used a body mass composition analysis device using bioimpedance spectroscopy (BCM, Fresenius Medical Care). The parameters for bioelectrical impedance were: TBW in litres (l), ECW in l, ICW in l, ECW/ICW, lean tissue mass (LTM) in kg, LTM% in percentage, lean tissue mass index in kg/m2, total fat (FAT) in kg, FAT% in percentage, phase angle (Phi 50), total cellular mass (TCM) in kg, and overhydration (OH) in l. The state of OH was calculated by standardising OH to ECW and considering OH to be present if >15%.4 As regards the bioelectrical impedance analysis parameters, PD patients had values of ICW (19.67±3.61 vs 16.51±3.36 litres, P=.010), LTM (37.20±8.65kg vs 32.57±8.72kg, P=.029), TCM (20.53±5.65kg vs 17.56±5.91kg, P=.033) and phase angle (5.81±0.86 vs 4.74±0.98, P=.000) greater than HD patients. Under the conditions the study was being carried out in, a total of 14 HD patients and 2 PD patients were overhydrated (22% and 10%, respectively). The clinical, biochemical, and bioelectrical impedance values were analysed after six months for both dialysis techniques. Statistical Analysis We performed all statistical analyses using SPSS statistical software version 12 (Chicago, Illinois, SL, USA). Normally distributed variables were expressed as a mean and standard deviation, and non-normal variables as a median and range (maximum and minimum). We compared the means between groups using Student’s t-tests or Mann-Whitney U-tests and/or chi-square tests according to the nature of the variable. Categorical variables were expressed as number and percentage. We set the value of statistical significance at P<.05. For the univariate analysis, we used Pearson’s or Spearman’s correlation coefficients, according to the nature of the variable. The multivariate analysis involved linear correlation, considering the increase in weight as the dependent variable, and individually introducing variables that were statistically significant in the univariate analysis. RESULTS We analysed the data from a total of 65 patients on HD and 19 on PD. The demographic, clinical, biochemical, and bioelectrical impedance values from the initial analysis are summarised in Table 1. The patients on PD were younger (50±10 years vs 57±14 years, P=.031), with a lower Charlson comorbidity index (4,8±3 vs 7,5±3, P<.001), less time on dialysis (16.9±18.01 months vs 51.88±68.79 months, P=.020), lower CRP [3 (3-9.3) vs 5.25 (1-76.4)] and 110 Diabetic patients had a higher mean systolic BP (P=.012) and lower phase angle (P=.008), with no difference in the rest of the parameters used to measure body composition. In patients on PD after 6 months (table 2), we observed a significant increase in weight (73.75±12.27kg vs 75.22±11.87kg, P=.027), with an increase in total fat (26.88±10kg vs 30.02±10kg, P=.011) and relative fat (35.75±9.87% vs 39.34±9.12%, P=.010), and decreased ICW (18.56±3.45l vs 17.65±3.69l, P=.009), total LTM (36.95±8.88kg vs 34±9.70kg, P=.008) and relative LTM (50.85±12.33% vs 45.40±11.95%, P=.012). There was a global correlation between the variation (∆) in weight and ∆ in fat, but not with ∆ in extracellular weight. In the multivariate analysis, the ∆ in weight was correlated with ∆ in fat (P<.001). There was also a correlation between the increase in fat and a decrease in LTM (P=.01). In the multivariate analysis that held the decrease in LTM as the dependent variable and a progressive introduction of age, sex, dialysis technique, and BMI, only age had an influence on the decrease in LTM (P=.012). Upon analysing the data according to the technique of PD used, we observed a tendency towards lower increase in fat when on APD (1.58±3.05 vs 3.5±3.05), despite a higher glucose load (206±58 vs 62.98±75.5, P=.006). In patients on HD, only ECW was significantly reduced (P=.001); all other parameters measured using bioelectrical impedance did not vary in the measurements taken over the 6-month interval (Table 3). DISCUSSION This study demonstrates the differences in body composition between patients on HD and those on PD. In the initial study, the differences in the nutritional parameters evaluated (TP, transferrin) and bioelectrical impedance (ICW, LTM, Nefrologia 2012;32(1):108-13 M. Cristina Di Gioia et al. Body composition in HD and PD short reviews Table 1. Clinical, biochemical, and bioelectrical impedance parameters from the initial analysis HD (n=65) PD (n=19) P (value) 57±14 50±10 0.031 Age Sex: % women 34% 39% 0.681 Charlson C I 7.5±3 4.8±3 0.001 Time on dialysis (months) 51.88±68.79 16.9±18.01 0.020 Weight (kg) 68.38±14.26 73.93±14.46 0.108 BMI 25.95±5.88 26.96±4.62 0.401 135±22 127±18 0.164 Systolic BP (mmHg) Diastolic BP (mmHg) 75±13 80.87±8.20 0.011 5.25 (0.6-76.4) 3.00 (3-9.30) 0.113 Creatinine (mg/dl) 8.38±2.21 8.35±1.97 0.954 Total protein (g/dl) 7.04±0.55 7.46±0.44 0.005 CRP (mg/l) Hb (g/l) 11±1.45 12.20±1.23 0.414 3.76±0.37 3.68±0.39 0.424 Transferrin (mg/dl) 185±29 205±41 0.024 ERI (u/kg/patients/Hb) 22±21 8±5 0.010 Albumin (g/dl) TBW (l) 31.80±5.71 34.17±6.4 0.101 ECW (l) 15.31±2.81 15.47±3.11 0.819 ICW (l) 16.51±3.36 19.67±3.61 0.010 ECW/ICW 0.94±0.14 0.83±0.10 0.001 LTM (kg) 35.26±8.72 37.20±8.65 0.029 Relative LTM (%) 49.33±16.16 51.17±11.63 0.621 FAT (kg) 25.19±11.45 26.98±9.82 0.500 Relative FAT (%) 35.52±11.98 35.81±8.86 0.914 TCM (kg) 17.56±5.91 20.53±5.65 0.033 Phi 50 4.74±0.98 5.81±0.86 0.000 Overhydration (OH) (l) 1.15±1.58 –0.69±1.70 Overhydration/ECW >0.15% 14 (22%) 2 (10%) Data are expressed as a mean±standard deviation or range, median (interquartile range or percentage). ECW: extracellular water; ICW: intracellular water; TBW: total body water; PD: peritoneal dialysis; HD: haemodialyis; Charlson C I: Charlson comorbidity index; BMI: body mass index; ERI: erythropoietin resistance index; TCM: total cellular mass; FAT: fat mass; relative FAT: relative percentage of fat mass; LTM: lean total mass; relative LTM: relative percentage of lean total mass; CRP: c-reactive protein; Phi 50: phase angle; OH: overhydration; BP: blood pressure. TCM, and phase angle) can be attributed to the younger age, less time on dialysis, and better nutritional state in the group on PD. Despite this, a significant weight gain is evident over the six-month observation period among patients on PD, which is not produced in patients on HD. The weight gain is primarily in the form of fat mass. These data could indicate on the one hand that the glucose input from PD could be responsible for the fat increase in these patients, and that it is more difficult to control extracellular volume in PD than in HD, as a consequence of the progressive reduction in RRF. Patients on HD experienced a decrease in ECW with no variations in the other parameters measured over the sixmonth period. Nefrologia 2012;32(1):108-13 LTM decreases in patients on PD, probably secondary to the more sedentary lifestyle associated to dialysis, which could contribute to the increase in fat content in PD patients, and also probably indicates the need for a physical exercise regimen. We were surprised that the decrease in LTM was not significant in HD patients, which we believe could be due to the short time span between the two measurements. We will continue with more prolonged follow-up times. In our study, 22% of the HD patients and 10% of the PD patients were overhydrated, according to the criteria established by other publications.4 We did not perform postdialysis bioelectrical impedance analysis, since it requires at least 30 minutes to carry out the procedure and the patients 111 M. Cristina Di Gioia et al. Body composition in HD and PD short reviews Table 2. Evolution after 6 months in the 14 patients on peritoneal dialysis Initial 6 months P ECW (l) 15.57±2.64 15.96±2.66 0.180 ICW (l) 18.56±3.45 17.65±3.69 0.009 ECW/ICW 0.84±0.10 0.91±0.10 0.003 LTM (kg) 36.95±8.88 34±9.70 0.008 Relative LTM (%) 50.85±12.33 45.40±11.95 0.006 26.88±10 30.02±10 0.011 FAT (kg) Relative FAT (%) 35.75±9.87 39.34±9.12 0.012 LTMI (kg/m2) 13.31±2.58 12.19±2.80 0.008 Weight (kg) 73.75±12.27 75.22±11.87 0.027 RRF (ml/min) 6.06±3.58 5.75±4.44 0.608 Diuresis ml/24h 2500±1115 2200±1030 0.742 887±445 871±432 0.828 Ultrafiltration cc/d Data expressed as mean±standard deviation or percentage. ECW: extracellular water; ICW: intracellular water; RRF: residual renal function; LTMI: lean total mass index; FAT: fat mass; relative FAT: relative percentage of fat mass; LTM: lean total mass; relative LTM: relative percentage of lean total mass. would not consent. Even so, in the case of HD, we evaluated the level of overhydration in the patients’ maximum state of overhydration (pre-HD), and we doubt that the two situations would be comparable. When pre and post-HD measurements were available, and overhydration was calculated over the mean at centres that took similar care to reach dry weight in each dialysis session, 10% of patients were overhydrated,11 similar to our patients on PD. Our results indicate that our patients on PD gain weight above all due to fat increase. Additionally, a slight increase in ECW, probably due to a slight decrease in diuresis, contributes to the weight gain observed, although these changes were not statistically significant. In our patients on PD, the prevalence of overhydration (10%) was lower than reported from other centres.15,16 As has been shown, BP control is harder on automated PD than CAPD due to a worse negative sodium balance on APD resulting from a sodium sieving coefficient in the peritoneal membrane.17,18 The use of icodextrin can favour improved control of the volume situation and reduce left ventricular mass.19 In our experience,20 there is no difference in controlling BP or residual renal function between the two types of PD, probably due to the insistence in restricting salt from the diet and ample use of icodextrin. The tendency in our patients on APD was for a lower increase in fat, despite a greater glucose load than in CAPD, Table 3. Evolution after 6 months in the 49 patients on haemodialysis ECW (l) ICW (l) ECW/ICW LTM (kg) Relative LTM (%) FAT (kg) Relative FAT (%) LTMI (kg/m2) Weight (kg) Initial 15.11±2.45 15.75±3.07 0.97±0.136 31.04±8.22 49.14±16.99 23.94±10.99 35.05±12.79 11.82±2.60 65.63±12.89 6 months 14.61±2.45 15.69±3.46 0.49±0.135 30.86±9.10 48.81±17.32 24.26±11.27 35.82±12.74 11.75±3.00 64.71 ±13.70 P 0.001 0.797 0.112 0.112 0.782 0.820 0.725 0.470 0.076 Data expressed as mean±standard deviation or percentage. ECW: extracellular water; ICW: intracellular water; LTMI: lean total mass index; FAT: fat mass; relative FAT: relative percentage of fat mass; LTM: lean total mass; relative LTM: relative percentage of lean total mass. 112 Nefrologia 2012;32(1):108-13 M. Cristina Di Gioia et al. Body composition in HD and PD perhaps due to the reduced time the glucose spends in the peritoneal cavity and thus lower total absorption. The high prevalence of arterial hypertension and volume overload in HD centres and the difficulty for establishing dry weight in dialysis patients21 situates bioelectrical impedance as another tool for evaluating the changes suffered in body composition that can orient the physician to establish dry weight in HD patients and to introduce changes in the liquids provided to PD patients. Conflicts of interest Authors declare no potential conflicts of interest. REFERENCES 1. Charra B. “Dry weight” in dialysis: the history of a concept. Nephrol Dial Transplant 1998;7:1882-5. 2. Wizemann V, Schilling M. Dilemma of assessing volume state- the use and limitations of a clinical score. Nephrol Dial Transplant 1995;10:2114-7. 3. Wang X, Axelsson J, Lindholm B, Wang T. Volume status and blood pressure in continous ambulatory peritoneal dialysis patients. Blood Purif 2005;23:373-8. 4. Wizemann V, Wabel P, Charnney P, Zaluska V, Moissl U, Rode C, et al. The mortality risk of overhydration in haemodialysis patients. Nephrol Dial Transplant 2009;24:1574-9. 5. Kooman J, van der Sande F, Leunissen K. Wet or dry in dialysis- Can new tecnologies help? Semin Dial 2009;22(1):9-12. 6. Cherlex E, Leunissen K, Janssen J. Echografy of the inferior vena cava is a simple and reliable tool for estimation of “dry weight” in haemodialysis patients. Nephrol Dial Transplant 1989;4:563-8. 7. Kraemer M, Rode C, Wizemann V. Detection limit of methods to assess fluid status changes in dialysis patients. Kidney Int 2006;69:1609-20. 8. Jaeger J, Metha R. Assessement of dry weight in hemodialysis. An overwiew. J Am Soc Nephrol 1999;10:392-403. 9. Leunissen K, Kouw P, Kooman J, Cheriex EC, deVries PM, Donker AJ, et al. New techniques to determine fluid status in hemodialysis patients. Kidney Int Suppl 1993;41:S50-6. short reviews 10. Chamney P, Kramer M, Rode C, Kleinekofort W, Wizemann V. A new technique for establishing dry weight in hemodialysis patients via whole body bioimpedance. Kidney Int 2002;61:2250-8. 11. Devolder I, Verleysen A, Vijt D, Vanholder R, Van Biesen W. Body composition, hydration, and related parameters in hemodialysis versus peritoneal dialysis patients. Perit Dial Int 2010;30:208-14. 12. Demirci MS, Demirci C, Ozdogan O, Kircelli F, Akcicek F, Basci A, et al. Relations between malnutrition-inflamation-atherosclerosis and volume status. The usefulness of bioimpedance analysis in peritoneal dialysis patients. Nephrol Dial Transplant 2011;26(5):1708-16. 13. Charlson M, Pompei P, Ales k, MacKenzie CR. A new method of classifiying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis 1987;40:373-83. 14. Wabel P, Moissl U, Chamney P, Jirka T, Machek P, Ponce P, et al. Towards improved cardiovascular management: the necessity of combining blood pressure and fluid overload. Nephrol Dial Transplant 2008;23:2965-71. 15. Lindley E, Devine Y, Hall L, Cullen M, Cuthbert S, Woodrow G, et al. A ward bases peocedure for assessment of fluid status in peritoneal dialysis using bioimpedance spectroscopy. Perit Dial Int 2005;25 Suppl 3:S46-8. 16. Plum J, Schoenicke G, Kleophas W, Kulas W, Steffens F, Azem A, et al. Comparison of body fluid distribution between chronic haemodialysis and peritoneal dialysis patients as assessed by biophysical and biochemical methods. Nephrol Dial transplant 2001;16:237885. 17. Rodríguez Carmona A, Pérez Fontán M. Sodium removal in patients undergoing CAPD and Automated Peritoneal Dialysis. Perit Dial Int 2002;22:705-13. 18. Ortega O, Gallar P, Carreño A, Gutiérrez M, Rodríguez I, Oliet A, et al. Peritoneal sodium mass removal in continuous ambulatory peritoneal dialysis and automated peritoneal dialysis: influence on blood pressure control. Am J Nephrol 2001;21:189-93. 19. Konings CJ, Kooman J, Schonck M, Gladziwa U, Wirtz J, van den Wall Bake AW, et al. Effect of icodextrin on volume status, blood pressure and echocardiographic parameters: A randomozed study. Kidney Int 2003;63:1556-63. 20. Gallar P, Ortega O, Rodríguez I, Mon C, Ortiz M, Herrero JC, et al. Control de la tensión arterial y balance peritoneal de sodio en DPCA y DPA. VI Reunión Anual de Diálisis Peritoneal. Vitoria, 2008. Libro de resúmenes. 21. Passauer J, Petrov H, Schleser A, Leitch J. Evaluation of clinical dry weight assessment in haemodialysis patients using bioimpedance spectroscopy: a cross-sectional study. Nephrol Dial Transplant 2010;25:545-51. Sent for review: 8 Jun. 2011 | Accepted: 22 Oct. 2011 Nefrologia 2012;32(1):108-13 113 letters to the editor http://www.revistanefrologia.com © 2012 Revista Nefrología. Official Publication of the Spanish Nephrology Society A) COMMENTS ON PUBLISHED ARTICLES The role of interleukin 6 in the pathogenesis of hyponatremia associated with Guillain-Barré syndrome Nefrologia 2012;32(1):114 doi: 10.3265/Nefrologia.pre2011.Oct.11115 To the Editor, We read with great interest the contribution by Monzón et al.1 They reported a significant case of a man who had Guillain-Barré syndrome (GBS) with syndrome of inappropriate antidiuretic hormone (SIADH) and speculated that increased sensitivity to vasopressin in the renal tubule and a long-lasting hypoosmolarity or antidiuretic substances might cause GBS-related SIADH. However, we would like to add a possible pathomechanism in the development of hyponatremia associated with GBS. Furthermore, Mastorakos et al.6 reported that plasma antidiuretic hormone levels were elevated after IL-6 injection in cancer patients, suggesting that IL-6 activated the magnocellular ADHsecreting neurons and that it might be involved in SIADH. Activation of the subfornical organ and the organum vasculosum of the lamina terminalis by IL-6 could eventually lead to thirst and increased vasopressin secretion by neurons from the supraoptic nucleus and The paraventricular nucleus.4 combination of antidiuresis and increased water intake may result in hyponatremia. Therefore, there is a possibility that IL-6 may play a central role in the pathogenesis of hyponatremia associated with GBS. However, further studies are necessary to elucidate if IL6 crosses the blood-brain barrier (BBB), or whether lipopolysaccharides cross the BBB and then increase IL-6 locally in the brain in the future. Conflicts of interest According to a previous study by Maimone et al.,2 interleukin (IL)-6, a multifunctional cytokine, might be implicated in the immunopathogenesis of GBS. In their study, serum IL-6 levels were increased in six (26%) of 23 GBS patients, and detectable levels of IL-6 were also found in the cerebrospinal fluid in 13 (57%).2 Using enzyme-linked immunospot assays, Press et al.3 found elevated numbers of IL-6-secreting blood mononuclear cells during the acute phase in patients with GBS. Quite recently and importantly, Swart et al.4 depicted the cascade-like fashion of events initiated by an inflammatory stimulus (lipopolysaccharides), with tumor necrosis factor-α secreted first, IL-1β second, and IL-6 last, suggesting possible pathways connecting IL-6 to vasopressin release. These proinflammatory cytokines are secreted into the systemic circulation to initiate the acute phase response which is involved in the innate immune system.5 114 The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Monzón Vázquez T, Florit E, Marqués Vidas M, Rodríguez Cubillo B, Delgado Conde P, Barrientos Guzmán A. Syndrome of inappropriate antidiuretic hormone hypersecretion associated with Guillain-Barré syndrome. Nefrologia 2011;31:498-9. 2. Maimone D, Annunziata P, Simone IL, Livrea P, Guazzi GC. Interleukin-6 levels in the cerebrospinal fluid and serum of patients with Guillain-Barré syndrome and chronic inflammatory demyelinating polyradiculoneuropathy. J Neuroimmunol 1993;47:55-61. 3. Press R, Ozenci V, Kouwenhoven M, Link H. Non-T(H)1 cytokines are augmented systematically early in Guillain-Barré syndrome. Neurology 2002;58:476-8. 4. Swart RM, Hoorn EJ, Betjes MG, Zietse R. Hyponatremia and inflammation: the emerging role of interleukin-6 in osmoregulation. Nephron Physiol 2011;118:45-51. 5. Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med 1999;340:448-54. 6. Mastorakos G, Weber JS, Magiakou MA, Gunn H, Chrousos GP. Hypothalamicpituitary-adrenal axis activation and stimulation of systemic vasopressin secretion by recombinant interleukin-6 in humans: potential implications for the syndrome of inappropriate vasopressin secretion. J Clin Endocrinol Metab 1994;79:934-9. Se Jin Park1, Ki Soo Pai1, Ji Hong Kim2, Jae II Shin2 Department of Pediatrics. Ajou University School of Medicine, Ajou University. Suwon (Korea). 2 Department of Pediatrics. Yonsei University College of Medicine, Severance Children’s Hospital. Seoul (Korea). Correspondence: Jae II Shin Department of Pediatrics. Yonsei University College of Medicine. Severance Children’s Hospital, 250 Seongsan-ro, Seodaemun-gu, 120-752, Seoul, Korea. [email protected] 1 Acyclovir and valacyclovir neurotoxicity in patients with renal failure Nefrologia 2012;32(1):114-5 doi:10.3265/Nefrologia.pre2011.Nov.11247 To the Editor, It was with great interest that we read the article by Quiñones et al1 in which they mention how toxicity secondary to starting new treatments in patients with renal failure can give rise to false diagnoses. One of the patients cited by the authors suffered from neurotoxicity due to acyclovir. Acyclovir and its ester, valacyclovir, are widely used in treating infection with the varicella zoster virus, Nefrologia 2012;32(1):114-32 letters to the editor and its Summary of Product Characteristics lists neurotoxicity as an extremely rare event. However, we have observed 3 episodes similar to that described by Quiñones et al in patients on haemodialysis receiving acyclovirvalacyclovir for metameric herpes zoster. Case 1. Female patient aged 61 years treated with oral acyclovir at 800mg/12 hours. After the third dose, she experienced a psychotic reaction with visual hallucinations and dysarthria. Antiviral treatment was suspended and the psychiatric symptoms resolved completely in 3 days. Case 2. Male patient aged 66 years undergoing treatment with oral valacyclovir (500mg/12 hours). After the second dose, he presented dysarthria and reduced consciousness. In light of a possible case of herpesviral encephalitis, treatment was changed to IV acyclovir at 400mg/day, with no noticeable response. The level of consciousness improved after each haemodialysis session, and then decreased again. When we suspected neurotoxicity caused by the antiviral agent, we reduced the acyclovir dose to 200mg/day and started daily haemodialysis sessions; the patient improved progressively and had recovered completely by the ninth day. Case 3. Female patient aged 83 years who was treated with valacyclovir at 1g/12 hours as prescribed by her general practitioner. Dysarthria began following the third dose. Valacyclovir was suspended and the patient underwent daily haemodialysis during 3 days, the speech disorder resolving completely. This last patient received a high dose of valacyclovir, but in the other two patients, acyclovir and valacyclovir doses were adjusted according to the stage of renal failure. A correlation between toxicity and plasma drug levels is under debate. Some authors state that there is a higher risk of toxicity when levels exceed 20 Nefrologia 2012;32(1):114-32 micromoles per litre,2 but others claim not to have witnessed symptoms in patients with levels greater than 30 micromoles, and it is therefore impossible to establish a safe therapeutic range.3 Furthermore, the early onset of the neurological symptoms was remarkable in our three cases: in all of the patients, symptoms appeared on the second day of treatment after the second or third oral dose, which would suggest that the cause was drug idiosyncrasy rather than drug accumulation. Haemodialysis was effective in reducing the levels of acyclovir and its metabolites.4 This is the most effective treatment for this type of neurotoxicity, and it is an important tool for the differential diagnosis of acyclovir neurotoxicity and viral encephalitis.2,5 The appearance of neurological or psychiatric changes in these patients should be taken into account in order to prevent misdiagnosis, as occurred in our own case 2 and in the case described by Quiñones et al. observational study. Nephrol Dial transplant 2003;18:1135-41. 5. Peces R, de la Torre M, Alcázar R. Acyclovir-associated encephalopathy in haemodialysis patients. Nephrol Dial Transplant 1996;11:752. Gloria Ruiz-Roso, Antonio Gomis, Milagros Fernández-Lucas, Martha Díaz-Domínguez, José L. Teruel-Briones, Carlos Quereda Servicio de Nefrología. Hospital Universitario Ramón y Cajal. Madrid. Spain. Correspondence: Gloria Ruiz Roso Servicio de Nefrología. Hospital Universitario Ramón y Cajal, Madrid. Spain. [email protected] Estimating glomerular filtration rate in order to adjust drug doses: confusion abounds Nefrologia 2012;32(1):115-7 doi:10.3265/Nefrologia.pre2011.Dec.11235 Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. To the Editor, Two recent events led to our writing this letter. 1. Quiñones Ortiz L, Suárez Laurés A, Pobes Martínez A, de la Torre M, Torres Lacalle A, Forascepi Roza R. Alerta ante medicación inesperada en hemodiálisis. Nefrologia 2011;31:611-2. 2. Gómez Campderá F, Verde E, Vozmediano MC, Valderrábano F. More about acyclovir neurotoxicity in patients on haemodialysis. Nephron 1998;78:228-9. 3. Haefeli WE, Schoenenberger RA, Weiss P, Ritz RF. Acyclovir-induced neurotoxicity: Concentration-side effect relationship in acyclovir overdose. Am J Med 1993;94:212-5. 4. Helldén A, Odar-Cederlöf I, Diener P, Barkholt L, Medin C, Svensson JO, et al. High serum concentrations of the acyclovir main metabolite 9carboxymethoxymethylguanine in renal failure patients with acyclovir-related neuropsychiatric side effects: an One. For 2 or 3 years now, our local biochemistry laboratories calculate the estimated glomerular filtration rate (eGFR) by means of the MDRD-IDMS formula (formerly MDRD) and the isolated creatinine value, as per National Kidney Foundation recommendations.1 Yet in October 2011, we still observe the following: - The constant used by some biochemistry laboratories for the MDRD-IDMS formula is 186, when it should be 175 since the calculation for the serum creatinine value is standardised by IDMS. - Some laboratories deliver MDRDIDMS results in ml/min instead of ml/min/1.73m2. Although it is dependent on an individual’s body surface area, this could lead one to 115 letters to the editor assume that the measurement is absolute, which could have consequences when adjusting doses. Two. While the new formula for measuring glomerular filtration rate (GFR),2 CKD-EPI, seems to improve on the current MDRD-IDMS formula in both accuracy and precision, it is likely to make things even more confusing when it comes to choosing an equation to adjust a drug dose. Furthermore, an article recently published in this journal comparing the MDRDIDMS and CKD-EPI formulas in a Spanish population3 contained what appears to be an erratum in Table 1, which describes the formulas used to calculate CKD-EPI: for males with creatinine levels >80 micromoles/litre, it states to divide by 0.7, and we believe that it should be by 0.9. In light of all of the above, we would like to make the following observation: From the 1980s until quite recently, GFR was estimated using the formula published by Cockcroft and Gault (CG) in 1976.4 The result of this equation (an estimation of creatinine clearance) was used to evaluate renal function and adjust the doses of any drugs that so required. We would like to stress that the value obtained by this formula is absolute. This means that it accounts for the individual’s size (since it includes weight among its variables) and gives a result in ml/min (if the body surface area differs greatly from the mean, using ideal rather than true weight is recommended.) In 1999, 23 years after the CG formula was published, Levey published a new formula for estimating GFR: the MDRD.5 Shortly afterwards, in 2002, the KDOQI proposed using this formula for early detection and classification of chronic kidney disease so that patients in earlier stages would have better access to nephrology care.1 The result given by this formula is dependent on body surface area (ml/min/1.73m2), as is also the case with the recently improved MDRDIMDS and CKD-EPI formulas.6,2 Since the result is dependent on a surface area of 1.73m2, we only need the variables age, sex, serum creatinine and race. This formula was recommended by such societies as the Spanish Society of Clinical Biochemistry and Molecular Pathology (SEQC) and the Spanish Society of Nephrology (S.E.N).7 Nevertheless, although generalised use of the MDRD method seems appropriate for categorising individuals in different stages of chronic kidney disease, it causes some problems in adjusting drug doses, especially if the value given by the laboratories is interpreted as an absolute value. If we consider only the relative results (ml/min/1.73m2) given by MDRD, Table 1. Recommended dabigatran dose adjustments according to the glomerular filtration rate estimated by different equations Formula used to estimate GFR Results Units Cockcroft-Gault 31.3 ml/min MDRD-4v 28.6 ml/min/1.73m Recommendation as SmPC Indicated 2 Contraindicated MDRD-4v adjusted for body surface area 35.1 ml/min Indicated CKD-EPI 26.3 ml/min/1.73m2 Contraindicated White male 85 years of age, 180cm height, 90kg, with a serum creatinine level of 2.2mg/dL. Estimated body surface area 2.1m . GFR=Glomerular filtration rate 2 116 MDRD-IMDS or CKD-EPI –those results given by biochemical laboratories– individuals with a body surface area >1.73m2 will have a higher absolute eGFR value. This could lead to underdosing the patient. If the patient’s body surface area is less than 1.73m2, the absolute eGFR will be lower, which could lead to overdosing the patient (Tables 1 and 2). On the other hand, the required dose of a certain drug may vary considerably depending on the equation used to estimate GFR, and this may have clinical repercussions.8 With this in mind, most published drug adjustment guidelines recommend a dose and/or drug interval according to the Cockcroft-Gault formula; very few guidelines make use of MDRD.9 In two recent examples, regulatory authorities based their recommendations on the CG formula: - The Spanish Agency for Medicines and Health Products (AEMPS) followed the European Medicines Agency recommendation and modified the SmPC for Pradaxa® (dabigatran) and issued an informative note on 27 October 2011 reminding doctors of the importance of checking renal function before and after treatment with this new drug. They informed that before starting dabigatran treatment, renal function must be assessed in all patients by calculating creatinine clearance (CrCl) in order to exclude patients with severe renal failure (CrCl<30ml/min).10 - The Food and Drug Administration (FDA)’s safety update of 1 September 2011 stated that the SmPC had been changed and issued a reminder that “Reclast should not be used (is contraindicated) in patients with creatinine clearance less than 35ml/min”.11 The FDA guidelines for the industry simply cite the CG and MDRD equations as being the most commonly used. However, experts do not agree on which of the formulas should be used for adjusting doses in patients with renal Nefrologia 2012;32(1):114-32 letters to the editor Table 2. Recommended daptomycin dose adjustments according to the glomerular filtration rate estimated by different equations Formula used to estimate GFR Cockcroft-Gault MDRD-4v Results Units Recommendation as SmPC 21.6 ml/min /48h 33 ml/min/1.73m2 /24h MDRD-4v adjusted for body surface area 27.5 CKD-EPI 31.5 ml/min ml/min/1.73m 11. /48h 2 /24h 12. White female 85 years of age, 150cm height, 50kg, with a serum creatinine level of 1.5mg/dl. Estimated body surface area 1.4m2. GFR=Glomerular filtration rate failure. Some advocate using the equation recommended by the pharmaceutical manufacturer, particularly in the case of elderly patients,12 while others13,14 state that the MDRD and CG equations are completely interchangeable. In summary, and as a general rule, using the equation recommended by the pharmaceutical manufacturer (mainly CG) seems reasonable. If there is no specific recommendation, the most reliable method of estimating GFR in the target population should be should. Regardless of which equation is used, we must remember that dose adjustments must be made using absolute GFR values, especially for patients whose body surface area differs greatly from 1.73m2. 4. 5. 6. 7. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. National Kidney Foundation. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis 2002;39(2 Suppl 1):S1-266. 2. Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro III AF, Feldman HI, et al., for the CKD-EPI (Chronic Kidney Disease Epidemiology Collaboration). A new equation to estimate glomerular filtration rate. Ann Intern Med 2009;150(9):604-12. 3. Montañés R, Bover J, Oliver A, Ballarín JA, Gracia S. Valoración de la nueva ecuación Nefrologia 2012;32(1):114-32 8. 9. 10. CKD-EPI para la estimación del filtrado glomerular. Nefrologia 2010;30(2):185-94. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron 1976;16(1):31-41. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of Diet in Renal Disease Study Group. Ann Intern Med 1999;130(6):461-70. Levey AS, Coresh J, Greene T, Marsh J, Stevens LA, Kusek JW, et al. Expressing the Modification of Diet in Renal Disease Study equation for estimating glomerular filtration rate with standardized serum creatinine values. Clin Chem 2007;53(4):766-72. Gracia S, Montañés R, Bover J, Cases A, Deulofeu R, Martín de Francisco AL, et al. Recomendaciones sobre la utilización de ecuaciones para la estimación del filtrado glomerular en adultos. Nefrologia 2006;26:658-65. Denetclaw TH, Oshima N, Dowling TC. Dofetilide dose calculation errors in elderly associated with use of the modification of diet in renal disease equation. The Ann Pharmacother 2011;45:e44. Dowling TC, Matzke GR, Murphy JE, Burckart GJ. Evaluation of renal drug dosing: prescribing information and clinical pharmacist approaches. Pharmacotherapy 2010;30(8):776-786. Nota informativa de la AEMPS, del 27 de octubre de 2011, sobre dabigatrán (Pradaxa®) y riesgo de hemorragia: nuevas recomendaciones de vigilancia de la función renal. Available at: http://www.aemps.gob.es/informa/notasI nformativas/medicamentosUsoHumano/seguri 13. 14. dad/2011/NI-MUH_21-2011.htm. [Accessed: Oct/31/2011]. Comunicado de la FDA, del 1 de septiembre de 2011, sobre la seguridad de los medicamentos: Nueva contraindicación y advertencia actualizada sobre el deterioro renal causado por Reclast (ácido zoledrónico). Available at: http://www.fda.gov/Drugs/DrugSafety/ucm27 0199.htm#ref. [Accessed: Oct/31/2011]. Corsonello A, Pedone C, Lattanzio F, Semeraro R, D’Andria F, Gigante M, et al. Agreement between equations estimating glomerular filtration rate in elderly nursing home residents and in hospitalised patients: implications for drug dosing. Age and Ageing 2011;40:583-9. Stevens LA, Nolin TD, Richardson MM, Feldman HI, Lewis JB, Rodby R, et al., on behalf of the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI). Comparison of drug dosing recommendations based on measured GFR and kidney function estimating equations. Am J Kidney Dis 2009;54:33-42. Jones G. Estimating renal function for drug dosing decisions. Clin Biochem Rev 2011;32:81-8. Javier Peral-Aguirregoitia1, Unax Lertxundi-Etxebarria2, Ramon Saracho-Rotaeche3, Sira Iturrizaga-Correcher4, M. José Martínez-Bengoechea5 1 Servicio de Farmacia Hospitalaria. Hospital Galdakao-Usansolo. Galdakao, Vizcaya. Spain. 2 Farmacéutico especialista en Farmacia Hospitalaria. Jefe de Servicio de Farmacia. Hospital Psiquiátrico de Álava. Vitoria-Gasteiz, Álava. Spain. 3 Servicio de Nefrología. Hospital Santiago Apóstol. Vitoria-Gasteiz, Álava. Spain. 4 Laboratorio de Análisis Clínicos. Hospital Txagorritxu. Vitoria-Gasteiz, Álava. Spain. 5 Farmacéutica especialista en Farmacia Hospitalaria. Jefa de Servicio de Farmacia. Hospital Galdakao-Usansolo. Galdakao, Vizcaya. Spain. Correspondence: Javier Peral Aguirregoitia Servicio de Farmacia Hospitalaria. Hospital Galdakao-Usansolo. Barrio Labeaga s/n, 48960 Galdakao, Vizcaya. Spain. [email protected] [email protected] 117 letters to the editor B) BRIEF PAPERS ON RESEARCH AND CLINICAL EXPERIENCES Is peripheral and/or catheter blood necessary for performing haemoculture in haemodialysis patients whose central venous catheter presents bacteraemia? Nefrologia 2012;32(1):118-20 doi:10.3265/Nefrologia.pre2011.Nov.11114 To the Editor, The profile of today’s patients in haemodialysis (HD) programmes has changed. HD patients are now older and have more co-morbidities. The factors responsible for their poorer cardiovascular and immunological health are mainly the increase in diabetes, followed by increased survival rates of patients within the programme. As a result, their blood vessels (arteries and veins) are in worse condition for creating an internal arteriovenous fistula (IAVF), the number of punctures and the risk of infection are higher, and the patients have poorer HD clearance. This leads to increased use of central venous catheters (CVC) and a higher probability of catheter dysfunction, which is accompanied by a higher risk of bacteraemia.1-4 The high percentage of patients who start HD treatment using catheters is well-known. One multi-centre study evaluated the onset of renal replacement therapy in 1504 patients from 35 different Spanish hospitals in 2003, and found that nearly half (46%) began with unscheduled dialysis sessions, and therefore used a CVC. Of these patients, 82% decided to continue with HD.5-6 In the region of the Canary Islands, the 2009 Dialysis and Transplant report by the S.E.N.7 reported an incidence rate of 129 patients per million population (pmp) on renal replacement therapy, with 85% undergoing HD. A total of 226 patients, 118 with a mean age of 66 years, underwent dialysis at our peripheral hospital in 2010; 43% were diabetic, 28% had a tunnelled CVC, and the length of stay was very little for patients with nontunnelled CVC. Despite the outsourcing efforts, we found that there are very long delays in achieving a permanent access site. This is due to delays in both the surgical procedure to create the access and in the arteriovenous fistula maturation time in the population described above. Furthermore, many of these patients refuse surgery repeatedly, and a large percentage do not have the option of a permanent surgical access. Furthermore, this patient population has a high rate of CVC-related bacteraemia.8 In Chapter 1 (procedures prior to creation of a vascular access) of the S.E.N. guidelines for vascular access in haemodialysis (November 2004), we find the following recommendations for preserving the venous network:9 1) Warn the patient about its importance. 2) Provide the patient with a card or recommend wearing a wrist band. 3) Recommend venipuncture in the back of the hand. 4) Use low-plasma laboratory techniques (capillary or dry samples). 5) Make other professionals aware of these problems. 6) Avoid implanting the CVC in the shoulder girdle, and especially in the subclavian vein. 7) Femoral vein catheters are recommended for patients who experience flare-ups in the course of their chronic kidney disease. 8) Stimulate muscular and vascular development through isometric physical exercises or venous dilation techniques. 9) Carefully monitor the venous network of peritoneal dialysis and kidney recipients as well. In kidney recipients, patients and professionals must be made aware of the importance of i) rescuing a thrombosed IAVF and ii) repairing rather than closing elbow IAVF in the absence of congestive heart failure. Haemodialysis unit personnel are aware that bacteraemia due to CVC is the most common complication in vascular accesses. The incidence rate of bacteraemia varies, but it is higher for non-tunnelled catheters (3.8-6.5 per 1000 catheters/day) than for tunnelled catheters. (1.6-5.5 per 1000 catheters/day).10-13 In our peripheral unit, the bacteraemia incidence rate for tunnelled CVC was 1.63 per 1000 catheters/day. Non-hospitalised HD patients (outpatients) and those with a CVC may develop bacteraemia after beginning dialysis, which suggests a systemic influx of bacteria and/or endotoxins from the intraluminal wall of the catheter. We must consider how to take blood samples for blood culture without interrupting dialysis, unless this is necessary due to haemodynamic instability or another major clinical complication. The definitive diagnosis of bacteraemia due to CVC requires that 1 the following criteria are met: - Positive blood cultures that find the same microorganism in the catheter and in a peripheral vein, with a bacterial colony count 5 times higher in the catheter or a difference in bacterial growth of more than 120 minutes. - Cultures of the same microorganism from both the tip of the catheter and from at least one peripheral blood culture. - Cultures of the same microorganism from two different peripheral blood cultures where there is no other source of infection. At least 2 blood cultures taken between 10 and 15 minutes apart. According to section 6.10.2 on infections,9 chapter 6 (central venous catheters) of the S.E.N. guidelines for vascular accesses in haemodialysis Nefrologia 2012;32(1):114-32 letters to the editor (November 2004), when fever is present in a patient with a CVC, samples must be drawn of peripheral blood and from both lumens of the catheter, and samples must be extracted simultaneously and cultured using quantitative techniques if possible. Evidence B. which may be needed for creating a permanent vascular access. At times, venipuncture is a labourious task which does not guarantee an aseptic field, in addition to causing added pain and suffering in a patient already subjected to a number of traumatic procedures. There is no controversy regarding the universal criteria for obtaining blood cultures in patients with CVC and bacteraemia. There is abundant literature on CVC implanted for other reasons, such as for administering drugs, parenteral nutrition and haemodynamic monitoring, as well as CVC in HD patients. However, it does not specify whether patients who have CVC for HD developed bacteraemia after beginning haemodialysis treatment.14-15 Although culture blood samples obtained by venipuncture have been held up as the gold standard for diagnosing bacteraemia, we must consider the extracorporeal system an extension of the circulatory system. It is not likely that there would be significant differences between the blood sample obtained by venipuncture and that extracted from the arterial line of the extracorporeal circuit.19 In this context, complying with criteria for obtaining blood cultures is difficult due to the following reasons: - Blood from the catheter: when dialysis is interrupted, disconnecting both lines to obtain blood cultures may even have an iatrogenic effect, given the risk of infection from handling the catheter16 in a clinical situation that is already complicated. Such a step also entails the possibility of clotting the entire extracorporeal system and wasting precious time, since we do not know if the patient will be able to continue with dialysis treatment or how long the patient will have to wait for the next session following catheter removal. On the other hand, extracting blood samples from both CVC lumens gives rise to false positives in more than 60% of cases. These are related to colonisation of the CVC by microorganisms from the skin.17-18 - Peripheral blood: obtaining peripheral blood in the population described above is very difficult in as much as 40% of all patients, especially if they are under heparin, with the risk of developing haematomas and damage to veins, Nefrologia 2012;32(1):114-32 In order to correctly perform haemodialysis through a catheter, maximum flow rates are required to overcome the deficit due to recirculation (where possible, blood flow rates of more than 300ml/min). Under these conditions, it is likely that large volumes of blood have circulated through the catheter in both directions (arterial and venous) and –when bacteraemia is present– the sample obtained from the catheter will not maintain the quantitative colony differential (with respect to peripheral blood) that is necessary to determine whether the bacteraemia arose in the CVC. However, this is not the case when obtaining samples from CVC implanted for other purposes or tunnelled/non-tunnelled CVC for HD during interdialysis periods. In conclusion, a universal protocol for obtaining blood cultures from the patients described here may do more harm than good, and we believe that the S.E.N. expert committee should review this matter to determine whether or not they should establish an exception. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Wasse H, Kutner N, Zhang R, Huang Y. Association of initial hemodialysis vascular access with patient-reported health status and quality of life. Clin J Am Soc Nephrol 2007;2:708-14. 2. Wasse H, Speckman R, Frankenfiel D, Rocco M,McClellan. Predictors of central venous catheter use at the initiation of Hemodialysis. Semin Dial 2008;21(4):34651. 3. Mermel LA. What is the predominant source of intravascular catheter infections? Clin Infect Dis 2011;52(2):211-2. 4. Matajira T, Félez I, Lacambra I, Azuara M, Álvarez Lipe R, Iñigo P. Endocarditis bacteriana por SAMR en paciente portador de catéter venoso central para hemodiálisis: uso de daptomicina. NefroPlus 2010;3(2):41-5. 5. Gil Cunquero JM, Marrón B. La realidad y la percepción de las infecciones en diálisis. Nefrologia 2010;1(Supl Ext 1):56-62. 6. Marrón B, Ortiz A, de Sequera P, MartínReyes G, de Arriba G, Lamas JM, et al.; Spanish Group for CKD. Impact of endstage renal disease care in planned dialysis start and type of renal replacement therapya Spanish multicentre experience. Nephrol Dial Transplant 2006;21 (Suppl 2):ii51-5. 7. Informe de Diálisis y Trasplante 2009 (Registro Español de Enfermos Renales). Congreso de Granada, 2010. 8. Lee T, Barker J, Allon M. Tunneled catheters in hemodialysis patients: reasons and subsequent outcomes. Am J Kidney Dis 2005;46(3):501. 9. Guías de Acceso Vascular en Hemodiálisis. Sociedad Española de Nefrología. Noviembre de 2004. 10. Weijmer MC, Vervloet MG, Piet M, ter Wee. Compared to tunnelled cuffed hemodialysis catheters, temporary untunnelled catheters are associated with more complications already 2 weeks of use. Nephrol Dial Transplant 2004;19:670-7. 11. Oliver MJ. Acute dialysis catheters. Semin Dial 2001;14(6):432-5. 12. Kairaitis LK, Gottlieb T. Outcome and complications of temporary hemodialysis catheters. Nephrol Dial Transplant 1999;14:1710-4. 13. Robinson D, Suhocki P, Schwab SJ. Treatment of infected tunneled venous access hemodialysis wih guidewire exchange. Kidney Int 1998;53:1792-4. 119 letters to the editor 14. Yébenes JC, Capdevila JA. Infección relacionada con catéteres intravasculares. Med Clin (Barc) 2002;119(13):500-7. 15. León C, Ariza J. Documento de consenso: Guías para el tratamiento de las infecciones relacionadas con catéteres intravasculares de corta permanencia en adultos: conferencia de consenso SEIMCSEMICYUC. Enferm Infecc Microbiol Clin 2004;22(2):92-101. 16. Albalate M, Pérez García R, De Sequera P, Alcázar R, Puerta M, Ortega M, et al. ¿ Hemos olvidado lo más importante para prevenir las bacteriemias en pacientes portadores de catéteres para hemodiálisis? Nefrologia 2010;30(5):573-7. 17. Gaur AH, Fynn PM, Heine DJ, Giannini MA, Shenep JL, Hayden RT. Diagnosis of catheter-related bloodstream infections among pediatric oncology patients lacking a peripheral culture, using differential time to detection. Pediatr Infect Dis J 2005;24:445. 18. Guembe M, Rodríguez-Créixems M, Sánchez-Carrillo C, Pérez-Parra A, Martín-Rabadán P, Bouza E. How many lumens should be cultured in the conservative diagnosis of catheterrelated bloodstream infections? Clin Infect Dis 2010; 50:1575. 19. García CP, Payá GE, Olivares CR, Cotera FA, Rodríguez TJ, Sanz RM. Documento de consenso: Diagnóstico de las infecciones asociadas a catéteres vasculares centrales. Rev Chilena Infectol 2003;20(1):41-50. Juan F. Betancor-Jiménez1, Francisco Alonso-Almán1, Yanet Parodis-López1, Beatriz Quintana-Viñau1, Sonia González-Martínez1, Cristina García-Laverick1, Patricia Pérez-Borges2, José C. Rodríguez-Pérez2 1 Centro de hemodiálisis RTS-GranCanaria. Las Palmas de Gran Canaria. Spain. 2 Servicio de Nefrología. Hospital Universitario de Gran Canaria Dr. Negrín. Las Palmas de Gran Canaria. Spain. Correspondence: José C. Rodríguez Pérez Servicio de Nefrología. Hospital Universitario de Gran Canaria Dr. Negrín. Las Palmas de Gran Canaria. Spain. [email protected] 120 Serial ultrasound of the vascular access Nefrologia 2012;32(1):120-2 doi:10.3265/Nefrologia.pre2011.Nov.11109 To the Editor, Current advances in nephrology and similar advances in other areas of medical knowledge mean that nephrologists must develop technical skills that are not provided by traditional training in nephrology. We present a case that illustrates that fact. The patient in question is an 83 year old male in a conventional haemodialysis (HD) programme with chronic kidney disease secondary to diabetic nephropathy. He had a history of type 2 diabetes mellitus with various diabetes-related complications, arterial hypertension and atypical chest pain with no evidence of ischaemic heart disease. The patient started HD via a tunnelled catheter in February 2010, with good haemodynamic tolerance and adaptation. A left humeral-cephalic arteriovenous fistula (AVF) was created one month later. Following a 30-day maturation period, we began venipuncture in the AVF and observed suboptimal maturation, difficult anatomical interpretation, venous collapse, ´frequent extravasations and impossibility of reaching a blood flow (Qb) greater than 250ml/min. Given these findings, we examined the vascular access (VA) with a portable vascular ultrasound machine (EcoAVP) in the HD room (Figure 1) and observed no stenosis in the arteriovenous fistula and a dual venous system with a collateral vessel branching off 3cm from the arterial anastomosis with a thickness similar to that of the two veins (diameter: 0.39cm vs 0.36cm; area: 0.12 vs 0.14cm2). We found 2 stenoses in the proximal part of the cephalic vein. The fistulography (Figure 2) confirmed the ultrasound findings, a haemodynamically A: cephalic vein; B: collateral vessel to cephalic vein Figure 1. First B-mode ultrasound image of the vascular access in which we see two veins of similar size significant (80%) stenosis at 10cm from the arteriovenous fistula and another smaller one in the proximal third of the cephalic vein. Percutaneous angioplasty was performed on the 2 stenoses with good angiographic results. The identified collateral vessel was not treated in any way. One month later, the AVF had progressed well, allowing for cannulation with no extravasations and an acceptable Qb rate. A second image from the EcoAVP (Figure 3) confirmed the increase in the diameter and the cross-sectional area of the main vein (diameter: 0.5cm, area: 0.24cm2) with a decrease in the size of the collateral vessel (diameter: 0.35, area: 0.08cm2). One year later, the AVF was functioning properly, with a Qb of 350ml/min and a normal venous pressure of 140mmHg. Table 1 shows the changes in some clinical parameters and ultrasound images taken after the treatment with percutaneous angioplasty. The use of an EcoAVP is not common in daily practice. However, it is very useful for approaching, monitoring, and diagnosing AVF complications.1 Ultrasound provides both morphological and functional information in a fast, reliable and noninvasive way, which helps us determine whether percutaneous or surgical treatment is necessary.2 The EcoAVP enables us to combine B-mode imaging, which estimates Nefrologia 2012;32(1):114-32 letters to the editor diagnosis and treatment of VA complications may reduce the number and duration of hospital stays associated with such problems, reduce the use of venous catheters, shorten waiting times for having an AVF, reduce costs derived from diagnostic and therapeutic procedures, and optimise prevention of complications in general.10 A: cephalic vein; B: collateral vessel to cephalic vein Figure 3. Second B-mode ultrasound of the arteriovenous fistula showing an increase in cephalic vein size and decrease in the width of the collateral vessel interventions,4,5,6,7 and estimated venous elastography as a tool that may predict AVF success (limited evidence at present).5 A: cephalic vein; B: collateral vessel to cephalic vein Figure 2. Fistulography image taken after percutaneous angioplasty to both stenoses vein volume, the presence of haematomas, parietal calcifications, intraluminal thrombi, collateral vessels and stenosis, with the Colour Doppler mode, which estimates blood flow, peak systolic velocity, the presence of turbulences, and the shape of pulse waves with the corresponding resistance indices.3 Ultrasound results must always be interpreted in conjunction with clinical findings.3 A broader view of the nephrologists’ participation in decision-making would include using ultrasound for arterial and venous mapping, which has been proven to increase success in surgical Nefrologia 2012;32(1):114-32 At present, guidelines do not set strict criteria for periodical ultrasound assessments of VA or recommend a time to initiate ultrasound monitoring. In some studies, the complications involved in VA failure, which can be detected with a EcoAVP, are present in AVF that still function normally.8 On the other hand, early dysfunction and primary failure in radiocephalic AVF and the frequent delayed maturation in diabetic patients leads us to recommend using a EcoAVP as a monitoring device for all patients on dialysis.4 Considering the increased mean age of patients in dialysis units and data on the high number of complications at any level and any type of VA in elderly patients,4,9 we can state that training in ultrasound examinations should be included in the nephrological curriculum. Active participation of nephrologists in the Despite a certain amount of dependence on specialties such as vascular surgery or interventional radiology in this field, the nephrologist is ultimately responsible for ensuring that the VA works correctly. This responsibility requires strict monitoring and early treatment of VA complications in a multidisciplinary area that encounters frequent administrative obstacles. Proper training in ultrasound examinations will enable professionals to make better treatment decisions in situations in which success depends upon swift action. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Tordoir J, Canaud B, Haage P, Konner K, Basci A, Kooman J, et al. EBPG on Vascular Access. Nephrol Dial Transplant 2007;22 (Suppl 2):ii88-117. 2. Chandra AP, Dimascio D, Gruenewald S, Nankivell B, Allen RD, Swinnen J. Colour duplex ultrasound accurately identifies focal stenoses in dysfunctional autogenousarteriovenous fistulae. Nephrology (Carlton) 2010;15(3):300-6. 3. Kerr SF, Krishan S, Lapham RC, Weston MJ. Duplex sonography in the planning and Table 1. Changes in certain study parameters following percutaneous transluminal angioplasty of the arteriovenous fistula Diameter of Diameter of Area of cephalic vein collateral vein cephalic vein Area of collateral vein Blood flow Pre-PTA 39mm 36mm 0.12cm2 0.14cm2 ≤250ml/min. Post-PTA 50mm 35mm 0.24cm 0.08cm > _350 ml/min. 2 2 PTA: percutaneous transluminal angioplasty 121 letters to the editor 4. 5. 6. 7. 8. 9. 10. evaluation of arteriovenous fistulae for haemodialysis. Clin Radiol 2010;65(9): 744-9. FerringM, Henderson J, Wilmink A, Smith S. Vascular ultrasound for the pre-operative evaluation prior to arteriovenous fistula formation for haemodialysis: review of the evidence. Nephrol Dial Transplant 2008;23:1809-15. Biswas R, Patel P, Park DW, Cichonski TJ, Richards MS, Rubin JM, et al. Venous elastography: Validation of a novel highresolution ultrasound method for measuring vein compliance using finite element analysis. Semin Dial 2010;23(1):105-9. Ives CL, Akoh JA, George J, VaughanHuxley E, Lawson H. Pre-operative vessel mapping and early post-operative surveillance duplex scanning of arteriovenous fistulae. J Vasc Access 2009;10(1):37-42. Davidson I, Chan D, Dolmatch B, Hasan M, Nichols D, Saxena R, et al. Duplex ultrasound evaluation for dialysis access selection and maintenance: a practical guide. J Vasc Access 2008;9(1):1-9. Pietura R, Janczarek M, Zaluska W, Szymanska A, Janicka L, Skublewska-Bednarek A, et al. Colour Doppler ultrasound assessment of well-functioning mature arteriovenous fistulas for haemodialysis access. Eur J Radiol 2005;55(1):113-9. Martín Navarro J, Petkov V, Ríos F, Gutiérrez Sánchez MJ, Alcázar de la Ossa JM. Acceso vascular para HD: elección en mayores de 75 años. Nefrologia 2011;31 (supl 2):80. Asif A, Merrill D, Briones P, Roth D, Beathard GA. Hemodialysis Vascular Access: Percutaneous Interventions by Nephrologists. Semin Dial 2004;17(6):528-34. Juan A. Martín-Navarro, M. José Gutiérrez-Sánchez, Vladimir Petkov-Stoyanov Servicio de Nefrología. Unidad de Hemodiálisis. Hospital del Tajo. Aranjuez, Madrid. Spain. Correspondence: Vladimir Petkov Stoyanov Servicio de Nefrología. Unidad de Hemodiálisis. Hospital del Tajo. Avda. Amazonas Central s/n, 28300 Aranjuez, Madrid. Spain. [email protected] [email protected] 122 Economic impact of estimating renal function in patients with systemic lupus erythematosus Nefrologia 2012;32(1):122-3 doi:10.3265/Nefrologia.pre2011.Oct.11097 To the Editor, Kidney injury is one of the most important morbidity and mortality factors in patients with systemic lupus erythematosus (SLE).1,2 Glomerular filtration rate (GFR) is the best indicator of renal function, and it is important in the diagnosis, determining the stage, gauging treatment response and dosing medications.3 The National Kidney Foundation (NKF) recommends estimating GFR using creatinine-based equations.4,5 On the other hand, the European Consensus of Lupus Glomerulonephritis suggests that renal function in SLE patients should be measured either by serum creatinine levels or by estimating renal function using serum creatinine-based equations, but where GFR is higher than 60ml/min/1.73m2, creatinine clearance should be used (CrCl).6 In a recent publication, we reported on the high frequency of inappropriate sample collection from SLE patients when CrCl is used.7 We took a survey of Mexican rheumatologists to better understand the use of NKF-recommended equations in evaluating renal function in SLE patients. We used the google.com survey tool to send questionnaires to members of the Mexican College of Rheumatology in September 2010. We evaluated their demographic data, including sex, years practicing medicine, number of SLE patients evaluated per week and the rheumatologist’s method for evaluating renal function in patients with SLE. We received responses from 45 rheumatologists throughout the country; the mean age of those responding was 40 years, with a mean of 9.5 years practicing medicine. Of those responding, 75.6% were male and 51.2% saw more than 10 SLE patients per week. Almost half of the rheumatologists (46.7%) use CrCl in all of their patients in order to estimate GFR; 17.8% use it in two-thirds of their patients, and only 13.3% do not use it at all. Only 28.9% of those responding used equations for estimating GFR (MDRD, CKD-EPI, Cockcroft-Gault, others). According to INEGI (Mexican National Institute of Statistics and Geography), nearly 112 million people lived in Mexico in 2010. As per the Peláez-Ballestas et al study, the SLE prevalence in Mexico is 0.06%.8 We evaluated the mean cost of CrCl (serum and urinary creatinine in 24 hours) and the mean cost of measuring only serum creatinine (in order to determine GFR by means of equations) in three laboratories in central Mexico. The difference in cost between taking a single GFR measurement by one method or the other is more than 500 000 dollars if the rheumatologist uses CKD-EPI or MDRD instead of 24 hour CrCl (Table 1). Despite the evidence suggesting a high frequency of inappropriate sample collecting and the recommendation made Table 1. Costs associated with a single estimate of GFR in Mexico Equations DCr Saving $ Saving USD Costs $ 47 $ 160 Population (Mexico) 112.337.000 112.337.000 Population total LES Cost total 67.402 $ 3.167.894 67.402 $ 10.784.320 $ 7.616.426 USD 647.396 $: Mexican pesos; CKD-EPI: the Chronic Kidney Disease Epidemiology Collaboration equation; CrCl: creatinine clearance. USD: dollars. Nefrologia 2012;32(1):114-32 letters to the editor by the NKF, Mexican rheumatologists continue to use CrCl to estimate GFR. The importance of disseminating studies in other diseases, in addition to the NKF guidelines, is firstly due to the fact that the methods mentioned above do not require 24 hour urine collection and that health care systems would save thousands of dollars if this practice were generalised (worldwide); considering that a number of controlled international clinical trials use CrCl to estimate GFR.9,10 These findings show that although the guidelines suggest the use of more exact, less expensive methods, Mexican rheumatologists continue to use methods that are both more expensive and less reflective of true GFR. We must promote studies among doctors showing the benefits for patients in terms of both economic sustainability and reproducibility. If our results among Mexican rheumatologists were similar on a global level, the savings incurred by using better estimation methods could amount to millions of dollars. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla P, et al. Morbidity and mortality in systemic lupus erythematosus during a 10-year period: a comparison of early and late manifestations in a cohort of 1,000 patients. Medicine (Baltimore) 2003;82(5):299-308. 2. Stoll T, Seifert B, Isenberg DA. SLICC/ACR Damage Index is valid, and renal and pulmonary organ scores are predictors of severe outcome in patients with systemic lupus erythematosus. Br J Rheumatol 1996;35(3):248-54. 3. Soares AA, Eyff TF, Campani RB, Ritter L, Camargo JL, Silveiro SP. Glomerular filtration rate measurement and prediction equations. Clin Chem Lab Med 2009;47(9):1023-32. 4. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis 2002;39(2 Suppl 1):S1-266. 5. Levey AS, Coresh J, Balk E, Kausz AT, Levin A, Steffes MW, et al. National Kidney Foundation Nefrologia 2012;32(1):114-32 6. 7. 8. 9. 10. practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med 2003;139(2):137-47. Gordon C, Jayne D, Pusey C, Adu D, Amoura Z, Aringer M, et al. European consensus statement on the terminology used in the management of lupus glomerulonephritis. Lupus 2009;18(3):257-63. Martínez-Martínez MU, Borjas-García JA, Magaña-Aquino M, Cuevas-Orta E, Llamazares-Azuara L, Abud-Mendoza C. Renal function assessment in patients with systemic lupus erythematosus. Rheumatol Int. 2011 May 21. [Epub ahead of print]. Peláez-Ballestas I, Sanin LH, Moreno-Montoya J, Alvarez-Nemegyei J, Burgos-Vargas R, GarzaElizondo M, et al. Epidemiology of the rheumatic diseases in Mexico. A study of 5 regions based on the COPCORD methodology. J Rheumatol Suppl 2011;86:3-8. Mak A, Mok CC, Chu WP, To CH, Wong SN, Au TC. Renal damage in systemic lupus erythematosus: a comparative analysis of different age groups. Lupus 2007;16(1):28-34. Mak SK, Lo KY, Lo MW, Chan SF, Tong GM, Wong PN, et al. Efficacy of enteric-coated mycophenolate sodium in patients with active lupus nephritis. Nephrology (Carlton) 2008;13(4):331-6. Marco U. Martínez-Martínez, Carlos Abud-Mendoza Unidad Regional de Reumatología y Osteoporosis. Hospital Central Dr. Ignacio Morones Prieto. San Luis Potosí (México). Correspondence: Carlos Abud-Mendoza Unidad Regional de Reumatología y Osteoporosis. Hospital Central Dr. Ignacio Morones Prieto, Av. V. Carranza 2395, 78240 San Luis Potosí, Mexico. [email protected] [email protected] chronic kidney disease (CKD) in women than in men, regardless of age.1,2 The study by Labrador et al assesses the prevalence of occult renal disease (defined as an estimated glomerular filtration rate (eGFR) below 60ml/min and serum creatinine within the normal range), and the authors found this condition in 43.5% of the women in a group with a mean age of 77 years.3 We therefore propose studying sex as a factor involved in GFR in a cohort of elderly patients with both normal and altered serum creatinine (sCr) levels. We will also analyse the effect of this factor in patients considered as carriers of occult renal disease. Between January and April 2006, we conducted a cross-sectional study in a population with a mean age of 83 years (range: 69-97 years) that was recruited when patients came in for scheduled check-ups with the Geriatric Medicine and General Nephrology Departments at the General Hospital of Segovia. In this group, 38 patients had sCr within the normal range: Group 1, sCr ≤1.1mg/dl (range 0.7-1.1): 6 males and 32 females; 42 had altered sCr. Group 2, sCr >1.1mg/dl (range 1.2-3): 19 males and 23 females.3% of the total had diabetes mellitus, and 81.3% had arterial hypertension. GFR was estimated using the abbreviated MDRD method4 and the CockcroftGault formula.5 Table 1 shows the mean GFR given by the formulae, broken down by group and sex. doi:10.3265/Nefrologia.pre2011.Dec.11249 Out of the patient total, 56 (70%) had a GFR (MDRD) <60ml/min. Of the patients with a GFR<60ml/min according to MDRD, 18 had sCr within the normal range (100% female), while 38 had a baseline sCr>1.1mg/dl (15 males [39.5%] and 23 females [60.5%], P=.001. To the Editor, In epidemiological studies we generally find a higher prevalence of The 18 patients with a normal sCr and GFR by MDRD <60ml/min (occult renal disease) had a mean age of 81.33±6 years. Chronic kidney disease in the elderly: the impact of patients’ sex Nefrologia 2012;32(1):123-4 123 letters to the editor Table 1. Mean estimated baseline glomerular filtration rates in the study group broken down by sex Group Group Group Group 1 1 2 2 (MDRD (ml/min) (Cockcroft-Gault) (ml/min) (MDRD (ml/min) (Cockcroft-Gault) (ml/min) Male 78.01 (7) 58.78 (11) 43.60 (15) 31.86 (16) Female 60.85 (8) 45.63 (9) 37.30 (8) 29.69 (6) P 0.000 0.007 NS NS Group 1: sCr < _1.1mg/dl (6 males, 32 females). Group 2: sCr>1.1mg/dl (19 males, 23 females). NS: not significant. In our study, we also found that eGFR (estimated using the two methods listed here) in women with sCr within the normal range was significantly lower than in men. However, these differences are not as pronounced in the patient group with altered sCr (Group 2). Our data therefore confirm a higher prevalence rate of CKD in women if they are evaluated by eGFR; this statement is especially true for the subjects in Group 1. The differences in GFR between the sexes and study groups may lie within the mathematical formulae used to estimate GFR. The mathematical formulae used in our study are based on sCr, which involves the patient’s muscle mass and nutritional state. Therefore, these significant differences in GFR between the sexes among patients with a normal sCr are more likely to show women’s smaller muscle mass rather than their true GFR. It is also important to note that the MDRD formula was designed in patients with altered renal function and not validated in a healthy population: applying the MDRD formula to estimate GFR in individuals with a normal sCr may underestimate true GFR by up to 50%.6 On the other hand, when we use the MDRD formula in patients with altered renal function, the differences between males and females are less pronounced. This means that the resulting eGFR may successfully show the presence of kidney disease rather than the patient’s nutritional state and/or muscle mass. In conclusion, sex is a factor to consider when checking for chronic kidney disease in the elderly. The systematic use of formulae based on creatinine levels can lead to healthy elderly women being considered carriers of occult renal disease. 2. Chadban SJ, Briganti EM, Kerr PG, Dunstan DW, Welborn TA, Zimmet PZ, et al. Prevalence of kidney damage in Australian adults: The AusDiab kidney study. J Am Soc Nephrol 2003;14: S131-38. 3. Labrador PJ, Mengotti T, Jiménez M, Macías M, Vicente F, Labrador J, et al. Insuficiencia renal oculta en Atención Primaria. ¿Un problema exclusivo de mujeres? Nefrologia 2007;27:716-20. 4. Levey AS, Greene T, Kusek JW, Beck GJ. Simplified equation to predict glomerular filtration rate from serum creatinine. J Am Soc Nephrol 2000;11:828. 5. Cockroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron 1976;16:31-41. 6. Heras M, Fernández-Reyes MJ, Guerrero MT. Sobre la estimación de la función renal en el anciano: implicaciones del uso sistemático de la fórmula Modification of Diet in Renal Disease para el ajuste farmacológico. Rev Esp Geriatr Gerontol 2010;45(1):50-1. 1. Zhang QL, Rothenbacher D. Prevalence of chronic kidney disease in population-based studies: systematic review. BMC Public Health 2008;8:117. Manuel Heras1, Pedro García-Cosmes2, M. José Fernández-Reyes1, M. Teresa Guerrero3, Rosa Sánchez1 1 Servicio de Nefrología. Hospital General de Segovia. Spain. 2 Servicio de Nefrología. Hospital Universitario de Salamanca. Spain. 3 Servicio de Geriatría. Hospital General de Segovia. Spain. Correspondence: Manuel Heras Servicio de Nefrología. Hospital General de Segovia. 40002 Segovia. Spain. [email protected] [email protected] laparoscopic surgery for an adrenal myelolipoma associated with primary hyperaldosteronism. Myelolipomas are rare tumours; they are benign, grow slowly, and vary in size. They are made up of adipose and haematopoietic tissue. These tumours are typically non-functional and if they reach a large size, they can cause pain, pressure on adjacent organs and acute intratumoural or retroperitoneal bleeding. The patient was a male aged 54 years, obese and a smoker, with long-standing hypertension (HT) and chronic kidney disease (CKD) secondary to malignant nephroangiosclerosis that was diagnosed by kidney biopsy in 2000. He started peritoneal dialysis in 2006 and underwent a deceased donor transplant in 2008. Previous x-ray studies already showed a right adrenal mass compatible with a myelolipoma; in 2005, it measured 5*5.4cm in Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. C) BRIEF CASE REPORTS Adrenal myelolipoma associated with primary hyperaldosteronism Nefrologia 2012;32(1):124-5 doi:10.3265/Nefrologia.pre2011.Nov.11195 To the Editor, We present the case of a kidney recipient who recently underwent 124 Nefrologia 2012;32(1):114-32 letters to the editor diameter. When we were monitoring the patient’s CKD in our department, the patient also presented persistent hypokalaemia due to hyperreninemic hyperaldosteronism secondary to the underlying disease (malignant HT), which ruled out the possibility of a functional adrenal mass. However, as part of the pre-transplant study in 2008, the patient underwent a CTguided needle biopsy of the adrenal mass, and the results from the histological study suggested a myelolipoma, thus confirming the initial diagnosis. As the tumour was benign, it did not contraindicate kidney transplantation. During the two-year outpatient monitoring period following the transplant, the patient presented refractory HT requiring six different hypotensive drugs to achieve rather poor blood pressure control. His renal function deteriorated slowly over this time, and he presented proteinuria and microhaematuria. The persistent hypokalaemia reappeared and doctors ordered a new hormonal study. This time, the study found high plasma aldosterone (1098pg/ml) and suppressed plasma renin activity (0.13ng/ml/h). The patient was then diagnosed with primary hyperaldosteronism and the CT and MRI scans were repeated; the adrenal mass had reached 12*5cm in diameter along the cranio-caudal plane and 10cm in diameter along the transversal plane. It contained mainly fatty tissue with dense soft tissue areas. The patient was referred to the General Surgery Department, and in March 2010, underwent laparoscopic right adrenalectomy with excellent and prompt recovery. The histological study showed an adrenal myelolipoma with hyperplasia of the adrenal cortex (zona glomerulosa) secondary to the pressure exerted by the large size of the myelolipoma. This explained the patient’s primary hyperaldosteronism, even though the tumour was benign and non-functional. Nefrologia 2012;32(1):114-32 We could reduce the hypotensive drugs by half in the post-operative phase. The patient now has excellent control over his hypertension with the aid of two hypotensive drugs and blood potassium levels are normal, which suggests that the renal hyperplasia was not bilateral and was clearly associated with the myelolipoma. We found cases of myelolipomas associated with arterial HT in the literature, but the tumours have never been shown to be functional. Arterial HT was rather explained by renovascular causes, due to pressure exerted by the tumour, or associated with obesity or endocrine conditions such as Cushing’s syndrome or Conn’s syndrome. This case is exceptional as primary hyperaldosteronism was caused by a myelolipoma, which could possibly be explained by the pressure exerted on the adrenal gland by the large tumour. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Villar del Moral JM, Rodríguez González JM, Moreno Llorente P, Martos Martínez JM, de la Quintana Barrasate A, Expósito Rodriguez A, et al. Adrenal surgery in Spain: final results of a national survey. Cir Esp 2011;89(10):663-9. 2. Brogna A, Scalisi G, Ferrara R, Bucceri AM. Giant secreting adrenal myelolipoma in a man: a case report. J Med Case Reports 2011;5:298. 3. Wani NA, Kosar T, Rawa IA, Qayun A. Giant adrenal myelolipoma: Incidentaloma with a rare incidental association. Urol Ann 2010;2(3):130-3. 4. Lu HS, Gan MF, Chen HS, Huang SQ. Adrenal myelolipoma within myxoid cortical adenoma associated with Conn´s syndrome. J Zhejiang Univ Sci B 2008;9(6):500-5. 5. Dluhy RG, Maher MM, Wu Ch-L. A 59Year-Old Woman with an Incidentally Discovered Adrenal Nodule. N Engl J Med 2005;352:1025-32. Vanesa Camarero-Temiño, Verónica Mercado-Ortiz, Badawi Hijazi-Prieto, Pedro Abaigar-Luquin Sección de Nefrología. Complejo Hospitalario Universitario de Burgos. Spain. Correspondence: Vanesa Camarero Temiño Sección de Nefrología. Complejo Hospitalario Universitario de Burgos, Avda. del Cid 96, 09005 Burgos. Spain. [email protected] Reactive haemophagocytic syndrome associated with parvovirus B19 in a kidney-pancreas transplant patient Nefrologia 2012;32(1):125-6 doi:10.3265/Nefrologia.pre2011.Oct.11179 To the Editor, Reactive haemophagocytic syndrome or secondary haemophagocytic lymphohistiocytosis (HLH) is a disorder of the mononuclear phagocyte system characterised by generalised, ineffective and uncontrolled histiocytic proliferation that leads to cell damage and multiple organ dysfunction with haemophagocytosis. The first description of secondary forms of this disease was by Risdall et al,1 who in 1979 described a syndrome characterised by a proliferation of histiocytes with haemophagocytic activity, associated with a viral infection. This syndrome was later described in association with infections of all types and with non-infectious diseases such as rheumatoid arthritis, lupus, leukaemia, lymphomas, myelodysplastic syndromes and carcinomas. Its pathogenesis is still unclear, although there are several hypotheses. The development of this syndrome is likely to be due to an immunological disorder that results in uncontrolled T-lymphocyte activation,2 causing hypercytokinaemia, and consequently, excessive macrophage activation. 125 letters to the editor It is diagnosed according to the criteria in HLH-20043 and the treatment focuses on the infectious process, as well as on the use of gamma globulin and immunosuppression.4 We describe the case of a patient aged 42 years with a history of type 1 diabetes, diabetic nephropathy, and chronic renal failure who underwent a kidney-pancreas transplant (October 2010). The patient’s maintenance immunosuppressants are deltisone B, everolimus, and tacrolimus (FK) in addition to prophylaxis with valgancyclovir and trimethoprim/ sulfamethoxazole (TMS). The patient experienced fever, vomiting and odynophagia for 2 weeks, and was treated with oral antibiotics. Fever, asthenia and dehydration persisted, so the patient was hospitalised. Laboratory analyses revealed pancytopoenia and renal and pancreatic dysfunction; the patient received subcutaneous insulin but not haemodialysis. Blood and urine cultures were performed, as well as a PCR (polymerase chain reaction) test for cytomegalovirus (CMV), and empirical treatment with ceftriaxone and ciprofloxacin was administered. Twenty-four hours after admission, the haemodynamic state had deteriorated severely and the patient was moved to intensive care, where all immunosuppressants except for corticosteroids were discontinued. Antibiotic coverage was increased through vancomycin, imipenem, fluconazole and ganciclovir. Seven days after admission, the patient was still feverish with positive cultures for common microbes and fungi and a negative PCR for CMV. We ordered PCR for parvovirus B19 due to the persistent pancytopoenia. The physical examination showed cutaneous and mucosal pallor, asthenia, adynamia and splenomegaly. The laboratory results were as follows: Hb: 8.6mg/dl, leukocytes: 900mm3, triglycerides: 317mg/dl; ferritin >1500mcg/l. In light of suspected haemophagocytic syndrome (5 criteria met), we performed a bone marrow biopsy, which revealed histiocytes with haemophagocytosis. The patient was treated with high doses of gamma globulin (400mg/kg) during 5 days. In the end, PCR was positive for parvovirus B19. 126 All of the patient’s low values improved (haematocrit 28%; Hb 9.4g/dl; leukocytes 1900mm3; platelets 203 000mm3) and immunosuppressant treatment was resumed. Pancreatic function remained weak, and the nephrology department found the renal function to be so severely affected that the patient needed haemodialysis. A kidney biopsy puncture was performed which yielded insufficient material. At 30 days of hospitalisation, the patient was once again feverish with a headache; lumbar puncture revealed normal cerebrospinal fluid, acid-alcohol resistant bacilli (AARB) negative; PCR for CMV, herpes simplex virus, Epstein-Barr virus, cryptococcal antigenaemia all negative; adenosine deaminase at the upper cut-off level; cerebral MRI showed no lesions. The thoracic radiography showed bilateral interstitial and alveolar infiltrates, which was confirmed by thoracic CT as bilateral radiodense infiltrates; fibrobronchoscopy with bronchoalveolar lavage was performed; negative for AARB and positive for pneumocystitis carinii (PCP) when TMS treatment began. Due to the persistent fever and the lack of culture isolation in a case with pulmonary lesions, empirical treatment with isoniazid, rifampicin, ethambutol and liposomal amphotericin was administered. Another kidney biopsy puncture was performed, but graft bleeding ensued and the patient had to go to the surgical ward. Doctors decided to extirpate both grafts, and observed mesenteric adenopathies and abundant purulent matter. This matter tested AARB (+) under direct examination, and therefore antibiotic and antifungal treatments were suspended, with the patient continuing tuberculosis treatment. Final culture was positive for tuberculosis. Patient’s fever subsided and overall condition improved; he returned to his home city and is monitored by his local haemodialysis centre. Haemophagocytic syndrome that reacts to associated infections is a severe and potentially fatal condition. Immunosuppressed patients who present with a fever and haematological abnormalities (cytopoenias) should be screened for haemophagocytosis as early diagnosis enables proper treatment and a favourable prognosis. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Risdall RJ, McKenna RW, Nesbit MF, Krivit W, Balfour HH Jr, Simmons RL, et al. Virusassociated hemophagocytic syndrome: A benign histyocitic proliferation distinct from malignant histiocytosis. Cancer 1979;44:993-02. 2. Alexei A. Groma and Elizabeth D. MellinsbMacrophage activation syndrome: advances towards understanding pathogenesis. Curr Opin Rheumatol 2010;22:561-6. 3. Henter JI, Horne A, Aricó M, Egeler RM, Filipovich AH, Imashuku S, et al. HLH2004: Diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer 2007;48(2):124-31. 4. Ardalan MR, Shoja MM, Tubbs RS, Esmaili H, Keyvani H. Postrenal Transplant Hemophagocytic Lymphohistiocytosis and Thrombotic Microangiopathy American Journal of Transplantation 2008; 8: 1340–1344Associated with Parvovirus B19 Infection. Am J Transplant 2008;8:1340-4. Maico Tavera1, Jorgelina Petroni2, Luis León3, Elena Minue3, Domingo Casadei3 1 Servicio de Nefrología. Trasplante Renal. Instituto de Nefrología Sa Buenos Aires. Capital Federal, Buenos Aires (Argentina). 2 Trasplante Renal y Pancreático. Instituto de Nefrología Sa Buenos Aires. Capital Federal, Buenos Aires (Argentina). 3 Trasplante Renal. Instituto de Nefrología Sa Buenos Aires. Capital Federal, Buenos Aires (Argentina). Correspondence: Luis León Trasplante Renal. Instituto de Nefrología Sa Buenos Aires, cabello 3889, 1425 Capital Federal, Buenos Aires, Argentina. [email protected] Nefrologia 2012;32(1):114-32 letters to the editor Emphysematous cystitis resolved with medical treatment. A case report and literature review Nefrologia 2012;32(1):127 doi:10.3265/Nefrologia.pre2011.Oct.11209 To the Editor, Emphysematous cystitis is a rare infectious complication mainly seen in diabetic patients. Here, we present the first case of this disease examined by our Department. A 69 years old male patient with insulindependent diabetes and poor metabolic control was admitted due to an acute fever accompanied by nausea, vomiting and pneumaturia. The patient also had arterial hypertension, stent revascularisation for coronary heart disease, and had been diabetic for 20 years. He presented diabetic retinopathy, neuropathy and nephropathy. He had been monitored for obstructive uropathy secondary to adenoma of the prostate. He was conscious and alert during the physical examination: arterial blood pressure 110/60; heart rate 80bpm; axillary temperature 38ºC; jugular veins flat. The rest of the examination was uneventful. Laboratory testing delivered the following results: haemoglobin: 12g/dl; leukocytes: 6500; glycaemia: 738mg/dl; blood urea nitrogen: 68mg/dl; creatininaemia: 1.4mg/dl; [Na+] = 127mmol; [K+] = 5.4mmol; [Cl-] = 88mmol; [HCO3-] = 27mmol. The urine test revealed leukocyturia, red blood cells >100 per field and abundant bacteria. The urine culture showed extended-spectrum beta lactamase-producing Escherichia coli. The CT urography (a specific CT technique) showed air within the bladder lumen near its ventral face. The patient received hydration, insulin therapy and treatment with ertapenem. Nefrologia 2012;32(1):114-32 Figure 1. CT intravesical air Urography showing Progress was excellent, with no septic shock and easy-to-manage hyperosmolar syndrome. Emphysematous cystitis was discovered by Eisenlohr1 at the end of the 19th century and Bailey2 described this illness. It may present asymptomatically until the onset of severe sepsis, or it may pass through stages with pneumaturia and acute abdomen. For the case in question, the key symptom was pneumaturia and the presence of air in the bladder as shown by the CT (Figure 1). In the series described by Thomas et al.3, the most common bacterial strain was E. coli, followed by Klebsiella. The mean age was 66 years, most affected individuals were women (64%) and most were diabetic (67%). The patient treated in our Department was similar in age to those in the other study, and also suffered diabetes with poor metabolic control. It has been postulated that air is produced by the fermentation of glucose in urine.4 Treatment is usually medical, as in our case, and the mortality rate is close to 7%. In isolated cases, treatment has been combined with surgery or a hyperbaric chamber. 2. Bailey H. Cystitis emphysematosa; 19 cases with intraluminal and interstitial collections of gas. Am J Roentgenol Radium Ther Nucl Med 1961;86:850-62. 3. Thomas AA, Lane BR, Thomas AZ, Remer EM, Campbell SC, Shoskes DA. Emphysematous cystitis: a review of 135 cases. BJU Int 2007;100(1):17-20. 4. González-Martín V, Rodrigo E, Arias M, Lastra P. Cistitis enfisematosa en un trasplantado renal. Nefrologia 2008;28(1):110-1. Héctor Parra-Riffo, Juan Lemus-Peñaloza, Paula Maira Unidad de Nefrología. Hospital FACH. Santiago de Chile (Chile). Correspondence: Héctor Parra Riffo Unidad de Nefrología, Hospital FACH, Av. Las Condes 8631, Santiago de Chile, 7560171 Santiago de Chile, Chile. [email protected] Rhabdomyolysis and acute renal failure following hard physical activity in a patient treated with rosuvastatin Nefrologia 2012;32(1):127-8 doi:10.3265/Nefrologia.pre2011.Oct.11118 To the Editor, Muscular problems constitute one of the most important adverse effects of statin drugs, and they range from myalgias to myositis/rhabdomyolysis.1,2 The incidence rates of myalgia and rhabdomyolysis due to statins are 5-10% and 0.01% respectively.1 However, this is the drug type that is most frequently involved in rhabdomyolysis.3 The authors declare they have no potential conflicts of interest related to the contents of this article. Risk factors for developing rhabdomyolysis due to statins include, but are not limited to, high doses, intense physical exercise and interactions with other drugs.2,4 1. Eisenlohr W. Das Interstielle vagina, darm und harnblasenemphysem zurückgefürt auf gasentwickelnde bakterien. Beitr Path Anat Allg Path 1888;3:101. We describe a patient treated with rosuvastatin who developed rhabdomyolysis and acute renal failure following intense physical activity. Conflicts of interest 127 letters to the editor Male patient diagnosed with dyslipidaemia and arterial hypertension 3 months prior to the event. He began treatment with rosuvastatin (10mg/day), olmesartan and torasemide; creatinine level was 1.1mg/dl. One week before being admitted, he was hired to install antennas, which required considerable physical effort. About three days before admission, he experienced muscle soreness in the lower limbs and took 2 ibuprofen 600mg tablets. The pain did not subside and he experienced nausea and vomiting, so he came to the hospital. Physical examination findings were normal. Diagnostic tests provided the following results; haemoglobin 14.6g/dl; urea 141 mg/dl; creatinine 9.33mg/dl; uric acid 12.2mg/dl; total calcium 7.7mg/dl; phosphate 4mg/dl; sodium 137mEq/l; potassium 6mEq/l; chlorine 103mEq/l; bicarbonate 24mEq/l; albumin 4g/dl; creatine kinase (CK) 6243U/l (nv: 38-174); lactate-dehydrogenase 768U/l (nv: 140300); triglycerides 253mg/dl; CRP 42.5mg/l; coagulation, platelets, and other basic biochemical parameters were within normal values. Normal thyrotropin. Urine: d 1030, proteinuria 30-70mg/dl; sediment 80/90 leukocytes/field, 5-10 red blood cells/field; sodium 59mEq/l; urine culture negative. Serology for hepatitis B, C and HIV was negative. Thoracic radiography: normal; electrocardiogram: sinus rhythm, right bundle branch block. Ultrasound showed normal kidneys. Medication was suspended and we prescribed hydration/electrolyte replacement. The patient maintained good urine production and creatinine levels and other biochemical parameters improved. After 11 days, the patient was discharged with a creatinine level of 1.29mg/dl and normal CK; the decision was made later to prescribe atorvastatin 20mg every other day. Rosuvastatin is a synthetic HMG-CoA reductase inhibitor with pharmacological characteristics that, in theory, imply a lower risk of myotoxicity: 1) it has a 128 low liposolubility, which makes it less able to penetrate muscle tissue;1 2) it is potent, and generally speaking, low doses of potent statins are less myotoxic than high doses of less potent drugs;1 and 3) it is eliminated through biliary (90%) and renal (10%) excretion. Metabolism by cytochrome P450 is minimal, and performed by the CYP2C9 and CYP2C19 subfamilies and not by CYP3A4. This means that there is less possibility of it interacting with other drugs.1,5 However, cases of rhabdomyolysis and kidney failure associated with rosuvastatin have been reported.6,7 Renal failure appears in approximately 40% of cases of rhabdomyolysis due to statins, which gives a poorer prognosis.3 In the case of this patient, the renal failure, which could also have been affected by the ibuprofen and the olmesartan, resolved favourably. The patient had been taking rosuvastatin with no incidents until he engaged in hard physical labour. Statin-related rhabdomyolysis is related to: 1) depletion of intermediary metabolites in the pathway of mevalonate, such as geranylgeranyl pyrophosphate and farnesyl pyrophosphate, causing myocyte apoptosis and 2) mitochondrial dysfunction.8 Decreases in Q10 coenzyme and changes in membrane stability due to lower cholesterol levels may also be contributing factors.2 Hard physical activity has an additional myotoxic effect by increasing oxygen consumption and provoking mitochondrial overload.8 The course of action to take with patients who develop statin-related rhabdomyolysis and need hypolipidaemic agents afterwards is a matter of debate. Some recommend atorvastatin every other day, or fluvastatin or rosuvastatin on alternate days or administered once weekly.1,2 To conclude, patients receiving statin drugs must be aware that hard physical labour, whether exercise or occupational, may trigger muscular complications. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Jacobson TA. Toward «pain-free» statin prescribing: clinical algorithm for diagnosis and management of myalgia. Mayo Clin Proc 2008;83:687-700. 2. Joy TR, Hegele RA. Narrative review: statin-related myopathy. Ann Intern Med 2009;150:858-68. 3. Oshima Y. Characteristics of drugassociated rhabdomyolysis: analysis of 8,610 cases reported to the U.S. Food and Drug Administration. Intern Med 2011;50:845-53. 4. Sirvent AE, Cabezuelo JB, Enríquez R, Amorós F, González C, Reyes A. [Rhabdomyolysis and anuric kidney failure induced by the treatment with a gemfibrozil-cerivastatin combination]. Nefrologia 2001;21:497-500. 5. Calza L. Long-term use of rosuvastatin: a critical risk benefit appraisal and comparison with other antihyperlipidemics. Drug Healthc Patient Saf 2009;1:25-33. 6. Buyukhatipoglu H, Sezen Y, Guntekin U, Kirhan I, Dag OF. Acute renal failure with the combined use of rosuvastatin and fenofibrate. Ren Fail 2010;32:633-5. 7. Pérez Díaz I, Sánchez Argaiz M, Sánchez Gómez E. Possible rosuvastatin-induced fatal rhabdomyolysis. Farm Hosp 2011;35:340-1. 8. Meador BM, Huey KA. Statin-associated myopathy and its exacerbation with exercise. Muscle Nerve 2010;42:469-79. Diana Martínez-López1, Ricardo Enríquez2, Ana E. Sirvent2, M. Dolores Redondo-Pachón2, Isabel Millán2, Francisco Amorós2 1 Servicio de Medicina Intensiva. Hospital General de Elche. Elche, Alicante. Spain. 2 Servicio de Nefrología. Hospital General de Elche. Elche, Alicante. Spain. Correspondence: Ricardo Enríquez Servicio de Nefrología. Hospital General de Elche. Camino de la Almazara, 03203 Elche, Alicante. Spain. [email protected] Nefrologia 2012;32(1):114-32 letters to the editor Intestinal pseudoobstruction secondary to persistent constipation due to lanthanum carbonate Nefrologia 2012;32(1):129 doi:10.3265/Nefrologia.pre2011.Nov.11191 To the Editor, Hyperphosphataemia is a complication of chronic kidney disease (CKD) which is often accompanied by hypocalcaemia and low serum vitamin D levels. Without treatment, these deficiencies generally lead to severe secondary hyperparathyroidism. Restricting dietary phosphate is key to treating this condition, but this approach alone is not sufficient to control hyperphosphataemia. As a result, oral phosphate binders are used by more than 90% of patients with CKD.1 Lanthanum carbonate is a non-calcium, metal-containing phosphate binder used in hyperphosphataemia treatment. This trivalent cation forms ionic bonds with phosphate. Precipitation of insoluble complexes then takes place in the intestinal lumen. Due to the very low degree of absorption by the gastrointestinal tract (<0.0013%), an abdominal radiography will show radiopaque images of lanthanum carbonate, which may slow intestinal transit as a side effect.2 Recent studies have shown that this may cause or worsen diverticular colitis, which would increase serum lanthanum levels. In conclusion, caution must be exercised when using this drug in patients with diverticula.3 We present the clinical case of a 55 year old patient with CKD treated with periodic haemodialysis, secondary hyperparathyroidism and severe hyperphosphataemia, mainly due to poor dietary compliance. Therefore, we prescribed a non-calcium phosphate binder, lanthanum carbonate (Fosrenol®), since the patient also presented significant peripheral vascular disease of ischaemic Nefrologia 2012;32(1):114-32 origin and the aim was to reduce the risk of vascular calcifications. The patient came to the Emergency Department on two occasions due to abdominal pain and constipation, and was admitted to the General Surgery Department the second time for suspected diverticulitis. The abdominal CT revealed uncomplicated sigmoid diverticulitis. One month after being discharged from the hospital, the patient returned with abdominal pain, constipation, nausea and vomiting, and was then admitted by our Nephrology Department. Examination showed peristaltic activity and diffuse abdominal pain. Laboratory test results were uneventful (absence of leukocytosis; amylase and lipase within normal ranges). In both the preliminary and current radiographies, as shown in Figure 1, we can observe abundant deposits of lanthanum carbonate in the colon and dilated intestinal loops consistent with the ingestion of lanthanum carbonate 3g/daily for 3 months. The diagnosis was intestinal pseudo-obstruction secondary to persistent constipation due to lanthanum carbonate. That drug was suspended; with administration of two cleansing enemas and the addition of an oral osmotic laxative (Duphalac®) the clinical symptoms resolved completely. Plain film abdominal radiography showing abundant radiopaque lanthanum carbonate remains in the colon and dilated intestinal loops. Figure 1. Intestinal pseudo-obstruction due to lanthanum carbonate Hyperphosphataemia is closely related to developing a risk of cardiovascular disease with tissue ischaemia and calciphylaxis in CKD, and with an increase in fibroblast growth factor 23 and secondary hyperparathyroidism. However, when faced with preventing such complications, we must be mindful of the fact that treating hyperphosphataemia with lanthanum carbonate presents a risk of developing abdominal symptoms such as those described here. We recommend closely monitoring tolerance-based doses in order to prevent related complications.4 Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Tonelli M, Pannu N, Manns B. Oral phosphate binders in patients with kidney failure. N Engl J Med 2010; 362:1312-24. 2. Turkmen K, Solak Y, Anil M, Polat H, Tonbul HZ. An unusual hurdle to renal transplantation: speckled abdominal opacities induced by lanthanum carbonate. Intern Med J 2010;40(12): e1-2. 3. Kato A, Takita T, Furuhashi M. Accumulation of lanthanum carbonate in the digestive tracts. Clin Exp Nephrol 2010;14(1):100-1. 4. Cronin RE, Quarles LD, Berns JS, Sheridan AM. Treatment of hyperphosphatemia in chronic kidney disease. Last literature review version 19.1: January 2011 | This topic last updated: February 9, 2011. [Available at: www.uptodate.com]. Vanesa Camarero-Temiño, Verónica Mercado-Valdivia, Badawi Hijazi-Prieto, Pedro Abaigar-Luquin Sección de Nefrología. Complejo Hospitalario Universitario de Burgos. Spain. Correspondence: Vanesa Camarero Temiño Sección de Nefrología. Complejo Hospitalario Universitario de Burgos. Avda del Cid, 96, 09005 Burgos. Spain. [email protected] [email protected] 129 letters to the editor Acute renal failure due to gabapentin. A case report and literature review Nefrologia 2012;32(1):130-1 doi:10.3265/Nefrologia.pre2011.Nov.11087 To the Editor, Gabapentin is an anticonvulsive that is widely used for a number of indications at present: diabetic neuropathy, neuropathic pain of other causes, epilepsy, etc. Some of its most common side effects include the following: ataxia, nystagmus, drowsiness, headaches, diplopia, fatigue and myoclonic twitches.1 All of these effects appear quite often in patients with chronic kidney disease, especially if they are undergoing dialysis and their doses are not adjusted to their glomerular filtration rates.2 We describe a new case of rhabdomyolysis and acute renal failure due to gabapentin in order to raise awareness of the importance of monitoring creatine kinase (Ck) and renal function, and of being on the alert for side effects every time this drug is used.1,3 The patient, aged 49 years, was taken to the Emergency Department due to delirium, deteriorating condition and myalgias evolving over 48 hours. The patient had visited the Emergency Department two days before due to lumbosacral pain, was diagnosed with mechanical low back pain, and began treatment with 600mg gabapentin every 8 hours. Relevant medical history included smoking 1 packet/day, active use of multiple substances (alcohol, heroin, cocaine, etc.) and a recent hospital admission. Arterial hypertension treated with eprosartan and bisoprolol. Anxiety-depression syndrome. No relevant nephrological or urological history. Ten days prior to being admitted, he underwent laboratory testing at the clinic. Tests showed normal renal function (creatinine 0.9mg/dl, urea 30mg/dl and no pathological findings in urinary sediment. His normal treatment consisted of paroxetine, mianserin, disulfiram, eprosartan, bisoprolol, and, during the last 48 hours, gabapentin. 130 The physical examination showed acceptable general condition, no fever, and low blood pressure (90/60mmHg). The patient was conscious, disoriented and drowsy, with myoclonic twitches. Eupnoea at rest. Normal breath sounds; no oedema in lower limbs, with mucous membrane dehydration. The neurological examination found no focal dystonia or neck stiffness, and the significant finding was that the patient trembled when at rest. Due to the patient history mentioned above, we screened urine for toxins, and it was positive for cocaine, heroin and morphine. We also ran a full blood and urine analysis which provided the following relevant results: - Blood: glucose: 146mg/dl; GOT: 246IU/l; GPT: 231IU/l; bilirubin: 0.8mg/dl; LDH: 2520U/l; C-reactive protein: 258; Ck: 14911U/l; creatinine: 13.5mg/dl; urea: 273mg/dl; Na: 136mmol/l; Ki: 6.8mmol/l; Ca: 6.1mg/dl; Pi: 16mg/dl; pH 7.2; bicarbonate: 12mmol/l. - Urine: specific gravity 1020; pH 5; proteins 30mg/dl; glucose negative; ketone bodies: present; leukocytes 70; erythrocytes 200/µl (after catheterisation). Given these findings and oligoanuria, doctors requested a kidney ultrasound that showed kidneys of normal size, shape and ecogenicity and no ureteral dilation. Medical treatment for hyperkalaemia and metabolic acidosis was initiated as well as plasma volume expansion. As oliguria, severe metabolic acidosis and delirium persisted with only minimal improvements after administration of 0.5mg flumazenil, we decided to place a femoral catheter and perform an emergency dialysis session. In order to avoid imbalance syndrome, we used a low cut-off dialyser with a flow of 200ml/min during 2 hours 30 minutes and neutral-pH Balance solution. Under this treatment, the patient improved partially from a clinical standpoint; diuresis resumed at 60ml/hour and the metabolic acidosis resolved. As the patient had a history of multiple drug use in addition to the delirium and the abnormal laboratory results described here, we performed a differential diagnosis to rule out other causes of delirium, such as Wernicke encephalopathy, neuroleptic malignant syndrome and sepsis. The biochemical study was expanded to measure thyroid hormones, vitamin B12 and folic acid; results were normal. Serological analyses for hepatitis B and C and HIV were negative. Cultures from blood and urine samples were negative. A cerebral MRI found non-specific demyelinating lesions in pale nuclei and the pyramidal tract that were not compatible with Wernicke-Korsakoff syndrome. Neuroleptic malignant syndrome was effectively ruled out by the absence of high fever and rigidity, in addition to the clinical response following the first dialysis session. We gathered information from family members, who confirmed that the patient had ingested at least 6 gabapentin tablets in the 24 hours prior to admission, along with the drugs cited above. After discontinuing gabapentin and providing hydration and an additional dialysis session in the following 12 hours, the patient’s encephalopathy improved progressively. The electrocardiogram taken at 72 hours showed no pathological findings, renal function became normal (creatinine: 1mg/dl; urea: 55mg/dl in 48 hours and 0.9mg/dl in 36 hours) and Ck values decreased progressively (7327 at 48 hours and 555 at 96 hours). Gabapentin toxicity and side effects are well-known among nephrologists and fully described in the literature as myoclonic twitches, myopathy, neurotoxicity, etc., particularly in dialysis patients.2,4 Rhabdomyolysis with associated acute renal failure is an uncommon side effect, but it has been described in earlier cases.1,3 The aetiology of rhabdomyolysis varies greatly. Its most frequent causes include trauma, intense physical exercise, infections, and drugs such as statins, fibrates, neuroleptics, colchicine and proton pump inhibitors.5,6 It is also Nefrologia 2012;32(1):114-32 letters to the editor associated with cocaine use, but unlike the case described here, rhabdomyolysis tends to be associated with hypertension and malignant nephrosclerosis. While our patient did use cocaine, this is unlikely to be the root of the problem7 given that the patient was originally hypotensive and experienced early renal function recovery. While gabapentin levels were not measured, the rapid resolution of the delirium and recovery of renal function after only two sessions of low cut-off haemodialysis seem to indicate that gabapentin caused the symptoms. In fact, gabapentin is eliminated through renal excretion only, and since it does not bind to proteins, a single dialysis session will eliminate nearly 35% of the total.8,9 In our case, this would explain the rapid improvement in symptoms. As in the other 2 cases of gabapentin-induced acute renal failure and rhabdomyolysis, the patients involved had multiple illnesses and were affected by multiple medications or other factors that might lead to rhabdomyolysis and renal failure. Another similarity was the rapid resolution of the condition and the improvement in Ck values after discontinuing the drug. In summary, we can conclude that although it happens infrequently, gabapentin may cause myotoxicity, rhabdomyolysis and renal failure even in patients whose renal function was previously normal. This is why we must take special care with its dosage, with concomitant medications and the patient’s co-morbidities, and why, after prescribing gabapentin, we must be watchful for any signs of muscle toxicity or kidney failure and quickly discontinue the drug if necessary. Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Bilgir O, Calan M, Bilgir F, Kebapçilar L, Yüksel A, Yildiz Y, et al. GabapentinNefrologia 2012;32(1):114-32 2. 3. 4. 5. 6. 7. 8. 9. 10. induced rhabdomyolysis in a patient with diabetic neuropathy. Intern Med 2009;48(12):1085-7. Bassilios N, Launay-Vacher V, Khoury N, Rondeau E, Deray G, Sraer JD. Gabapentin neurotoxicity in a chronic haemodialysis patient. Nephrol Dial Transplant 2001;16(10):2112-3. Tuccori M, Lombardo G, Lapi F, Vannacci A, Blandizzi C, Del Tacca M. Gabapentininduced severe myopathy. Ann Pharmacother 2007;41(7):1301-5. Lipson J, Lavoie S, Zimmerman D. Gabapentin-induced myopathyin 2 patients on short daily hemodialysis. Am J Kidney Dis 2005;45(6):e100-4. Guis S, Mattei JP, Cozzone PJ, Bendahan D. Pathophysiology and clinical presentations of rhabdomyolysis. Joint Bone Spine 2005;72: 382-91. Marinella MA. Rhabdomyolysis associated with haloperidol withoutevidence of NMS. Ann Pharmacother 1997;31:927-8. Horowitz BZ, Panacek EA, Jouriles NJ. Severe rhabdomyolysis with renal failure after intranasal cocaine use. J Emerg Med 1997;15(6):833-7. Bluma RA, Pharm D, Thomas J, Schultz RW, Keller E, Reetze P, et al. Pharmacocinetics of gabapentin in subjects with various degrees of renal function. Clin Pharmacol Ther 1994;56:154-9. Wong MO, Eldon MA, Keane WF, Türck D, Bockbrader HN, Underwood BA, et al. Disposition of gabapentin in anuric subjects on hemodialysis. J Clin Pharmacol 1995;35(6):622-6. Naranjo CA, Busto U, Sellers EM, Sandor P, Ruiz I, Roberts EA, et al. A method for estimatingthe probability of adverse drug reactions. Clin Pharmacol Ther 1981;30:239-45. Eduardo Torregrosa-de Juan, Pau Olagüe-Díaz, Pilar Royo-Maicas, Enrique Fernández-Nájera, Rafael García-Maset Sección de Nefrología. Hospital de Manises. Manises, Valencia. Spain. Correspondence: Eduardo Torregrosa de Juan Sección de Nefrología. Hospital de Manises, Av. Generalitat Valenciana, 50, 46940 Manises. Valencia. Spain. [email protected] [email protected] Haemorrhagic fever with renal failure syndrome: a case report Nefrologia 2012;32(1):131-2 doi:10.3265/Nefrologia.pre2011.Dec.11225 To the Editor, Haemorrhagic fever with renal syndrome (HFRS) is a clinical condition secondary to infection with a hantavirus (Hantaan, Seoul, No Name, Andes virus, Puumala and Dobrava); the latter two varieties are endemic in rural areas of Eastern Europe, and of the two, infection with the Puumula virus has a better long-term prognosis.1 Rodents are the natural carriers of hantavirus, which is transmitted to humans when they come into direct contact with rodent secretions (urine, faeces and saliva). The natural evolution of the disease entails 4 successive phases following an incubation period of about 3 weeks. The first phase is characterised by fever, followed by a phase with shock and oliguria; patients who survive this phase enter a phase with polyuria, which in turn is followed by a convalescence period of variable duration. Thrombocytopoenia is common and may produce haemorrhages at any location. Renal symptoms include proteinuria, haematuria and decreased glomerular filtration rate. Direct vascular endothelial lesions and tubulo-interstitial nephritis mediated by cytokines have been proposed as the underlying physiopathological cause.2 Diagnosis is based on a strong clinical suspicion, and confirmed by specific serological methods.3 Kidney biopsy is not necessary.4 A renal ultrasound can show the increase in kidney size and in resistance indices. Perirenal fluid collection is also a common finding (in addition to pleural or pericardial effusion or ascites). No vaccine or specific treatment exists; supportive therapy is of vital importance. One double-blind study showed decreased mortality given early treatment with ribavirin.5 131 letters to the editor Below, we describe our hospital’s experience with a case of HFRS secondary to infection with the Puumala virus. morphologically normal kidneys with no ureteral dilation, with perirenal and pelvic free liquid. (Figure 1) The patient was a male aged 18 years with no relevant prior history who came to the Emergency Department due to fever, muscle aches and a frontal headache evolving over 3 days with no improvement following his doctor’s prescription of amoxicillin-clavulanic acid and paracetamol. Two weeks before, he had been hiking in a rural area of Slovenia. The patient was admitted for observation, which is why he was initially given empirical antibiotic treatment with ceftriaxone and levofloxacin after the samples were extracted. The day after admission, he suffered a conjunctival haemorrhage (Figure 2), tendency toward oliguria, the appearance of oedemas, and decreased renal function; his creatinine level was 2.7mg/dl. As a hantavirus infection was suspected, we opted for antiviral treatment with ribavirin (500mg/i.v. every 8 hours). The same day, 8 hours later, he suffered an episode of dyspnoea and tachypnoea with desaturation (89%) and hypoxia (pO2: 63), and the decision was made to send him to the Intensive Care Unit. On the fourth day after admission, the patient presented epistaxis that subsided with anterior nasal packing. The oliguria became more pronounced, oedemas increased and the renal function worsened with creatinine levels of 5.6mg/dl. We then decided to start haemodialysis by means of a temporary catheter in the right femoral vein. After 4 session of haemodialysis, we observed improvements in urinary volume and renal function. The immunological study (antinuclear antibodies, anti-neutrophil cytoplasmic antobodies, anti-DNA, anti-GBM antibodies, complementary components, immunoglobulins, protein electrophoresis, cryoglobulin, circulating immune complexes) and serology study for hepatitis B and C, HIV, Epstein-Barr virus, cytomegalovirus, leptospira, parvovirus B19 and toxoplasms were negative or normal. On day 7, the laboratory reported IgG (+) 1/512 for Puu- The physical examination found a fever of 39.2ºC, normal blood pressure, no adenopathy, and diffuse pain upon abdominal palpation. The Emergency Department laboratory blood test found 10 010 leukocytes/mm3 with no eosinophilia; thrombocytopoenia of 32 000/mm3 with no platelet additives; haemoglobin: 18; haematocrit: 48%; creatinine: 1.35mg/dl; urea: 47mg/dl; lactate dehydrogenase (LDH): 324U/l. All other blood tests were normal. Microhaematuria was present. Thoracic and abdominal radiographies were normal. The abdominal ultrasound and the abdominal and pelvic CT showed Figure 1. Abdominal and pelvic computed tomography 132 Figure 2. Conjunctival haemorrhage appearing the day after patient was admitted mala virus, and antibiotic treatment was consequently discontinued. On day 13, the patient was discharged after completing the antiviral treatment, showing recovered renal function and a creatinine level of 1.25. At follow-up 5 weeks later, his renal function was completely normal (creatinine: 0.69, estimated glomerular filtration rate >60). Conflicts of interest The authors declare they have no potential conflicts of interest related to the contents of this article. 1. Miettinen MH, Mäkelä SM, Ala-Houhala IO, Huhtala HS, Kööbi T, Vaheri AI, et al. Tenyear prognosis of Puumala hantavirusinduced acute intersticial nephritis. Kidney int 2006;69:2043-8. 2. Mäkelä S, Mustonen J, Ala-Houhala I, Hurme M, Koivisto AM, Vaheri A, et al. Urinary excretion of inerleukin-6 correlates whith proteinuria in acute Puumala hantavirus - induced nephritis. Am J Kidney Dis 2004;43:809-16. 3. Vapalahti O, Mustonen J, Lundkvist A, Henttonen H, Plyusnin A, Vaheri A. Hantavirus infections in Europe. Lancet Infect Dis 2003;3:653-61. 4. Kim S, Sung SH, An HR, Jun YH, Yu M, Ryu DR, et al. A case report of crescentic glomerulonephritis associated with hantavirus infection. Nephrol Dial Transplant 2010;25(8):2790-2. 5. Huggins JW, Hsiang CM, Cosgrriff M, Guang MY, Smith JI, Wu ZO, et al. Prospective, double-blind, concurrent, placebo-controlled clinic trial of intravenous ribavirin therapy of hemorrhagic fever with renal syndrome. J Infect Dis 1991;164:1119-27. Rodrigo Avellaneda-Campos Unidad de Nefrología. Hospital Universitario Donostia. San Sebastián, Gipuzkoa. Spain. Correspondence: Rodrigo Avellaneda Campos Unidad de Nefrología. Hospital Universitario Donostia. San Sebastián, Gipuzkoa. Spain. [email protected] Nefrologia 2012;32(1):114-32