vol.49 no.4 december 2015 Prva in edina samostojna kemoterapija, ki v primerjavi z ostalimi možnostmi zdravljenja z enim zdravilom, pri bolnicah s predhodno že večkratno zdravljenim metastatskim rakom dojke, dokazano značilno podaljša celokupno preživetje.1,2 NOVA SMER DO PODALJŠANJA CELOKUPNEGA PREŽIVETJA Halaven (eribulin): ne-taksanski zaviralec dinamike mikrotubulov, prvo zdravilo iz nove skupine kemoterapevtikov, imenovanih halihondrini. Zdravilo HALAVEN je indicirano za zdravljenje bolnic z lokalno napredovalim ali metastatskim rakom dojke, ki je napredoval po vsaj enem režimu kemoterapije za napredovalo bolezen. Predhodna zdravljenja morajo vključevati antraciklin in taksan, bodisi kot adjuvantno zdravljenje ali za zdravljenje metastatskega raka dojke, razen če to zdravljenje za bolnice ni bilo primerno.1 Priporočeni odmerek 1,23 mg/m2, intravensko, v obliki 2-do 5-minutne infuzije, 1. in 8. dan vsakega 21-dnevnega cikla. Ena 2 ml viala vsebuje 0,88 mg eribulina. Raztopina, pripravljena za uporabo, redčenje ni potrebno. SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA HALAVEN 0,44 mg/ml raztopina za injiciranje (eribulin) TERAPEV TSKE INDIKACIJE: Zdravljenje lokalno napredovalega ali metastatskega raka dojke, ki je napredoval po vsaj enem režimu kemoterapije za napredovalo bolezen vključno z antraciklinom in taksanom (adjuvantno zdravljenje ali zdravljenje metastatskega raka dojke), razen če to ni bilo primerno. ODMERJANJE IN NAČIN UPORABE: Halaven se daje v enotah, specializiranih za dajanje citotoksične kemoterapije, in le pod nadzorom usposobljenega zdravnika z izkušnjami v uporabi citotoksičnih zdravil. Odmerjanje: Priporočeni odmerek eribulina v obliki raztopine je 1,23 mg/m2 i.v. v obliki 2- do 5-minutne infuzije 1. in 8. dan vsakega 21-dnevnega cikla. Bolnikom je lahko slabo ali bruhajo. Treba je razmisliti o antiemetični profilaksi, vključno s kortikosteroidi. Preložitev odmerka med zdravljenjem: Dajanje Halavena je treba preložiti, če se pojavi kaj od naslednjega: absolutno število nevtrofilcev (ANC) < 1 x 109/l, trombociti < 75 x 109/l ali nehematološki neželeni učinki 3. ali 4. stopnje. Zmanjšanje odmerka med zdravljenjem: Za priporočila za zmanjšanje odmerka ob pojavu hematoloških ali nehematoloških neželenih učinkov glejte celoten povzetek glavnih značilnosti zdravila. Okvara jeter zaradi zasevkov: Priporočeni odmerek pri blagi okvari jeter (stopnje A po Child-Pughu) je 0,97 mg/m2 v obliki 2- do 5-minutne i.v. infuzije 1. in 8. dan 21-dnevnega cikla. Priporočeni odmerek pri zmerni okvari jeter (stopnje B po Child-Pughu) je 0,62 mg/m2 v obliki 2- do 5-minutne i.v. infuzije 1. in 8. dan 21-dnevnega cikla. Pri hudi okvari jeter (stopnje C po Child-Pughu) se pričakuje, da je treba dati še manjši odmerek eribulina. Okvara jeter zaradi ciroze: Zgornje odmerke se lahko uporabi za blago do zmerno okvaro, vendar se priporoča skrbno nadziranje, saj bo odmerke morda treba ponovno prilagoditi. Okvara ledvic: Pri hudi okvari ledvic (očistek kreatinina < 40 ml/min) bo morda treba odmerek zmanjšati. Priporoča se skrbno nadziranje varnosti. Način uporabe: Odmerek se lahko razredči z do 100 ml 0,9 % raztopine natrijevega klorida (9 mg/ml) za injiciranje. Ne sme se ga redčiti v 5 % infuzijski raztopini glukoze. Pred dajanjem glejte navodila glede redčenja zdravila v celotnem povzetku glavnih značilnosti zdravila ter se prepričajte, da obstaja dober periferni venski dostop ali prehodna centralna linija. Ni znakov, da bi eribulin povzročal mehurje ali dražil. V primeru ekstravazacije mora biti zdravljenje simptomatsko. KONTRAINDIKACIJE: Preobčutljivost na zdravilno učinkovino ali katerokoli pomožno snov. Dojenje. POSEBNA OPOZORILA IN PREVIDNOSTNI UKREPI: Mielosupresija je odvisna od odmerka in se kaže kot nevtropenija. Pred vsakim odmerkom eribulina je treba opraviti pregled celotne krvne slike. Zdravljenje z eribulinom se lahko uvede le pri bolnikih z vrednostmi ANC . 1,5 x 109/l in s trombociti > 100 x 109/l. Bolnike, pri katerih se pojavijo febrilna nevtropenija, huda nevtropenija ali trombocitopenija, je treba zdraviti v skladu s priporočili v celotnem povzetku glavnih značilnosti zdravila. Hudo nevtropenijo se lahko zdravi z uporabo G-CSF ali enakovrednim zdravilom v skladu s smernicami. Bolnike je treba skrbno nadzirati za znake periferne motorične in senzorične nevropatije. Pri razvoju hude periferne nevrotoksičnosti je treba odmerek prestaviti ali zmanjšati. Če začnemo zdravljenje pri bolnikih s kongestivnim srčnim popuščanjem, z bradiaritmijami ali sočasno z zdravili, za katera je znano, da podaljšujejo interval QT, vključno z antiaritmiki razreda Ia in III, in z elektrolitskimi motnjami, je priporočljivo spremljanje EKG. Pred začetkom zdravljenja s Halavenom je treba popraviti hipokaliemijo in hipomagneziemijo in te elektrolite je treba občasno kontrolirati med zdravljenjem. Eribulina ne smemo dajati bolnikom s prirojenim sindromom dolgega intervala QT. To zdravilo vsebuje majhne količine etanola (alkohola), manj kot 100 mg na odmerek. Eribulin je pri podganah embriotoksičen, fetotoksičen in teratogen. Halavena se ne sme uporabljati med nosečnostjo, razen kadar je to nujno potrebno. Ženske v rodni dobi naj ne zanosijo v času, ko same ali njihov moški partner dobivajo Halaven, in naj med zdravljenjem in še do 3 mesece po njem uporabljajo učinkovito kontracepcijo. Moški naj se pred zdravljenjem posvetujejo o shranjevanju sperme zaradi možnosti nepopravljive neplodnosti. INTERAKCIJE: Eribulin se izloča do 70 % prek žolča. Sočasna uporaba učinkovin, ki zavirajo jetrne transportne beljakovine, kot so beljakovine za prenos organskih anionov in beljakovine, odporne na številna zdravila, z eribulinom se ne priporoča (npr. ciklosporin, ritonavir, sakvinavir, lopinavir in nekateri drugi zaviralci proteaze, efavirenz, emtricitabin, verapamil, klaritromicin, kinin, kinidin, dizopiramid itd). Sočasno zdravljenje z indukcijskimi učinkovinami, kot so rifampicin, karbamazepin, fenitoin, šentjanževka, lahko povzroči znižanje koncentracij eribulina v plazmi, zato je ob sočasni uporabi induktorjev potrebna previdnost. Eribulin je blag inhibitor encima CYP3A4. Priporočljiva je previdnost in spremljanje glede neželenih učinkov pri sočasni uporabi snovi, ki imajo ozko terapevtsko okno in se odstranjujejo iz telesa predvsem preko CYP3A4 (npr. alfentanil, ciklosporin, ergotamin, fentanil, pimozid, kinidin, sirolimus, takrolimus). NEŽELENI UČINKI: Povzetek varnostnega profila Neželeni učinek, o katerem najpogosteje poročajo v zvezi s Halavenom, je supresija kostnega mozga, ki se kaže kot nevtropenija, levkopenija, anemija, trombocitopenija s pridruženimi okužbami. Poročali so tudi o novem začetku ali poslabšanju že obstoječe periferne nevropatije. Med neželenimi učinki, o katerih poročajo, je toksičnost za prebavila, ki se kaže kot anoreksija, navzea, bruhanje, driska, zaprtost in stomatitis. Med drugimi neželenimi učinki so utrujenost, alopecija, zvečani jetrni encimi, sepsa in mišičnoskeletni bolečinski sindrom. Seznam neželenih učinkov: Zelo pogosti (. 1/10): nevtropenija (57,0 %) (3./4. stopnje: 49,7 %), levkopenija (29,3 %) (3./4. stopnje: 17,3 %), anemija (20,6 %) (3./4. stopnje: 2,0 %), zmanjšan apetit (21,9 %) (3./4. stopnje: 0,7 %), periferna nevropatija (35,6 %) (3./4. stopnje: 7,6 %), glavobol (17,2 %) (3./4. stopnje: 0,8 %), dispnea (13,9 %) (3./4. stopnje: 3,1 %), kašelj (13,6 %) (3./4. stopnje: 0,6 %), navzea (33,8 %) (3./4. stopnje: 1,1 %), zaprtost (19,6 %) (3./4. stopnje: 0,6 %), driska (17,9 %) (3./4. stopnje: 0,8 %), bruhanje (17,6 %) (3./4. stopnje: 0,9 %), alopecija, artralgija in mialgija (19,4 %) (3./4. stopnje: 1,1 %), bolečina v hrbtu (13,0 %) (3./4. stopnje: 1,5 %), bolečina v udu (10,0 %) (3./4. stopnje: 0,7 %), utrujenost/astenija (47,9 %) (3./4. stopnje: 7,8 %), pireksija (20,4 %) (3./4. stopnje: 0,6 %), zmanjšanje telesne mase (11,3 %) (3./4. stopnje: 0,3 %). Pogosti (. 1/100 do < 1/10): okužba sečil (8 %) (3./4. stopnje: 0,5 %), pljučnica (1,2 %) (3./4. stopnje: 0,8 %), ustna kandidiaza, ustni herpes, okužba zgornjih dihal, nazofaringitis, rinitis, limfopenija (4,9 %) (3./4. stopnje: 1,4 %), febrilna nevtropenija (4,7 %) (3./4. stopnje: 4,5 %), trombocitopenija (4,3 %) (3./4. stopnje: 0,7 %), hipokaliemija (6,1 %) (3./4. stopnje: 1,7 %), hipomagneziemija (2,9 %) (3./4. stopnje: 0,2 %), dehidracija (2,8 %) (3./4. stopnje: 0,5 %), hiperglikemija, hipofosfatemija, nespečnost, depresija, disgevzija, omotičnost (7,9 %) (3./4. stopnje: 0,5 %), hipoestezija, letargija, nevrotoksičnost, obilnejše solzenje (6,0 %) (3./4. stopnje: 0,1 %), konjunktivitis, vrtoglavica, tahikardija, vročinski valovi, orofaringealna bolečina, epistaksa, rinoreja, bolečina v trebuhu, stomatitis (9,3 %) (3./4. stopnje: 0,8 %), suha usta, dispepsija (5,9 %) (3./4. stopnje: 0,2 %), gastroezofagealna refluksna bolezen, razjede v ustih, distenzija trebuha, zvišanje alanin-aminotransferaze (7,6 %) (3./4. stopnje: 2,1 %), zvišanje aspartat-aminotransferaze (7,4 %) (3./4. stopnje: 1,5 %), zvišanje gama-glutamiltransferaze (1,8 %) (3./4. stopnje: 0,9 %), hiperbilirubinemija (1,5 %) (3./4. stopnje: 0,3 %), izpuščaj, pruritus (3,9 %) (3./4. stopnje: 0,1 %), bolezni nohtov, nočno potenje, suha koža, eritem, hiperhidroza, bolečina v kosteh (9,6 %) (3./4. stopnje: 1,7 %), mišični spazmi (5,1 %) (3./4. stopnje: 0,1 %), mišično-skeletna bolečina in mišično­skeletna bolečina v prsih, mišična oslabelost, disurija, vnetje sluznice (8,3 %) (3./4. stopnje: 1,1 %), periferni edem, bolečina, mrzlica, bolečina v prsih, gripi podobna bolezen. Občasni (. 1/1.000 do < 1/100): sepsa (0,5 %) (3./4. stopnje: 0,2 %), nevtropenična sepsa (0,1 %) (3./4. stopnje: 0,1 %), herpes zoster, tinitus, globoka venska tromboza, pljučna embolija, hepatotoksičnost (1,0 %) (3./4. stopnje: 0.6 %), palmarno-plantarna eritrodisestezija, hematurija, proteinurija, odpoved ledvic. Redki (. 1/10.000 do < 1/1.000): diseminirana intravaskularna koagulacija, intersticijska pljučna bolezen, pankreatitis, angioedem. Za popoln opis neželenih učinkov glejte celoten povzetek glavnih značilnosti zdravila. Vrsta ovojnine in vsebina: viala z 2 ml raztopine. Režim izdaje: H Imetnik dovoljenja za promet: Eisai Europe Ltd, European Knowledge Centre, Mosquito Way, Hatfield, Hertfordshire, AL10 9SN, Velika Britanija HAL-270614, julij 2014 Pred predpisovanjem in uporabo zdravila prosimo preberite celoten povzetek glavnih značilnosti zdravila! Viri: (1) Povzetek glavnih značilnosti zdravila Halaven, junij 2014; (2) Cortes J et al. Lancet 2011; 377: 914–23. Odgovoren za trženje v Sloveniji: PharmaSwiss d.o.o., Brodišče 32, 1236 Trzin telefon: +386 1 236 47 00, faks: +386 1 283 38 10 HAL-0714-01, julij 2014 Publisher Association of Radiology and Oncology Affiliated with Slovenian Medical Association – Slovenian Association of Radiology, Nuclear Medicine Society, Slovenian Society for Radiotherapy and Oncology, and Slovenian Cancer Society Croatian Medical Association – Croatian Society of Radiology Societas Radiologorum Hungarorum Friuli-Venezia Giulia regional groups of S.I.R.M. Italian Society of Medical Radiology Aims and scope Radiology and Oncology is a journal devoted to publication of original contributions in diagnostic and interventional radiology, computerized tomography, ultrasound, magnetic resonance, nuclear medicine, radiotherapy, clinical and experimental oncology, radiobiology, radiophysics and radiation protection. Editor-in-Chief Gregor Serša, Institute of Oncology Ljubljana, Department of Experimental Oncology, Ljubljana, Slovenia Executive Editor Viljem Kovač, Institute of Oncology Ljubljana, Department of Radiation Oncology, Ljubljana, Slovenia Editorial Board Sotirios Bisdas, University Clinic Tübingen, Department of Neuroradiology, Tübingen, Germany Karl H. Bohuslavizki, Facharzt für Nuklearmedizin, Hamburg, Germany Serena Bonin, University of Trieste, Department of Medical Sciences, Trieste, Italy Boris Brkljačić, University Hospital “Dubrava”, Department of Diagnostic and Interventional Radiology, Zagreb, Croatia Luca Campana, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy Christian Dittrich, Kaiser Franz Josef - Spital, Vienna, Austria Metka Filipič, National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Ljubljana, Slovenia Maria Gődény, National Institute of Oncology, Budapest, Hungary Janko Kos, University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia Robert Jeraj, University of Wisconsin, Carbone Cancer Center, Madison, Wisconsin, USA Advisory Committee Tullio Giraldi, University of Trieste, Faculty of Medicine and Psychology, Trieste, Italy Vassil Hadjidekov, Medical University, Department of Diagnostic Imaging, Sofia, Bulgaria Deputy Editors Andrej Cör, University of Primorska, Faculty of Health Science, Izola, Slovenia Maja Čemažar, Institute of Oncology Ljubljana, Department of Experimental Oncology, Ljubljana, Slovenia Igor Kocijančič, University Medical Centre Ljubljana, Institute of Radiology, Ljubljana, Slovenia Karmen Stanič, Institute of Oncology Ljubljana, Department of Radiation Oncology, Ljubljana, Slovenia Primož Strojan, Institute of Oncology Ljubljana, Department of Radiation Oncology, Ljubljana, Slovenia Tamara Lah Turnšek, National Institute of Biology, Ljubljana, Slovenia Damijan Miklavčič, University of Ljubljana, Faculty of Electrical Engineering, Ljubljana, Slovenia Luka Milas, UT M. D. Anderson Cancer Center, Houston , USA Damir Miletić, Clinical Hospital Centre Rijeka, Department of Radiology, Rijeka, Croatia Häkan Nyström, Skandionkliniken, Uppsala, Sweden Maja Osmak, Ruder Bošković Institute, Department of Molecular Biology, Zagreb, Croatia Dušan Pavčnik, Dotter Interventional Institute, Oregon Health Science Universityte, Oregon, Portland, USA Geoffrey J. Pilkington, University of Portsmouth, School of Pharmacy and Biomedical Sciences, Portsmouth, UK Ervin B. Podgoršak, McGill University, Montreal, Canada Matthew Podgorsak, Roswell Park Cancer Institute, Departments of Biophysics and Radiation Medicine, Buffalo, NY ,USA Marko Hočevar, Institute of Oncology Ljubljana, Department of Surgical Oncology, Ljubljana, Slovenia Miklós Kásler, National Institute of Oncology, Budapest, Hungary Csaba Polgar, National Institute of Oncology, Budapest, Hungary Dirk Rades, University of Lubeck, Department of Radiation Oncology, Lubeck, Germany , Mirjana Rajer, Institute of Oncology Ljubljana, Department of Radiation Oncology, Ljubljana, Slovenia Luis Souhami, McGill University, Montreal, Canada Borut Štabuc, University Medical Centre Ljubljana, Department of Gastroenterology, Ljubljana, Slovenia Katarina Šurlan Popovič, University Medical Center Ljubljana, Clinical Institute of Radiology, Ljubljana, Slovenia Justin Teissié, CNRS, IPBS, Toulouse, France Gillian M.Tozer, University of Sheffield, Academic Unit of Surgical Oncology, Royal Hallamshire Hospital, Sheffield, UK Andrea Veronesi, Centro di Riferimento Oncologico- Aviano, Division of Medical Oncology, Aviano, Italy Branko Zakotnik, Institute of Oncology Ljubljana, Department of Medical Oncology, Ljubljana, Slovenia Stojan Plesničar, Institute of Oncology Ljubljana, Department of Radiation Oncology, Ljubljana, Slovenia Tomaž Benulič, Institute of Oncology Ljubljana, Department of Radiation Oncology, Ljubljana, Slovenia Radiol Oncol 2015; 49(4): A. Editorial office Radiology and Oncology Zaloška cesta 2 P. O. Box 2217 SI-1000 Ljubljana Slovenia Phone: +386 1 5879 369 Phone/Fax: +386 1 5879 434 E-mail: gsersa@onko-i.si Copyright © Radiology and Oncology. All rights reserved. Reader for English Vida Kološa Secretary Mira Klemenčič Zvezdana Vukmirović Design Monika Fink-Serša, Samo Rovan, Ivana Ljubanović Layout Matjaž Lužar Printed by Tiskarna Ozimek, Slovenia Published quarterly in 400 copies Beneficiary name: DRUŠTVO RADIOLOGIJE IN ONKOLOGIJE Zaloška cesta 2 1000 Ljubljana Slovenia Beneficiary bank account number: SI56 02010-0090006751 IBAN: SI56 0201 0009 0006 751 Our bank name: Nova Ljubljanska banka, d.d., Ljubljana, Trg republike 2, 1520 Ljubljana; Slovenia SWIFT: LJBASI2X Subscription fee for institutions EUR 100, individuals EUR 50 The publication of this journal is subsidized by the Slovenian Research Agency. Indexed and abstracted by: • Celdes • Chemical Abstracts Service (CAS) • Chemical Abstracts Service (CAS) - SciFinder • CNKI Scholar (China National Knowledge Infrastructure) • CNPIEC • DOAJ • EBSCO - Biomedical Reference Collection • EBSCO - Cinahl • EBSCO - TOC Premier • EBSCO Discovery Service • Elsevier - EMBASE • Elsevier - SCOPUS • Google Scholar • J-Gate • JournalTOCs • Naviga (Softweco) • Primo Central (ExLibris) • ProQuest - Advanced Technologies Database with Aerospace • ProQuest - Health & Medical Complete This journal is printed on acid- free paper On the web: ISSN 1581-3207 http://www.degruyter.com/view/j/raon http://www.radioloncol.com • ProQuest - Illustrata: Health Sciences • ProQuest - Illustrata: Technology • ProQuest - Medical Library • ProQuest - Nursing & Allied Health Source • ProQuest - Pharma Collection • ProQuest - Public Health • ProQuest - Science Journals • ProQuest - SciTech Journals • ProQuest - Technology Journals • PubMed • PubsHub • ReadCube • SCImago (SJR) • Summon (Serials Solutions/ProQuest) • TDOne (TDNet) • Thomson Reuters - Journal Citation Reports/Science Edition • Thomson Reuters - Science Citation Index Expanded • Ulrich's Periodicals Directory/ulrichsweb • WorldCat (OCLC) Radiol Oncol 2015; 49(4): B. contents contents review 315 Immunotoxin – a new treatment option in patients with relapsed and refractory Hodgkin lymphoma Barbara Jezersek Novakovic nuclear medicine 320 F-18 FDG-PET/CT parameters as predictors of outcome inoperable NSCLC patients Antonio Nappi , Rosj Gallicchio , Vittorio Simeon , Anna Nardelli , Alessandra Pelagalli , Angela Zupa , Giulia Vita , Angela Venetucci , Michele Di Cosola , Francesco Barbato , Giovanni Storto 327 Optimal scan time for evaluation of parathyroid adenoma with [18F]­fluorocholine PET/CT Sebastijan Rep, Luka Lezaic, Tomaz Kocjan, Marija Pfeifer, Mojca Jensterle, Urban Simoncic, Petra Tomse, Marko Hocevar radiology 334 Endobronchial ultrasound elastography strain ratio for mediastinal lymph node dianosis Ales Rozman, Mateja Marc Malovrh, Katja Adamic, Tjasa Subic, Viljem Kovac, Matjaz Flezar 341 Analysis of risk factors for perifocal edema after endovascular embolization of unruptured intracranial arterial aneurysms Snežana Lukić, Slobodan Janković, Katarina Popovic Šurlan, Dragić Banković, Peter Popovič. Milan Mijailović experimental oncology 347 Estimation of cell response in fractionation radiotherapy using different methods derived from linear quadratic model Safoora Nikzad, Bijan Hashemi, Golshan Mahmoudi, Milad Baradaran-Ghahfarokhi clinical oncology 357 Urokinase plasminogen activator (uPA) and plasminogen activator inhibitor type-1 (PAI-1) in breast cancer - correlation with traditional prognostic factors Maja Lampelj, Darja Arko, Nina Cas-Sikosek, Rajko Kavalar, Maja Ravnik, Barbara Jezersek-Novakovic, Sarah Dobnik, Nina Fokter Dovnik, Iztok Takac Radiol Oncol 2015; 49(4): C. contents 365 Prognostic value of some tumor markers in unresectable stage IV oropharyngeal carcinoma patients treated with concomitant radiochemotherapy Erika Soba, Marjan Budihna, Lojze Smid, Nina Gale, Hotimir Lesnicar, Branko Zakotnik, Primož Strojan 371 Clinical results of proton beam therapy for twenty older patients with esophageal cancer Takashi Ono, Tatsuya Nakamura, Yusuke Azami, Hisashi Yamaguchi, Yuichiro Hayashi, Motohisa Suzuki, Yoshiomi Hatayama, Iwao Tsukiyama, Masato Hareyama, Yasuhiro Kikuchi, Kenji Nemoto 379 Cutaneous melanoma frequencies and seasonal trend in 20 years of observation of a population characterised by excessive sun exposure Serena Bonin, Antonio Albano, Nicola di Meo, Alessandro Gatti, Giuseppe Stinco, Fabrizio Zanconati, Giusto Trevisan 386 Release of growth factors after mechanical and chemical pleurodesis for treatment malignant pleural effusion: a randomized control study Aljaž Hojski, Maja Leitgeb, Anton Crnjac 395 Giant solitary fibrous tumour of the pleura. Case report and review of the literature Anton Crnjac, Bojan Veingerl, Damjan Vidovič, Rajko Kavalar, Aljaž Hojski 402 Impact of comorbidity on the outcome in men with advanced prostate cancer treated with docetaxel Andrej Zist, Eitan Amir, Alberto F. Ocana, Bostjan Seruga 409 Clinical impact of post-progression survival on overall survival in patients with limited-stage disease small cell lung cancer after first-line chemoradiotherapy Norimitsu Kasahara, Hisao Imai, Kyoichi Kaira, Keita Mori, Kazushige Wakuda, Akira Ono, Tetsuhiko Taira, Hirotsugu Kenmotsu, Hideyuki Harada, Tateaki Naito, Haruyasu Murakami, Masahiro Endo, Takashi Nakajima, Masanobu Yamada, Toshiaki Takahashi I slovenian abstracts Radiol Oncol 2015; 49(4): D. 315 review Immunotoxin – a new treatment option in patients with relapsed and refractory Hodgkin lymphoma Barbara Jezersek Novakovic Division of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia Radiol Oncol 2015; 49(4): 315-319. Received 15 July 2015 Accepted 16 August 2015 Correspondence to: Assoc. Prof. Barbara Jezeršek Novaković, M.D., Ph.D., Division of Medical Oncology, Institute of Oncology Ljubljana, Zaloška 2, SI-1000 Ljubljana, Slovenia. Phone: +386 1 587 9280; Fax: +386 1 587 9305; E-mail: bjezersek@onko-i.si Disclosure: No potential conflicts of interest were disclosed. Background. Even though Hodgkin lymphoma is a highly curable disease, some of the patients have either a re­fractory disease or experience a relapse following a successful primary therapy. Durable responses and remissions in patients with relapsed or refractory disease may be achieved in approximately one-half with salvage chemotherapy followed by high dose chemotherapy (HDT) and autologous hematopoietic cell rescue (SCT). On the other hand, patients who relapse after HDT and autologous SCT or those who have failed at least two prior multi-agent chemo­therapy regimens and are not candidates for HDT have limited treatment options. Conclusions. A new treatment option in this population is an immunotoxin Brentuximab vedotin composed of a CD30 directed antibody linked to the antitubulin agent monomethyl auristatin E. It has demonstrated a substantial ef­fectiveness and an acceptable toxicity. In the pivotal study, the overall response rate was 75% with 34% of complete remissions. The median durations of response were 20.5 and 6.7 months for those with complete remission and all responding patients, respectively. The median overall survival was 40.5 months (3-years overall survival 54%) and the median progression-free survival 9.3 months. The most common non-hematologic toxicities were peripheral sensory neuropathy, nausea, and fatigue while the most common severe side effects were neutropenia, thrombocytopenia, anemia, and peripheral sensory neuropathy. Key words: Hodgkin lymphoma; relapsed and refractory; new treatment option; treatment effectiveness; toxicity Introduction Hodgkin lymphoma (HL) is a highly curable dis­ease. However, some of the patients have either a primary refractory disease or experience a relapse following a successful initial therapy.1-5 Primary re­fractory disease refers to those patients who do not achieve a complete remission after initial therapy. The incidence of primary refractory disease varies depending upon the stage of disease at diagnosis and the treatment regimen used and it occurs in approximately 10 to 15% of patients undergoing primary treatment.4-6 The likelihood of relapse of HL from initial therapy in the era of systemic or combined modality therapy is approximately 10 to 15% for localized HL and 20 to 40% for ad­vanced disease, dependent on prognostic factors. Approximately one-half of these relapses occur in the first 12 months from induction and an addition­al one-quarter occurs at one to three years thereaf­ter. Late relapses (i.e., more than 3 years following treatment) occur at the rate of a few percent per year, extending up to 12 years post-treatment.1-3 Durable responses and remissions in patients with primary refractory disease may be achieved in approximately one-half with second line chemo­therapy that incorporates drugs not used in initial treatment followed by high dose chemotherapy and autologous hematopoietic cell rescue (HDT and SCT). The treatment of relapsed disease pri­marily consists of combination chemotherapy with or without radiotherapy; radiotherapy alone is usu­ 316 ally not used. Salvage therapy with second or third line regimens can achieve responses in approxi­mately 50% of these patients, although long-term disease-free survival after the treatment of relapse with chemotherapy alone is less common. The choice of therapy is usually based upon prognostic features and patients with a localized, asympto­matic relapse occurring more than 12 months after the initial treatment are usually treated with con­ventional salvage chemotherapy, often combined with radiation therapy with or without HDT and autologous SCT. The value of HDT is uncertain in this group and may be unnecessarily toxic. High dose chemotherapy and autologous SCT should be considered as the treatment of choice for treatment of early relapses (less than 12 months after treat­ment) or induction failures, second relapses after conventional treatment for first relapse or general­ized systemic relapses even beyond 12 months.7-12 However, it is of great importance that complete remission is achieved prior to transplantation. This is supported by emerging experience with PET scans obtained at the end of salvage chemo­therapy, but prior to high-dose therapy with au­tologous transplant. In this setting, a negative scan has a markedly positive predictive value with 93% progression-free survival at two years in one se­ries.13 In comparison, the majority of PET-positive patients relapse despite high-dose therapy. Radiation therapy is in the relapsed setting in­dicated for patients with localized residual disease after salvage chemotherapy. In addition, patients with a localized late relapse may achieve long-term remission with chemotherapy followed by involved-field radiation therapy (IF-RT) rather than chemotherapy followed by HDT. The role of IF-RT in the treatment of patients who achieve a complete response to chemotherapy and plan to proceed with HDT is less clear.6,14,15 Risk factors for relapse after second line therapy include not only the patient and tumor specific markers (e.g., CD68 expression), but also the initial response to therapy and the duration of this ini­tial response. The German Hodgkin’s Lymphoma Study Group (GHSG) identified three adverse risk factors for second relapse following various forms of salvage therapy, which included hematopoietic cell transplantation in one-third of the cases. These adverse risk factors included time to first recur­rence of less or equal to 12 months, stage III or IV disease at first relapse and anemia at the time of first relapse.1 Patients who relapse following autologous SCT have limited treatment options. A second autolo­gous SCT is an option only in highly selected pa­tients because of their limited bone marrow reserve and due to the fact that the disease that relapses af­ter a first autologous SCT is generally more chemo­therapy resistant.16 Single agent chemotherapy is often used in this setting, but there are no guide­lines for the selection of agents. Other options in­clude the use of brentuximab vedotin, bendamus­tine, rituximab, mTOR inhibitors (e.g., everolimus), immunomodulators (e.g., lenalidomide), histone deacetylase inhibitors, adoptive immunotherapy, local regional irradiation, or allogeneic SCT.17­26 In addition, the anti PD-1 antibody nivolumab has shown quite impressive responses in patients with HL who relapsed after prior autologous SCT.27 Allogeneic SCT is usually offered to patients with HL as a salvage therapy following relapse or progression after autologous SCT. Long term remissions can be obtained after allogeneic SCT in a select group of patients. An advantage of an allogeneic SCT graft is the use of tumor-free graft and the transfer of robust immune system from a healthy donor that can mediate the “graft versus lymphoma” effect. Brentuximab vedotin Brentuximab vedotin (SGN-35) is an immunotox­in composed of a CD30 directed antibody linked to the antitubulin agent monomethyl auristatin E (MMAE).17,28 The antibody drug conjugate enables the antibody to selectively deliver a chemotherapy agent to CD30 positive tumor cells. The antibody and MMAE are joined together by a protease-cleav­ 317 able linker that is stable in plasma but degraded by lysosomal enzymes. This conjugate binds to CD30, which is expressed on the surface of HL cells, then gets internalized and traffics to lysosome, where the MMAE is released. The MMAE then disrupts the microtubule network, leading to cell cycle ar­rest in G2/M phase and apoptosis (Figure 1). Brentuximab vedotin has been approved by FDA and later EMA for the treatment of patients with relapsed or refractory CD30+ HL after failure of autologous hematopoietic SCT or after failure of at least two prior therapies in patients who are not candidates for HDT and autologous SCT or multi-agent chemotherapy.29 A subset of patients treated with brentuximab vedotin in this setting has been able to proceed with successful reduced intensity conditioning followed by allogeneic SCT.30 This drug is effective and has been approved also in re­lapsed or refractory systemic anaplastic large cell lymphoma (sALCL). Several studies have demonstrated the activity of brentuximab vedotin in patients with HL.17,31-36 In a phase I trial SG035-0001 of brentuximab vedo­tin in 45 patients with CD30 positive hematopoietic cancers (42 of whom had HL) relapsed or refrac­tory after a median of three prior therapies, there were 11 complete remissions (CR), six partial re­missions (PR), and 19 cases with stable disease.31 Overall response rate in patients receiving 1.8 mg/ kg was 50% and median duration of objective re­sponse was 9.7 months with a median progression-free survival of 5.9 months. Side effects were gen­erally mild with the most common being fatigue, pyrexia, diarrhea, nausea, neutropenia, and pe­ripheral neuropathy. The dose limiting toxicity at 2.7 mg/kg was acute renal failure, hyperglycemia, prostatitis, and febrile neutropenia. The maximum tolerated dose was defined as 1.8 mg/kg i.v. every three weeks. Another phase I trial SG035-0002 was a dose-es­calation study (0.4–1.4 mg/kg) of brentuximab ve­dotin given on days 1, 8, and 15 of 28-day cycles.37 Forty-four patients with relapsed or refractory CD30+ hematological malignancies were included, 38 of them with relapsed or refractory HL, 68% of all patients were previously treated with HDT. The maximum tolerated dose was defined as 1.2 mg/kg, the most common side effects were peripheral sen­sory neuropathy (66%), fatigue (52%), and nausea (50%). The overall response rate for all dose groups was 59% (34% CR), and with a median follow-up of 45.1 weeks the median progression-free survival was 28.7 weeks and the median overall survival has not been reached yet. In a pivotal phase II multicenter trial SG035­0003, 102 patients with relapsed or refractory HL after prior autologous SCT were treated with bren­tuximab vedotin (1.8 mg/kg every three weeks for up to 16 cycles).17 The overall response rate was 75% (34% CR) with median times to objective response and CR of 5.7 and 12 weeks, respectively. Median durations of response were 20.5 and 6.7 months for those with CR and all responding patients, respec­tively. At a median follow-up of 18.5 months, the median progression-free survivals were 21.7 and 5.6 months for patients with CR and all patients, re­spectively. After responding to brentuximab vedo­tin, eight patients received an allo-SCT as their first subsequent therapy - five patients with CR and three patients with PR. All eight patients were alive and remained in follow-up at the time of data cut­off. The most common non-hematologic toxicities were peripheral sensory neuropathy (42%), nausea (35%), and fatigue (34%). The most common severe (grade 3/4) side effects were neutropenia (20%), thrombocytopenia (8%), anemia (6%), and periph­eral sensory neuropathy (8%). The data of the SG035-0003 were updated with a median follow-up of 33.3 months post first bren­tuximab vedotin dose.38 The updated median over­all survival and progression-free survival were es­timated at 40.5 months and 9.3 months, respective­ly. Improved outcomes were observed in patients who achieved a CR, with estimated 3-year overall survival and progression-free survival rates of 73% and 58%, respectively, in this group (and medians not reached). Of the 34 patients who obtained CR, 16 (47%) remained progression-free after a median of 53.3 months of observation, 12 patients remained progression-free without a consolidative alloge­neic stem cell transplant. Those patients in remis­sion tended to be younger, predominately females, diagnosed with HL for a shorter period prior to receiving brentuximab vedotin, having a relapsed rather than refractory disease, with a lower ECOG performance score and a smaller disease burden prior to enrollment. They also received more cy­cles of brentuximab vedotin but with an equal in­cidence and severity of adverse events relative to other patients. In another phase II multicenter trial, 25 patients with relapsed HL after prior allogeneic SCT were treated with brentuximab vedotin (1.2 mg/kg or 1.8 mg/kg every three weeks for up to 16 cycles).32 The overall response rate was 50% (38% CR) with a me­dian time to response of 8.1 weeks. At a median fol­low-up of 34 weeks, the median progression-free survival was 7.8 months and the median overall 318 survival has not been reached. The most common toxicities were cough, fatigue, and pyrexia (52% each), nausea and peripheral sensory neuropathy (48% each), and dyspnea (40%). The most com­mon severe (grade 3/4) toxicities were neutropenia (24%) and anemia (20%). Rothe et al.39 reported effectiveness and safety in 16 patients with HL (14 patients) or sALCL (2 pa­tients) who have not undergone a prior HDT from GHSG enrolled in a named patient program with brentuximab vedotin 1.8 mg/kg every three weeks. Five patients achieved CR and 6 patients PR giv­ing an overall response rate of 69%. Six of the 16 patients proceeded to HDT with autologous trans­plant in 4 and allogeneic transplant in 2 patients. The 12-month overall survival for all patients was 68% and the 12-month progression-free survival 22%. The toxicity profile was similar as reported in previous studies. Rothe et al.33 also evaluated brentuximab vedotin in relapsed and refractory HL. Results were pooled from the multicentric study by the GHSG including 45 HL patients (34 patients from the named patient program) and SGN35-007 study (11 patients), 87% of them receiving a previous transplant. The over­all response rate was 60%, with 22% of CR and 38% of PR. The median progression-free survival was 8 months and the progression-free survival at 12 months 43% with an overall survival of 83% at 12 months and median overall survival not reached. Grade 3-4 toxicities were of similar frequencies as reported before - there were 13% of neutropenia/ sepsis, 7% of thrombocytopenia, and 7% of infec­tions with no grade 3-4 neuropathy reported. Brentuximab vedotin is usually well tolerated with an acceptable toxicity profile in these usually heavily pretreated patients.17,31-33,37-43 Infusion reac­tions are uncommon but severe anaphylaxis has been reported.29 Peripheral polyneuropathy is one of the most common side effects of brentuximab ve­dotin treatment with 53% of patients experiencing at least one treatment emerging event. However, in 80% of these patients the neuropathy presented as partially or completely reversible.17 Concomitant administration of brentuximab vedotin and bleo­mycin is contraindicated due to unacceptable pul­monary toxicity. Progressive multifocal leukoen­cephalopathy (PML) and acute pancreatitis are rare but potentially fatal complications of brentuximab vedotin treatment.42,43 In PML, other possible con­tributory factors beside brentuximab include prior therapies and underlying disease that may cause immunosuppression.41 Brentuximab vedotin has been available since 2012 also in Slovenia. Up till now, we have treated five adult patients with relapsed or refractory HL. All of them were heavily pretreated-in three pa­tients brentuximab vedotin was more than third line treatment (in two of them being the seventh line of treatment), in one patient it was given fol­lowing the autologous and in one patient following the allogeneic SCT. Patients received from 3 to 16 cycles of brentuximab but no CRs were achieved-there were two PRs and in two patients the disease progressed while the fifth patient is still under treatment awaiting first treatment evaluation. The treatment was very well tolerated in all patients and no dose reductions were needed except in the fifth patient who is receiving brentuximab vedotin at a reduced dosage due to a preexisting neuropathy. Regarding the inferior effectiveness in our patients in comparison with the one reported in the above studies17,31,38 we conclude that it must be on account of the heavy pretreatment of all our patients sug­gesting an earlier use of brentuximab vedotin. Conclusions Patients with relapsed of refractory HL have lim­ited treatment options - especially those who re­lapse after HDT and autologous SCT or those who have failed at least two prior multi-agent chemo­therapy regimens and are not candidates for HDT. Brentuximab vedotin represents an additional treatment option with substantial effectiveness and acceptable toxicity in this select population. References 1. Josting A, Franklin J, May M, Koch P, Beykirch MK, Heinz J, et al. New prognostic score based on treatment outcome of patients with relapsed Hodgkin’s lymphoma registered in the database of the German Hodgkin’s lymphoma study group. J Clin Oncol 2002; 20: 221-30. 2. Bonfante V, Santoro A, Viviani S, Devizzi L, Balzarotti M, Soncini F, et al. Outcome of patients with Hodgkin’s disease failing after primary MOPP­ABVD. J Clin Oncol 1997; 15: 528-34. 3. Radman I, Basić N, Labar B, Kovacević J, Aurer I, Bogdanić V, et al. Long-term results of conventional-dose salvage chemotherapy in patients with refractory and relapsed Hodgkin’s disease (Croatian experience). Ann Oncol 2002; 13: 1650-5. 4. Santoro A, Bonadonna G, Valagussa P, Zucali R, Viviani S, Villani F, et al. Long-term results of combined chemotherapy-radiotherapy approach in Hodgkin’s disease: superiority of ABVD plus radiotherapy versus MOPP plus radiotherapy. J Clin Oncol 1987; 5: 27-37. 5. Canellos GP, Anderson JR, Propert KJ, Nissen N, Cooper MR, Henderson ES, et al. Chemotherapy of advanced Hodgkin’s disease with MOPP, ABVD, or MOPP alternating with ABVD. N Engl J Med 1992; 327: 1478-84. 6. Brice P. Managing relapsed and refractory Hodgkin lymphoma. Br J Haematol 2008; 141: 3-13. 319 7. Lavoie JC, Connors JM, Phillips GL, Reece DE, Barnett MJ, Forrest DL, et al. High-dose chemotherapy and autologous stem cell transplantation for pri­mary refractory or relapsed Hodgkin lymphoma: long-term outcome in the first 100 patients treated in Vancouver. Blood 2005; 106: 1473-8. 8. Czyz J, Szydlo R, Knopinska-Posluszny W, Hellmann A, Gozdzik J, Hansz J, et al. Treatment for primary refractory Hodgkin’s disease: a comparison of high-dose chemotherapy followed by ASCT with conventional therapy. Bone Marrow Transplant 2004; 33: 1225-9. 9. Morabito F, Stelitano C, Luminari S, Mammi C, Marcheselli L, Callea V, et al. The role of high-dose therapy and autologous stem cell transplantation in patients with primary refractory Hodgkin’s lymphoma: a report from the Gruppo Italiano per lo Studio dei Linfomi (GISL). Bone Marrow Transplant 2006; 37: 283-8. 10. Nieto Y, Popat U, Anderlini P, Valdez B, Andersson B, Liu P, et al. Autologous stem cell transplantation for refractory or poor-risk relapsed Hodgkin’s lymphoma: effect of the specific high-dose chemotherapy regimen on outcome. Biol Blood Marrow Transplant 2013; 19: 410-7. 11. Popat U, Hosing C, Saliba RM, Anderlini P, van Besien K, Przepiorka D, et al. Prognostic factors for disease progression after high-dose chemotherapy and autologous hematopoietic stem cell transplantation for recurrent or re­fractory Hodgkin’s lymphoma. Bone Marrow Transplant 2004; 33: 1015-23. 12. Puig N, Pintilie M, Seshadri T, Al-Farsi K, Nagy T, Franke N, et al. Different response to salvage chemotherapy but similar post-transplant outcomes in patients with relapsed and refractory Hodgkin’s lymphoma. Haematologica 2010; 95: 1496-502. 13. Castagna L, Bramanti S, Balzarotti M, Sarina B, Todisco E, Anastasia A, et al. Predictive value of early 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) during salvage chemotherapy in relapsing/refrac­tory Hodgkin lymphoma (HL) treated with high-dose chemotherapy. Br J Haematol 2009; 145: 369-72. 14. Collins GP, Parker AN, Pocock C, Kayani I, Sureda A, Illidge T, et al. Guideline on the management of primary resistant and relapsed classical Hodgkin lymphoma. Br J Haematol 2014; 164: 39-52. 15. The National Comprehensive Cancer Network. NCCN Guidelines. [citated 2015 Jul 6]. Available at http://www.nccn.org/professionals/physician_gls/ pdf/hodgkins.pdf. 16. Thomson KJ, Peggs KS, Blundell E, Goldstone AH, Linch DC. A second au­tologous transplant may be efficacious in selected patients with Hodgkin’s lymphoma relapsing after a previous autograft. Leuk Lymphoma 2007; 48: 881-4. 17. Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol 2012; 30: 2183-9. 18. Devizzi L, Santoro A, Bonfante V, Viviani S, Balzarini L, Valagussa P, Bonadonna G. Vinorelbine: an active drug for the management of patients with heavily pretreated Hodgkin’s disease. Ann Oncol 1994; 5: 817-20. 19. Moskowitz AJ, Hamlin PA Jr, Perales MA, Gerecitano J, Horwitz SM, Matasar MJ, et al. Phase II study of bendamustine in relapsed and refractory Hodgkin lymphoma. J Clin Oncol 2013; 31: 456-60. 20. Younes A, Oki Y, McLaughlin P, Copeland AR, Goy A, Pro B, et al. Phase 2 study of rituximab plus ABVD in patients with newly diagnosed classical Hodgkin lymphoma. Blood 2012; 119: 4123-8. 21. Kasamon YL, Jacene HA, Gocke CD, Swinnen LJ, Gladstone DE, Perkins B, et al. Phase 2 study of rituximab-ABVD in classical Hodgkin lymphoma. Blood 2012; 119: 4129-32. 22. Johnston PB, Inwards DJ, Colgan JP, Laplant BR, Kabat BF, Habermann TM, et al. A Phase II trial of the oral mTOR inhibitor everolimus in relapsed Hodgkin lymphoma. Am J Hematol 2010; 85: 320-4. 23. Venkatesh H, Di Bella N, Flynn TP, Vellek MJ, Boehm KA, Asmar L. Results of a phase II multicenter trial of single-agent gemcitabine in patients with relapsed or chemotherapy-refractory Hodgkin’s lymphoma. Clin Lymphoma 2004; 5: 110-5. 24. Provencio M, Sánchez A, Sánchez-Beato M. New drugs and targeted treat­ments in Hodgkin’s lymphoma. Cancer Treat Rev 2014; 40: 457-64. 25. Kharfan-Dabaja MA, Hamadani M, Sibai H, Savani BN. Managing Hodgkin lymphoma relapsing after autologous hematopoietic cell transplantation: a not-so-good cancer after all! Bone Marrow Transplant 2014; 49: 599-606. 26. Younes A, Sureda A, Ben-Yehuda D, Zinzani PL, Ong TC, Prince HM, et al. Panobinostat in patients with relapsed/refractory Hodgkin’s lymphoma after autologous stem-cell transplantation: results of a phase II study. J Clin Oncol 2012; 30: 2197-203. 27. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lym­phoma. N Engl J Med 2015; 372: 311-9. 28. Okeley NM, Miyamoto JB, Zhang X, Sanderson RJ, Benjamin DR, Sievers EL, et al. Intracellular activation of SGN-35, a potent anti-CD30 antibody-drug conjugate. Clin Cancer Res 2010; 16: 888-97. 29. eMC. Adcetris 50 mg powder for concentrate for solution for infusion. [citated 2015 Jul 6]. Available at https://www.medicines.org.uk/emc/ medicine/27173. 30. Chen R, Palmer JM, Thomas SH, Tsai NC, Farol L, Nademanee A, et al. Brentuximab vedotin enables successful reduced-intensity allogeneic he­matopoietic cell transplantation in patients with relapsed or refractory Hodgkin lymphoma. Blood 2012; 119: 6379-81. 31. Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers EL, Forero-Torres A. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med 2010; 363: 1812-21. 32. Gopal AK, Ramchandren R, O’Connor OA, Berryman RB, Advani RH, Chen R, et al. Safety and efficacy of brentuximab vedotin for Hodgkin lymphoma recurring after allogeneic stem cell transplantation. Blood 2012; 120: 560-8. 33. Rothe A, Sasse S, Goergen H, Eichenauer DA, Lohri A, Jäger U, et al. Brentuximab vedotin for relapsed or refractory CD30+ hematologic malig­nancies: the German Hodgkin Study Group experience. Blood 2012; 120: 1470-2. 34. Forero-Torres A, Fanale M, Advani R, Bartlett NL, Rosenblatt JD, Kennedy DA, Younes A. Brentuximab vedotin in transplant-naive patients with re­lapsed or refractory hodgkin lymphoma: analysis of two phase I studies. Oncologist 2012; 17: 1073-80. 35. Gibb A, Jones C, Bloor A, Kulkarni S, Illidge T, Linton K, Radford J. Brentuximab vedotin in refractory CD30+ lymphomas: a bridge to allogeneic transplanta­tion in approximately one quarter of patients treated on a Named Patient Programme at a single UK center. Haematologica 2013; 98: 611-4. 36. Zinzani PL, Viviani S, Anastasia A, Vitolo U, Luminari S, Zaja F, et al. Brentuximab vedotin in relapsed/refractory Hodgkin’s lymphoma: the Italian experience and results of its use in daily clinical practice outside clini­cal trials. Haematologica 2013; 98: 1232-6. 37. Fanale MA, Forero-Torres A, Rosenblatt JD, Advani RH, Franklin AR, Kennedy DA, et al. A Phase I Weekly Dosing Study of Brentuximab Vedotin in Patients with Relapsed/Refractory CD30-Positive Hematologic Malignancies. Clin Cancer Res 2012; 18: 248-55. 38. Gopal AK, Chen R, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, et al. Durable remissions in a pivotal phase 2 study of brentuximab vedotin in relapsed or refractory Hodgkin lymphoma. Blood 2015; 125: 1236-43. 39. Rothe A, Sasse S, Goergen H, Eichenauer DA, Lohri A, Jäger U, et al. Brentuximab vedotin (SGN-35) in patients with relapsed/refractory CD30­positive hematologic malignancies without prior high-dose chemotherapy and stem cell transplantation. [ASH Annual Meeting Abstracts]. Blood 2012; 120: Abstract 2743. 40. Younes A, Connors JM, Park SI, Fanale M, O’Meara MM, Hunder NN, et al. Brentuximab vedotin combined with ABVD or AVD for patients with newly diagnosed Hodgkin’s lymphoma: a phase 1, open-label, dose-escalation study. Lancet Oncol 2013; 14: 1348-56. 41. Wagner-Johnston ND, Bartlett NL, Cashen A, Berger JR. Progressive multifo­cal leukoencephalopathy in a patient with Hodgkin lymphoma treated with brentuximab vedotin. Leuk Lymphoma 2012; 53: 2283-6. 42. Jalan P, Mahajan A, Pandav V, Bekker S, Koirala J. Brentuximab associated progressive multifocal leukoencephalopathy. Clin Neurol Neurosurg 2012; 114: 1335-7. 43. Urru SA, Mariotti E, Carta P, Massidda S, Marcias M, Murru R, et al. Acute pancreatitis following brentuximab vedotin therapy for refractory Hodgkin lymphoma: a case report. Drugs R D 2014; 14: 9-11. 320 research article [F-18] FDG-PET/CT parameters as predictors of outcome in inoperable NSCLC patients Antonio Nappi1, Rosj Gallicchio1, Vittorio Simeon2, Anna Nardelli3, Alessandra Pelagalli3, Angela Zupa2, Giulia Vita2, Angela Venetucci1, Michele Di Cosola4, Francesco Barbato5, Giovanni Storto1 1 Nuclear Medicine Department, 2 Research Laboratories, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Centro di Riferimento Oncologico di Basilicata (CROB), Rionero in Vulture, Italy 3 Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche, Napoli, Italy 4 Universita degli Studi di Foggia, Italy 5 CMO Oplonti Medical Centre, Italy Radiol Oncol 2015; 49(4): 320-326. Received 19 May 2015 Accepted 11 September 2015 Correspondence to: Giovanni Storto, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Centro di Riferimento Oncologico di Basilicata (CROB), Via P. Pio 1, 85028 Rionero in Vulture, Italy. Phone: +39 0972 726560; Fax: +39 0972 726399; E-mail: giosto24@hotmail.com Disclosures: No potential conflicts of interest were disclosed. Background. We evaluated the prognostic significance of standardized uptake value (SUVmax), metabolic tumour volume (MTV), and total lesion glycolysis (TLG) in [F-18] FDG PET/CT findings in patients with inoperable non-small-cell lung cancer (NSCLC). Patients and methods. One hundred and three patients (mean age, 65.6 ± 16 years) underwent [F-18] FDG PET/ CT before the chemotherapy. The SUVmax value, the MTV (cm3; 42% threshold) and the TLG (g) were registered. The patients were followed up to 18 months thereafter (range 12-55 months). Failure to respond without progression, pro­gression and/or disease-related death constituted surrogate end-points. The optimal SUVmax, MTV and TLG cut-off to predict the patients’ outcome were estimated. PET/CT results were then related to disease outcome (progression free survival; PFS). Results. The Kaplan-Meier survival analysis for SUVmax showed a significant shorter PFS in patients presenting with lower values as compared to those with higher (p < 0.05, log-rank test). MTV and TLG were not suitable for predicting PFS apart from the subset of patients with mediastinal nodal involvement. Conclusions. Despite the availability of new tools for the quantitative assessment of disease activity on PET/CT, the SUVmax rather than MTV and TLG remains the only predictor for PFS in NSCLC patients. MTV holds a value only when concomitant nodal involvement occurs. Key words: Non-small-cell lung cancer; [F-18] FDG PET/CT; quantitative assessment; glycolytic activity; survival Introduction Lung cancer constitutes the most common cause of cancer death around the world and is the second most common gender unrelated cancer.1-4 Non– small-cell lung cancer (NSCLC) includes up to 85% of all lung cancer cases.1 The treatment and prog­nosis of NSCLC depend mostly upon the stage out­lined according to the American Joint Committee on Cancer (AJCC) staging system.1,5-7 Although NSCLC remains a deadly cancer, to identify prog­nostic factors represents a clinical challenge since a modulated therapy is still possible. The TNM stag­ing as well as the stage grouping (I–IV) have been largely disputed so far. Other patients’ specific factors such as age, pulmonary performance, and co-morbidity might also influence the selection for treatment options.1,5 In fact, for the early-stage NSCLC, surgical resection is the standard of care, whereas in patients with unresectable, locally ad­vanced tumour, as per stage III NSCLC, the chem­otherapy in combination with radiation therapy 321 represents the best option.7-9 Systemic chemothera­py is reserved for the stage IV patients.10-12 Over the paste decade FDG (18F-fluoro-2-deoxy-D-glucose) PET/CT has demonstrated to be a powerful tool for staging and assessment of the treatment response in patients suffering from NSCLC.13,14 Even if PET/ CT is widely used in this setting, only limited data are presently available to describe in concert the role of PET/CT quantitative parameters for the prediction of the disease outcome (with special emphasis to those recently introduced).15,16 The findings are discordant being alternatively allotted as valuable or worthless.17-19 At this time, the meta­bolic tumour volume (MTV) and the total lesion glycolysis (TLG) are measures of the metabolic ac­tivity of tumours derived from the [F-18] FDG up­take on PET/CT images. Initial data have recently addressed their value in NSCLC.20 The MTV and the TLG can be easily calculated in the primary tumour by means of a segmentation technique. The manual or semiautomatic measurement of the pre-treatment MTV has been shown to be better than SUVmax for predicting patients prognosis in different solid neoplasms such as head and neck cancer, with or without metastases.21,22 Our study was undertaken to investigate the relationship be­tween the functional tumour parameters at staging (SUVmax, MTV, and TLG) and the progression free survival in inoperable patients with NSCLC, presenting with stage IIIB/IV, for whom a chemo­therapy (alone or in combination with radiothera­py) was shortly planned. Patients and methods Patients Two hundred and ninety patients suffering from inoperable NSCLC were referred for a staging PET/ CT scan prior to the start of chemotherapy between January 2008 and January 2012. Among them 103 (17 women; mean age 65,6 ± 16 years) strictly ful­filled inclusion criteria, which were: age at entry of 18 years or older; negative pregnancy test; stage IIIB and stage IV without distant metastases out of chest (inoperable tumour, CT staged, histologically proven). Patients whose lesions were excised surgi­cally before PET/CT imaging or who had received neo-adjuvant chemo-radiotherapy within the pre­ceding 6 months prior to PET scan were excluded from this study. Individual data are summarized in Table 1. Our institutional review board provided approval for the procedures included in the study. All patients who underwent PET/CT scan signed an informed consent form in accordance with the Declaration of Helsinki. Imaging technique The patients were well-hydrated before receiving [F-18] FDG intravenously (444–555 MBq). Sixty minutes after the tracer injection, PET and CT scans were obtained using a commercial PET/CT scanner (GE Discovery VCT scanner; Waukesha, WI) that combines a PET scanner and a Light Speed VCT sixty-four row MDCT system. MDCT (pitchx 1.5; 120 mAs; 120 kVp) was performed without the use of an intravenous and/or oral contrast medium. The PET scanning was subsequently performed, ac­quiring 4 minutes per bed position and six to eight bed positions per patient, depending on patient height. The raw CT data were reconstructed into transverse images with a 3.75-mm section thick­ness. Sagittal and coronal CT images were gener­ated by reconstruction of the transverse data. Raw PET data were reconstructed with and without at­tenuation correction into transverse, sagittal, and coronal images. Attenuation correction was based on the CT attenuation coefficients, which were de­termined by iterative reconstruction. The patients were kept fasting for at least 6 hours prior to the imaging. Blood glucose levels were measured in all patients before [F-18] FDG administration; patients with values above 7.77 mMol/L (140 ml/dl) were excluded from examination. Imaging evaluation All images were reviewed by using a PET/CT fusion software (Volumetrix for PET-CT, GE Healthcare, Waukesha, WI, USA). Each PET/CT study was interpreted by two experienced nuclear medicine physicians (G.S., and A.N., each with fif­teen years of expertise); one of them also being a board certified radiologist. They were blinded to the patient histories. The examiners first evaluated the CT images alone. The primary lesion size was visually estimated and measured for minimum and maximum diameters by using vendor-provid­ed software. A primary lesion was defined as soli­tary pulmonary nodule or peripheral nodule with an ill-defined, irregular, and spiculated border as well as identifiable mass or densitometric modi­fications of lung parenchyma (as per tissue thick­ening with or without air bronchogram findings and pseudocavitation/focal lucency, ground-glass opacification). Peribronchial/mediastinal lymph 322 TABLE 1. Overall patient characteristics Age at diagnosis, years, median (range) Gender ratio (M : F) Histological type Adenocarcinoma (%) Squamous cell carcinoma (%) Broncho-alveolar carcinoma (%) Adeno-squamous carcinoma (%) Others (%) Stage at diagnosis IIIB (%) IV (%) Chemotherapy (%) Radiotherapy (%) Radio-chemotherapy combination (%) Locoregional/mediastinal lymph node involvement Yes (%) No (%) Final patient status No response/Progression (%) Death (%) 68 (48–79) 86 :17 67 (65) 27 (26) 5 (5) 2 (2) 2 (2) 75 (73) 28 (27) 22 (21) 21 (20) 60 (59) 63 (61) 40 (39) 12 (43) 16 (57) nodes larger than 1 cm in minimum diameter and with altered shape or hilus, assessed with chest window CT settings, were considered suspicious. The PET studies were evaluated both visually and semi-quantitatively. Then, the maximum stand­ardized uptake values and body weight corrected (SUVmax) as well as the metabolic tumour volume (MTV; cm3; 42% threshold) and total lesion glyco­lysis (TLG; g) were determined by using the same vendor-provided software for the primary lesion. The MTV was defined as the volume with SUV over 42% of SUVmax. TLG was calculated as the product of SUVmean and MTV (TLG = SUVmean × MTV). Finally, lymph nodes were evaluated sys­tematically according to the topographic criteria on the PET/CT scan using the SUVmax to determine their metabolic activity, if any. As previously reported, a conventional SUVmax cut-off value of 2.5 (settled in studies using ROC curve analysis) has been considered to provide ex­cellent specificity and sensitivity for detecting le­sions.15,19,23 Accordingly, a significant uptake was reported to be higher than 2.5. Follow-up assessment The patients were categorized into two groups ac­cording to SUVmax, MTV and TLG cut-off points determined by receiver-operator-curve (ROC) analysis. The performance status and the status of disease were followed up to 18 months there­after (range 12–55). The evaluation was carried out by means of clinical and laboratory param­eters during scheduled or unscheduled visits, on the basis of diagnostic imaging (i.e. CT) results, if any, as well as by phone interview. Failure to re­spond without disease progression, disease pro­gression after four cycles of chemotherapy and/ or disease-related death were defined as surrogate end-points. Disease progression was identified as documentation of a new lesion or enlargement of a previous existing lesion; when there was missing information, the date of unscheduled new, alterna­tive treatment was considered. Clinical parameters and PET/CT results were then correlated to the progression free survival (PFS). PFS was defined as the time from PET/CT until end point occurrence or the time of last censor. Statistical analysis Continuous data were expressed as the mean ± 1 SD and median, as appropriate. Comparisons be­tween the mean values were performed with an unpaired Student’s t test (two-tailed probability) or Wilcoxon rank-sum test, according to skew­ness and kurtosis test for normality test. The ROC analysis was performed to estimate the optimal cut-off of SUVmax, MTV and TLG for differen­tiating patients at high risk of end-points occur­rence. Sensitivity and specificity were computed according to the standard method. Both univariate and multivariate regression analysis were used. A multivariate binary logistic regression analysis (enter method) was used to test the chosen (ho­mogeneously available) independent variables such as age, gender, the presence of loco-regional nodal involvement and the lesion SUVmax, MTV and TLG for their association with unsuccessful outcome. The PET/CT results were then correlated to the PFS. The Kaplan-Meier method was used to plot PFS. The predefined cut-off point for SUVmax, MTV and TLG were adopted, and the curves were compared by log-rank testing. Survival analysis was performed by univariate Cox proportional hazard regression analysis. A probability (p) value < 0.05 was considered statistically significant. Results Patient characteristics Patients with NSCLC were evaluated during the study period. Among those enrolled, none had a surgical intervention. All the patients were wild-type (no EGFR mutations, no ALK rearrangements detected) and underwent chemotherapy (from 4 to 323 6 cycles) after the PET/CT study; they were treated with cisplatin-based regimens with or without ra­diation therapy according to established dose or fractionation schedules up to 60 Gy–70 Gy (in 6–7 weeks). Overall individual patient characteristics are reported in Table 1. Energetic turnover measurements The PET/CT was reported as positive in all pa­tients (i.e. significant uptake; visually detectable and SUV higher than 2.5). The median SUVmax value was 7.3 (range 2.7–44), median MTV was 16.5 cm3 (range 3.7–38.1) and median TLG was 274.5 g (range 72.6–1039.9). According to nodal status, the median SUVmax value was 4.6 (range 2.7–23) and 10.7 (range 2.5–44) in patients without and with nodal involvement, respectively (p < 0.05), whereas median MTV was 15.2 (3.7–88) cm3 and 18.6 (5.1– 38.1) cm3, respectively (p = 0.07). TLG values were not significantly different as well. Prediction and discriminating values of SUVmax, MTV and TLG The ROC curve analysis recognizing the cut off value of SUVmax, MTV and TLG is showed in Figure 1. The area under the curve (AUC) for SUVmax was 0.64 and 6.3 was established as the cut off value. The AUC for MTV and TLG was 0.52 and 0.6, respectively whereas MTV and TLG cut off values were 8.4 and 259.0, respectively. The sensi­tivity of SUVmax cut-off on PET/CT for predicting the outcome was 75% whereas specificity was 57%. The sensitivity and specificity of MTV and TLG cut-off were 85% and 33%, and 61% and 52%, re­spectively. At univariate logistic regression analy­sis both the SUVmax (p < 0.01; OR 3.9) and pres­ence of nodal involvement (p < 0.001; OR 4.3) were predictive of the end-points, whereas MTV, TLG, age and gender were not. At multivariate logistic regression analysis only the presence of loco-re­gional nodal involvement on PET/CT contributed to the prediction of the end points occurrence (p < 0.05; OR 3.2), whereas MTV, TLG, age and gender did not. The SUVmax showed a trend for predict­ing the outcome (p= 0.08; OR 2.6), (Table 2). Clinical end points and follow up Overall 28 of 103 patients (27%) reached the end­point, 12 experienced no response/progression (43%) and 16 (57%) died. The mean SUVmax value in patients who reached an end point was higher as compared to those who did not (11.2±4.4 vs 8.4±5.9; p < 0.05, Wilcoxon rank-sum test). The median follow up was 18 months (range 12– 55 months). The Kaplan-Meier survival analysis for SUVmax showed a significant difference in PFS (p < 0.05, log-rank test). Shorter PFS was observed in patients with lesion SUVmax over 6.3 as compared to those with SUVmax values below this value (me­dian survival: 33 vs 41 months; p < 0.05, cox regres­sion) (Figure 2a). Patients with nodal involvement showed a shorter PFS as compared to those who did not (median survival: 30 vs 37 months; p < 0.05, cox regression) (Figure 2b). When nodal involve­ment was aggregated to higher SUVmax the pa­tients showed a shorter PFS as compared to those without nodal involvement and lower SUVmax (Figure 2c). Noteworthy, in the sub-group with nodal in­volvement, patients with lower MTV showed better outcome as compared to those with higher MTV value (median MTV 10.8; range 5.1–40 vs 17.6; range 9.7–381; p < 0.05). At ROC curve analy­ (N-, solid line) and with (N+, dotted line) nodal concern. (C) Survival by combination of SUVmax and nodal positivity. Kaplan-Meier graph of both SUVmax and nodal positivity and Progression Free Survival. SUVmax+ and SUVmax- indicate values of SUVmax above and below the cut off value of 6.3, respectively. N+ and N- indicate the presence and the absence of locoregional lymph-nodal involvement discovered at PET/CT. TABLE 2. Logistic regression analysis promise.24 This distressed scenario imposes the re-weighting of the prognostic factors available, es- Risk of progression pecially those allowing the correct understanding of tumour biology and its therapeutic sensitivity.25 OR 95% CI p OR 95% CI p Despite the efficacy of the new therapeutic agents, Age 0.95 0.9–1.007 0.093 - - - the necessity arises for directing their strength in Gender 2.25 0.75–6.76 0.149 - - - accordance to the tumour intrinsic features since SUVmax* MTV* 3.9 2.9 1.38–11.0 0.85–9.88 0.010 0.088 2.65 - 0.87–8.06 - 0.085 - metabolic pathways differ from subject to subject, claiming for a further specific treatment approach. Nowadays, [F-18] FDG PET/CT provides a global TLG* 2.6 0.89–7.81 0.080 - - - assessment of NSCLC patients for staging, re-stag­ Lymph-node 4.37 1.6–11.95 0.004 3.2 1.09–9.25 0.034 ing and therapy evaluation, having a prognostic * Dichotomized variables on ROC analysis basis; OR = odds ratio; CI = confidence interval; SUVmax = standardized uptake value; MTV = metabolic tumour volume; TLG = total lesion glycolysis. sis for this subgroup the AUC was 0.74 and 10.9 was the cut off value. The sensitivity and specificity of MTV cut-off on PET/CT for predicting the out­come were 94% and 54%, respectively. However, in this subgroup the MTV constituted a risk factor when settled as continuous variable (hazard ratio 1.03 per unit increment; p < 0.05; 95% CI: 1.005– 1.060), but was not able to predict PFS once used as dichotomous variable (hazard ratio 2.14; p = 0.47; 95% CI: 0.27–17.3). Discussion Current treatment approach based on stage of the disease (operable and inoperable tumours) is not entirely satisfactory in NSCLC, whilst the imple­mentation of more novel therapeutic strategies targeting specific receptors continues to hold great value as well.26 In addition, the PET/CT methodol­ogy allows a reproducible semi-quantitative assess­ment by means of several uptake indices, including the estimate of the whole tumour burden.20 Among the PET parameters, SUVmax is the most com­monly used. It reflects tumour glucose metabolism of the most aggressive cell component, given that previous studies have suggested the association between SUVmax and tumour aggressiveness.27 Conversely, the metabolic tumour volume as well as the total lesion glycolysis allow to estimate the tumour energetic turn-over throughout the volume of the lesion above a minimum threshold designed to exclude background activity. Since all the quantitative parameters derived from PET/CT have not yet been comprehensively investigated as prognostic factors in inoperable NSCLC patients, we evaluated, in a similar setting, the predictive impact of SUVmax, MTV, TLG in NSCLC patients, before radio-chemotherapy. The main finding of the present study is that SUVmax constitutes the only metabolic parameter, among the others, able to predict the progression 325 free survival in inoperable NSCLC patients having stage IIIB or chest confined stage IV, whereas MTV holds a slightly but predictive value only in case of loco-regional lympho-nodal involvement. According to previous studies, patients pre­senting with higher values of SUVmax showed a poor outcome as compared to those having lower, indicating that the high magnitude of glycolytic activity, rather than the extent of metabolic tu­mour burden, predicts a poor response to subse­quent therapy.28 As a result, lower SUVmax val­ues showed a virtually “protective” significance, whereas the MTV and the TLG did not impact on the outcome of patients. Unfortunately, this dichot­omous standpoint does not stratify the grey zone of patients discovered/confirmed to have nodal metabolic load (at PET/CT finding). Such an aspect has not been assessed, if not partially, in previous studies.29 The patients who showed nodal involve­ment associated to higher SUVmax had similar PFS to those presenting lower SUVmax values. As expected, the “protective value” of a low SUVmax fails when a lymph nodal involvement occurs; the quote of our patients with nodal involvement had a poor outcome irrespective of SUVmax values. It is well known, however, that, apart from distant metastases, positive mediastinal lymph nodes have the most significant prognostic value for recur­rence and death in NSCLC patients. Interestingly, the MTV acquired a prognostic significance in this sub-setting since it was slightly, but significantly associated with the outcome. This finding could be related to the impact of the amount of metaboli­cally active burden when multiple lesions are con­comitantly present, which better explains the risk of mediastinal nodal metastases.30 Some authors have already reported similar results but in more compromised, surgically treated patients whose nodal involvement was confirmed by histology and, not by PET/CT.29 Additionally, they described the incremental risk of developing nodal involve­ment due to MTV whereas in our setting it was re­lated to PFS. From a patho-physiological point of view, this finding is not surprising since SUVmax represents an index of cell glucose utilization and substan­tially reflects the first step of aerobic glycolysis, in­cluding cell uptake, whilst MTV better depicts the comprehensive metabolic tumour burden. Considering the reasonable natural history of the NSCLC, it could be envisaged that the persis­tent metabolism of glucose to lactate even in aero­bic conditions is an adaptation to intermittent hy­poxia in initial lesions, those supposed to be not yet metastatic (see SUVmax significance). However, once upregulation of glycolysis has taken place, cell populations acquire acid resistant phenotypes and powerful growth advantage. Such a condition is presumably thought to favour further, continu­ous hypoxia moving tumour and non-tumour cells automatically toward a type of anaerobic respira­tion. As a consequence, SUVmax depicting the sole hottest pixel within a tumour, might no longer indicate the whole tumour burden. On the other hand, the MTV, which embodies a volumetric rep­resentation of the metabolic charge, out of the more active pixel, seems to acquire a prognostic signifi­cance when tumour has dimensionally progressed and metastasized giving an incremental risk of event per unit increase. From a methodological point of view, it is con­ceivable that the lack of actually standardized cri­teria for determining MTV and TLG implies that these volumetric measurements might not repli­cate the absolute values produced elsewhere and continue to hold some intrinsic limits. However, several studies are being performed accounting for and validating their value. The prognostic impact of F-18 FDG PET/CT quantification parameters in non-surgical NSCLC patients remains to be completely established. In view of that, our results demonstrate, as a rule, that SUVmax is a better predictive indicator of progres­sion free survival than new volumetric parameters even if the MTV purchases a prognostic power in case of concomitant nodal metabolic concern. These findings endorse the idea that metabolic quantification parameters may be implemented in daily practice redirecting the concept that size (morphologically assessed) and histology consti­tute the only key-points for determining the bio­logical aggressiveness of lung cancer. This study included NSCLC patients that strictly fulfilled inclusion criteria (non-operable tumour ) and it was performed in a single centre, which gives reason for the relatively small samples size. Conclusions The quantitative assessment by F18-FDG PET/CT quantification parameters may be helpful to man­age inoperable NSCLC patients before chemother­apy. In this setting, the magnitude of glycolytic ac­tivity rather than the tumour burden extent seems to be generally predictive of response to subse­quent therapy apart from a loco-regional metastat­ic concern. 326 Acknowledgments The manuscript has been reviewed by a biostatis­tician. The authors wish to thank Mrs. A. Di Leo, Mrs. N. D’Errico and Mrs. A. Piccolella for their valuable contribution in performing the studies. References 1. AJCC cancer staging manual. 7th edition. Edge S, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, editors. New York: Springer Verlag; 2010. 2. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin 2010; 60: 277-300. 3. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, et al. Cancer incidence and mortality patterns in Europe: esti­mates for 40 countries in 2012. Eur J Cancer 2013; 49: 1374-403. 4. Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, et al. GLOBOCAN 2012 v1.0, Cancer incidence and mortality worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer; 2013. Available at: http://globocan.iarc.fr. 5. Schild SE, Tan AD, Wampfler JA, Ross HJ, Yang P, Sloan JA. A new scoring system for predicting survival in patients with non-small cell lung cancer. Cancer Med 2015; 4: 1334-43. 6. McCoach CE, Kelly K. Treatment paradigms in advanced non-small-cell lung cancer. Clin Adv Hematol Oncol. 2013; 11: 629-39. 7. Van Rens MT, de la Riviere AB, Elbers HR, van Den Bosch JM. Prognostic assessment of 2361 patients who underwent pulmonary resection for non-small cell lung cancer, stage I, II and IIIa. Chest 2000; 117: 374-79. 8. Socinski MA, Morris DE, Masters GA, Lilenbaum R; American College of Chest Physicians. Chemotherapeutic management of stage IV non-small cell lung cancer. Chest 2003; 123: 226S-43S. 9. Aydiner A, Sen F, Saglam EK, Oral EN, Eralp Y, Tas F, et al. Induction chemo­therapy with triweekly docetaxel and cisplatin followed by concomitant chemoradiotherapy with or without surgery in stage III non-small-cell lung cancer: a phase II study. Clin Lung Cancer. 2011; 12: 286-92. 10. Masters GA, Temin S, Azzoli CG, Giaccone G, Baker S Jr, Brahmer JR, et al. Systemic Therapy for Stage IV Non-Small-Cell Lung Cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol 2015; 33: 3488-515. 11. Zwitter M, Kovac V, Smrdel U, Vrankar M, Zadnik V. Gemcitabine in brief versus prolonged low-dose infusion, both combined with cisplatin, for advanced non-small cell lung cancer: a randomized phase II clinical trial. J Thorac Oncol 2009; 4: 1148-55. 12. Noonan K, Tong KM, Laskin J, Zheng YY, Melosky B, Sun S, et al. Evaluation of a ‘Watch and Wait’ Approach for Chemotherapy in Patients with Newly Diagnosed Advanced Non-small Cell Lung Cancer from a Diverse Community Population. Clin Oncol (R Coll Radiol) 2015; 27: 505-13. 13. Mac Manus MP, Hicks RJ, Matthews JP, McKenzie A, Rischin D, Salminen EK, et al. Positron emission tomography is superior to computed tomography scanning for response- assessment after radical radiotherapy or chemora­diotherapy in patients with non-small-cell lung cancer. J Clin Oncol 2003; 21: 1285-92. 14. Ryu JS, Choi NC, Fischman AJ, Lynch TJ, Mathisen DJ. FDG-PET in staging and restaging non-small cell lung cancer after neoadjuvant chemoradiotherapy: correlation with histopathology. Lung Cancer 2002; 35: 179-87. 15. Grgic A, Yüksel Y, Gröschel A, Schäfers HJ, Sybrecht GW, Kirsch CM, et al. Risk stratification of solitary pulmonary nodules by means of PET using (18) F-fluorodeoxyglucose and SUV quantification. Eur J Nucl Med Mol Imaging 2010; 37: 1087-94. 16. Zwitter M, Stanic K, Rajer M, Kern I, Vrankar M, Edelbaher N, et al. Intercalated chemotherapy and erlotinib for advanced NSCLC: high propor­tion of complete remissions and prolonged progression-free survival among patients with EGFR activating mutations. Radiol Oncol 2014; 48: 361-8. 17. Olivier A, Petyt G, Cortot A, Scherpereel A, Hossein-Foucher C. Higher predictive value of tumour and node [18F]-FDG PET metabolic volume and TLG in advanced lung cancer under chemotherapy. Nucl Med Commun 2014; 35: 908-15. 18. Hoang JK, Hoagland LF, Coleman RE, Coan AD, Herndon JE 2nd, Patz EF Jr. Prognostic value of fluorine-18 fluorodeoxyglucose positron emission tomography imaging in patients with advanced-stage non-small-cell lung carcinoma. J Clin Oncol 2008 20; 26: 1459-64. 19. Hashimoto Y, Tsujikawa T, Kondo C, Maki M, Momose M, Nagai A, et al. Accuracy of PET for diagnosis of solid pulmonary lesions with 18F-FDG uptake below the standardized uptake value of 2.5. J Nucl Med 2006; 47: 426-31. 20. Liao S, Penney BC, Wroblewski K, Zhang H, Simon CA, Kampalath R, et al. Prognostic value of metabolic tumor burden on 18F-FDG PET in nonsurgical patients with non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2012; 39: 27-38. 21. Lee P, Bazan JG, Lavori PW, Weerasuriya DK, Quon A, Le QT, et al. Metabolic tumor volume is an independent prognostic factor in patients treated definitively for non–small-cell lung cancer. Clin Lung Cancer 2012; 13: 52-8. 22. Xie P, Yue JB, Zhao HX, Sun XD, Kong L, Fu Z, et al. Prognostic value of 18F-FDG PET-CT metabolic index for nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2010; 136: 883-9. 23. Giorgetti A, Sorace O, Pisani P, Salvadori PA, Mariani G. Accuracy of Qualitative and Semiquantitative Analysis of 18FDG Positron Emission Tomography Scans in the Evaluation of Primary and Metastatic Lesions. Clin Positron Imaging 2000; 3: 182. 24. Zhang H, Berezov A, Wang Q , Zhang G, Drebin J, Murali R, et al. “ErbB receptors: from oncogenes to targeted cancer therapies”. J Clin Invest 2007; 117: 2051-8. 25. Ma BB, Hui EP, Mok TS. Population-based differences in treatment outcome following anticancer drug therapies. Lancet Oncol 2010; 11: 75-84. 26. Storto G, Nicolai E, Salvatore M. [18F]FDG-PET-CT for early monitoring of tu­mor response: when and why. Q J Nucl Med Mol Imaging 2009; 53: 167-80. 27. Berghmans T, Dusart M, Paesmans M, Hossein-Foucher C, Buvat I, Castaigne C, et al. European Lung Cancer Working Party for the IASLC Lung Cancer Staging Project. Primary tumor standardized uptake value (SUVmax) meas­ured on fluorodeoxyglucose positron emission tomography (FDG-PET) is of prognostic value for survival in non-small cell lung cancer (NSCLC): a systematic review and meta-analysis (MA) by the European Lung Cancer Working Party for the IASLC Lung Cancer Staging Project. J Thorac Oncol 2008; 3: 6-12. 28. Tong AN, Han SR, Yan P, Gong H, Zhao H, Yao H, et al. Prognostic value of FDG uptake in primary inoperable non-small cell lung cancer. Med Oncol 2014; 31: 780. 29. Lebioda A, Makarewicz R, Małkowski B, Dancewicz M, Kowalewski J, Windorbska W. Measurement of primary tumor volume by PET-CT to evalu­ate risk of mediastinal nodal involvement in NSCLC patients with clinically negative N2 lymph nodes. Rep Pract Oncol Radiother 2013; 18: 76-81. 30. Casali C, Cucca M, Rossi G, Barbieri F, Iacuzio L, Bagni B, et al. The variation of prognostic significance of Maximum Standardized Uptake Value of [18F]­fluoro-2-deoxy-glucose positron emission tomography in different histologi­cal subtypes and pathological stages of surgically resected Non-Small Cell Lung Carcinoma. Lung Cancer 2010; 69: 187-93. 327 research article Optimal scan time for evaluation of parathyroid adenoma with [18F]-fluorocholine PET/CT Sebastijan Rep1, Luka Lezaic1, Tomaz Kocjan2, Marija Pfeifer2, Mojca Jensterle Sever2, Urban Simoncic3, Petra Tomse1, Marko Hocevar4 1 Department for Nuclear Medicine, University Medical Centre Ljubljana, Slovenia 2 Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Slovenia 3 Jozef Stefan Institute, Ljubljana, Slovenia 4 Department of Oncological Surgery, Institute of Oncology Ljubljana, Ljubljana, Slovenia Radiol Oncol 2015; 49(4): 327-333. Received 6 January 2015 Accepted 31 March 2015 Correspondence to: Marko Hočevar, M.D., Ph.D., Department of Oncological Surgery, Institute of Oncology Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia. Phone: +386 1 5879 909; Fax: +386 1 5879 998; Email: mhocevar@onko-i.si Disclosure: No potential conflicts of interest were disclosed. Background. Parathyroid adenomas, the most common cause of primary hyperparathyroidism, are benign tumours which autonomously produce and secrete parathyroid hormone. [18F]-fluorocholine (FCH), PET marker of cellular pro­liferation, was recently demonstrated to accumulate in lesions representing enlarged parathyroid tissue; however, the optimal time to perform FCH PET/CT after FCH administration is not known. The aim of this study was to determine the optimal scan time of FCH PET/CT in patients with primary hyperparathyroidism. Patients and methods. 43 patients with primary hyperparathyroidism were enrolled in this study. A triple-phase PET/CT imaging was performed five minutes, one and two hours after the administration of FCH. Regions of interest (ROI) were placed in lesions representing enlarged parathyroid tissue and thyroid tissue. Standardized uptake value (SUVmean), retention index and lesion contrast for parathyroid and thyroid tissue were calculated. Results. Accumulation of FCH was higher in lesions representing enlarged parathyroid tissue in comparison to the thyroid tissue with significantly higher SUVmean in the second and in the third phase (p < 0.0001). Average retention index decreased significantly between the first and the second phase and increased significantly between the sec­ond and the third phase in lesions representing enlarged parathyroid tissue and decreased significantly over all three phases in thyroid tissue (p< 0.0001). The lesion contrast of lesions representing enlarged parathyroid tissue and thyroid tissue was significantly better in the second and the third phase compared to the first phase (p < 0.05). Conclusions. According to the results the optimal scan time of FCH PET/CT for localization of lesions representing enlarged parathyroid tissue is one hour after administration of the FCH. Key words: [18F]-fluorocholine PET/CT; lesions representing enlarged parathyroid tissue; triple-phase; standardized up­take value; retention index; lesion contrast Introduction Primary hyperparathyroidism is an endocrine disorder that develops as a result of autonomous production and secretion of parathyroid hormone (PTH) from parathyroid glands. The most com­mon cause of primary hyperparathyroidism is soli­tary adenoma (80–85%). Multi-glandular disease (multiple adenomas and parathyroid hyperplasia) is much rarer (15–20%).1 Parathyroid adenomas are benign monoclonal tumours that arise from neoplastic proliferation of a single abnormal cell. They are composed of main cells, oxyphil cells or a combination of both. In normally functioning para­thyroid glands only a small number of cells are in the growth phase while in primary hyperparathy­ 328 roidism the number of cells in the growth phase increases. The highest degree of proliferation is in parathyroid adenomas, followed by parathyroid gland hyperplasia.2,3 Symptomatic primary hyperparathyroidism is routinely treated with parathyroidectomy with a cure rate greater than 95% and a complication rate below 4%.4 Traditional surgical approach was a bilateral neck exploration with identification of all four parathyroid glands.5,6 Because most primary hyperparathyroidism cases can be attributed to a single adenoma7, a minimally invasive parathy­roidectomy with selective exploration and excision of only abnormally functioning parathyroid glands can be performed. The first minimally invasive parathyroidectomy was performed in 1996.8 Since then, the minimally invasive parathyroidectomy became a mainstay treatment for single adenoma with primary hyperparathyroidism, providing de­creased cost and patient discomfort9,10, and similar cure rates as a classic bilateral neck exploration.11 A prerequisite for successful minimally invasive par­athyroidectomy is accurate preoperative localiza­tion of lesions representing enlarged parathyroid tissue (LREPT). [99mTc]-sestaMIBI (MIBI) single-photon emission computed tomography/com­puted tomography (SPECT/CT)12 is a current gold standard for preoperative localization of LREPT with the sensitivity in the identification of a single adenoma of 80–90%.13-15 The ultrasonography that is often used in the preoperative or intraoperative setting as an adjunct for patients with negative MIBI scans, have the accuracy in identifying sin­gle adenomas of 70–80%.16,17 Both techniques have significantly lower accuracy for the detection of multi-glandular disease. Therefore, better imaging technique for preoperative localization of enlarged parathyroid glands is needed for wider acceptance of a minimally invasive parathyroidectomy. [11C]-choline and [18F]-fluorocholine (FCH), the PET markers of cellular proliferation, was inci­dentally demonstrated to accumulate in LREPT.18 Therefore; FCH was proposed for the preoperative localization in patients with a primary hyperpar­athyroidism.19 In a pilot study, our group was the first to demonstrate the effectiveness of FCH in the preoperative localization of LREPT in patients with a primary hyperparathyroidism. In a group of 24 patients the performance of FCH PET/CT was su­perior to standard MIBI SPECT/CT particularly in patients with multiglandular disease.20 There are very limited data in the literature on the tissue kinetics of FCH and additionaly they are all from studies in prostate cancer patients. The time course of FCH accumulation and release from LREPT and adjacent thyroid tissue has not yet been described. The aim of present study was to deter­mine the optimal scan time, i.e. time between ra­diopharmaceutical administration and FCH PET/ CT imaging in patients with a primary hyperpar­athyroidism. At the optimal scan time the highest values of lesion contrast should be expected, but also the radiopharmaceutical properties, such as administered activity and isotope half-life as well as particular department logistics, especially radi­opharmaceutical availability and scanner availabil­ity should be considered. Patients and methods From May 2012 to May 2014, FCHPET/CT triple-phase point imaging was performed in addition to conventional MIBI SPECT/CT and neck ultra­sound in 43 patients with a biochemically proven primary hyperparathyroidism (8 male and 35 fe­male, mean age 59.6 ± 11 years; range 36–77 years). All patients had increased levels of preoperative calcium (mean 2.8 mmol/l; range 2.6–4.1 mmol/l; normal range 2.1–2.6 mmol/l) and increased pre­operative iPTH levels (mean 311.5 ng/l; range 70.6– 2022 ng/l; normal range 10–65 ng/l). Patients with known history of malignant and/or inflammatory disease of the area of head and neck (other than autoimmune thyroid disease) were excluded from the study. All patients underwent surgery and had a histopathological examination of the removed parathyroid tissue. National medical ethics com­mittee approved the study and informed consent was obtained from all patients. After the administration of 100 MBq FCH (range: 96.8–104.5 MBq; mean 99.6 ± 2.2 MBq) the PET/CT imaging was performed at three time points: 5 min­utes (first phase), one hour (second phase) and two hours (third phase). An integrated PET/CT scanner (Biograph mCT, Siemens) was used. At all three time points of the imaging process the neck and upper mediastinum were scanned in a single bed position with a scan time of 4 minutes, the time-of­flight information capture being enabled. The pro­tocol included a low dose (120 kV; 25 mA) non-en­hanced CT scan of the neck and upper mediastinum for the attenuation correction, followed by 3D PET acquisition in the same anatomical area. The imag­es were reconstructed with iterative reconstruction with 2 iterations and 21 subsets, utilizing the scan­ner-specific point spread function. Data sets were reconstructed into standard 200×200×109 matrix 329 size using a 4×4×2 mm3 voxel size. A 3D Gaussian post-reconstruction filtration with 4 mm full-width at half maximum was applied. All the images were acquired at the Department of Nuclear Medicine, University Medical Centre Ljubljana. All FCH PET/CT images were masked and in­terpreted by two experienced observers on OASIS SEGAMI processing software. The image findings were scored for 5 different locations of LREPT: up­per right/left, lower right/left and ectopic. Focally increased uptake outside the normal FCH biodis­tribution was estimate as positive for LREPT. Scans of all three phases of FCH PET/CT were shown simultaneously on the monitor in the sag­ittal, transverse and coronary plane. Accumulated activity of FCH in LREPT was (semi)quantitative­ly valuated by placing a circular region of inter­est (ROI) adjusted to the metabolic volume of the gland. Maximum and mean standardized uptake values corrected for body weight (SUVmax and SUV) were obtained and SUV was used for meanmean analysis. In the thyroid gland, the circular ROI was positioned in an area in the lateral lobe without any thyroid pathology on metabolic and anatomi­cal (low dose CT) images. ROIs were copied and transferred to scans of all three time points, with automatic placement of the ROI in the appropri­ate location through automatic linking feature and manual correction if needed. Placement of ROIs was repeated four times to evaluate potential measurement error/dispersion. For patients positive in all three phases, SUVmean values were used to calculate the retention index (RI) and the lesion contrast (LC) – a quantitative measure obtained to evaluate the differential dy­namics of tracer uptake in LREPT and thyroid tis­sue in order to determine optimal FCH PET/CT scan time. RI determines the percentage variation of the standard uptake values in LREPT and thy­roid tissue and was compared between first-second and second-third phase.26 RI was calculated specifi­cally for LREPT and thyroid gland. RI was calcu­lated as: [1] where the RIt,p is retention index of LREPT (p) or thyroid tissue (t), and SUVmean is standardized up­take values in LREPT (p) or thyroid tissue (t) for corresponding phase.21 LC refers to the difference in the visual intensity of LREPT and thyroid gland in the image that cor­responds to different levels of radiopharmaceutical accumulation in these tissues. LC is important for TABLE 1. Comparison of binary classification test between all triple-phase FCH PET/ CT Sensitivity 90.5% 93.6% 93.6% 95.3% Specificity 98.2% 98.2% 98.2% 98.2% PPV 96.6% 96.7% 96.7% 96.8% NPV 94.7% 96.4% 96.4% 97.3% Accuracy 94.1% 96.5% 96.5% 97.0% the visual assessment of FCH PET/CT images and its higher value assists in the identification of ab­normalities, because the radiotracer accumulation in LREPT is higher in most cases, but may also be lower in comparison to surrounding tissue; in such cases, the lesion contrast is negative. LC was calcu­lated as: [2] where the LC is lesion contrast, PSUVmean is mean SUV in LREPT and TSUVmean is mean SUV in the thyroid tissue. LC was calculated for all three phas­es using the SUVmean of LREPT and thyroid tissue for corresponding phase.22 FCH PET/CT results were compared with his­topathological results, and sensitivity, specificity, positive predictive value (PPV), negative predic­tive value (NPV) and accuracy were calculated. SUVmean, RI and LC values are shown as average ± standard deviation (range). Group means were compared by two-tailed Student’s t-test for paired 330 TABLE 2. Comparison of average SUVmean LREPT and thyroid tissue LREPT Thyroid tissue p 5.29 ± 2.29 (1.9% to 11.8%) 4.48 ± 1.55 (2.5% to 7.9%) 0.03 4.69 ± 2.31 (1.6% to 11.0%) 3.15 ± 1.11 (1.8% to 5.8%) <0.0001 4.77 ± 2.39 (1.6% to 11.5%) 3.04 ± 1.13 (1.8% to 5.7%) <0.0001 of the FCH PET/CT were higher in the second and third phase compared to the first phase (Table 1). Triple-phase PET/CT images showed a different distribution of FCH in the LREPT in comparison to the thyroid tissue (Figure 1, Table 2). On average, SUVmean in LREPT was highest in the first phase, and then decreased significantly in the second phase (p < 0.0001), and increased non-significant­ly in the third phase (p = 0.2). Average SUVmean in thyroid tissue was also highest in the first phase, then decreased significantly in the second (p < 0.0001) and the third phase (p = 0.009). The differ­ence of the average SUVmean between the LREPT and background thyroid tissues was significant in the second and the third phase. Figure 2 shows RI between the first and the second phase and be­tween the second and the third phase. Average RI decreased significantly in LREPT between the first and the second phase, and increased significantly between the second and the third phase (both at p < 0.0001) (Table 3). Observed LC of LREPT and thyroid tissue was 31.5% ± 60.8% in the first phase, 70.4% ± 95.5% in the second phase and 75.6% ± 121.5% in the third phase (Table 4).The difference in LC was statisti­cally significant between the first and the second phase (p = 0.012) and between the first and the third phase (p = 0.015). The positive LC had values of up to 600% and the maximum negative LC had a value of 52%. Distribution of number of lesions along the ranges of LC values in all three phases is presented in Figure 3a. Additionally, in Figure 3b, the number of both – the positive and the negative - contrast lesions having the absolute LC greater than the selected value is presented. Discussion MIBI SPECT/CT is the current gold standard for preoperative localization of LREPT in patients with a primary hyperparathyroidism with sen­sitivity in the identification of a single gland dis­ease of 80-90%.13-15 However, in the case of multi-glandular disease the diagnostic performance of MIBI SPECT/CT is significantly lower.23 Therefore, better imaging techniques for preoperative locali­zation of LREPT are being searched. Among dif­ferent radiopharmaceuticals tested for PET scan in preoperative diagnosis of a primary hyperpar­athyroidism, [11C]-methionine is the most common one. However, the results of reported studies were not convincing enough to replace MIBI SPECT/ CT.24-26 The first report of FCH accumulation in or unpaired data, as appropriate. A p-value < 0.05 was considered statistically significant. Statistical analysis was performed with the use of the SPSS software (version 16). Results Sixty lesions of enlarged parathyroid tissue were localized by the FCH PET/CT scanning 43 patients. A primary hyperparathyroidism resolved and se­rum calcium normalized in 40/43 patients after surgery in which 60 parathyroid glands were re­moved (1.4 parathyroid gland/patient). According to histopathological diagnosis there were 34 soli­tary adenomas, one double adenoma, one cancer and hyperplasia in 7 patients (2/7 patients had a combination of primary and secondary hyperpar­athyroidism). Sensitivity, PPV, NPV and accuracy 331 parathyroid adenomas and hyperplasia was based on incidental findings of Quak and Mapelli in pa­tients with prostate cancer using FCH and [11C]­choline.18-19 It was our group that published the first study of comparison between FCH PET/CT and MIBI SPECT/CT and concluded that the FCH PET/CT is an accurate, efficient imaging modality for localization of hyper functioning parathyroid tissue, particularly in patients with multi glandu­lar disease, where by its diagnostic performance is superior to the standard MIBI SPECT/CT.20 Better spatial resolution and LC are most probably re­sponsible for higher sensitivity of FCH PET/CT. The aim of our present study was to determine the optimal FCH uptake period that maximizes tumor-to-normal-tissue activity ratio. In order to determine the optimal scan time, SUVmean, RI and LC in LREPT and thyroid tissue were measured at three time points - 5 minutes (first phase), one hour (second phase) and two hours (third phase) after the administration of FCH (Figure 4). Average accumulation of FCH was higher in LREPT in comparison to thyroid tissue in all three phases. However, the statistically signifi­cant difference in tracer uptake between LREPT and thyroid tissue, as assessed by SUVmean, was only found in the second and the third phase; in these two phases, LC was also significantly higher in comparison to the first phase. Additionally, in comparison to LREPT a higher SUVmean in the thy­roid tissue was found in more than a third (18/57; LREPT positive in all three phases) of lesions (8 solitary adenomas and 10 multi-glandular diseas­es). An underlying thyroid disease might be an ex­planation for a higher accumulation of FCH in the thyroid tissue. Three of these patients indeed had autoimmune thyroiditis; unfortunately, we did not have clinical data on thyroid disease status in the rest of these patients, but there was no known his­tory of thyroid disease. The highest SUVmean value in LREPT was achieved shortly after the FCH administration and decreased gradually between the first and the sec­ond phase in the majority of lesions. Surprisingly, in approximately half of these lesions a slight in­crease of SUVmean value in LREPT was observed between the second and the third phase. There is very limited data in the literature on the kinetics of FCH – all these studies include prostate cancer patients. Giussani et al.27 have developed a model of FCH kinetics based on biodistribution measure­ments that describes recirculation of radiopharma­ceutical from major organs of early uptake (liver, spleen, kidneys) back into the blood pool, which A B FIGURE 3. (A) The number of lesions in ranges of lesion contrast (LC) values for all three phases; for both positive and negative LC. (B) The number of both positive and negative lesions having absolute LC value equal or greater to the value on horizontal axis. TABLE 3. Comparison of average RI in LREPT and thyroid tissue Average RI Tissue between first and second between second and third phase phase -11.1% ± 18.5% 1,7% ± 10.2% LREPT (-54.4% to 26.3%) (-22.8% to 22.2%) -29.8% ± 12.8% -4.5% ± 8.4% Thyroid tissue (-51.7% to 3.8%) (-26.3% to 1.1%) p <0.0001 0.001 TABLE 4. Lesion contrast calculation and temporal comparison 5 min 1 h 2 h 31,1% ± 60.8% 70.4% ± 95.5% 75.6% ± 121.5% LC (-52% to 217.7%) (-44.8% to 410.5%) (-40.3% to 592.8%) Between first and Between second Between first second phase and third phase and third phase Lesion contrast 0.012 0.8 0.015 comparison (p) 332 FIGURE 4. A secluded lower left LREPT. FCH PET/CT was performed in triple-phase after administration of 100 MBq of FCH. The LREPT is well delineated according to the thyroid tissue on PET axial image in the first (A1), the second (A2) and the third phase (A3). SUVmean in the LREPT and the thyroid tissue was in the first phase 6.3 and 3.6, in the second phase 7.1 and 3, and in the third phase 6.6 and 2.9. The CT axial image of the LREPT and the thyroid tissue in all three phases (B1, B2, B3), and the FCH PET/ CT axial fusion image in all three phases (C1, C2, C3). may provide an explanation for late SUV increase in the parathyroid tissue. Tavola et al.28 concluded in their study that the simple linear model cannot adequately describe the kinetics of FCH, due to non-linear kinetics, which is associated with the re­lease of FCH from the organs back into the blood. The non-linear kinetic model caused a slight over­estimation of the activity in the liver and kidneys, most probably due to a physiological activity. In addition to a higher accumulation of FCH in most LREPT in all three phases, in comparison to the thyroid tissue, there was also a slower (efflux) release of FCH from the LREPT, reflected by the highest LC between LREPT and thyroid tissue in the third phase. However, the difference in LC be­tween the second and the third phase is not statis­tically significant, allowing PET/CT investigations to be performed from one to two hours after the administration of the radiopharmaceutical. Since the daily radiopharmaceutical dose was delivered to the department in the mornings, and its activ­ity diminished relatively fast due to the 110 min­utes half-life of 18F29, earlier scanning times were prefered. Therefore we are suggestting the optimal scan time of one hour with 4 minutes acquisition time. Such scanning protocol did allow us to im­age up to 12 patients with a primary hyperpar­athyroidism, each with small activities (100 MBq) administration of the radiopharmaceutical. Despite the generally higher LC between LREPT and thyroid tissue in the second and the third phase, there was a single patient in our study with the uptake only in the first phase. In this patient’s case, an intense accumulation was present in the bone marrow in the second and the third phase, while almost no activity could be perceived in LREPT and thyroid tissue. A possible explanation for this unusual situation could be polycythemia ru­bra vera, which the patient was treated for. In order to avoid negative test results, early imaging might also be recommended in patients with potentially extensive hypermetabolic tissues, such as haema­tological and other malignancies. Due to poor contrast between LREPT and thy­roid gland in the first phase, three lesions were not localized, while in the second and the third phase the LC improved due to rapid wash out of FCH from the thyroid tissue and all three lesions were correctly localized. In one patient with hyperplasia two lesions were correctly localized as double ade­nomas, while two were false positive – histological results showed they were lymph nodes. Conclusions Preoperative localization of parathyroid glands in patients with a primary hyperparathyroidism is possible with FCH PET/CT imaging. Optimal imag­ing time is one hour after the administration of FCH. Due to rare comorbidities, lesion uptake may be present exclusively in the early phase (immediately after administration) therefore if logistically possi­ble, early phase imaging is recommended as well. References 1. Lew JI, Solorzano CC. Surgical management of primary hyperparathy­roidism: state of the art. Surg Clin North Am 2009; 89: 1205-25. 2. Wada Y, Kunimura T, Sato S, Hisayuki T, Sato M, Imataka H, et al. Proliferating potential and apoptosis in the development of secondary hyperpar­athyroidism: a study based on Ki-67 immunohistochemical staining and the terminal dUTP nick-end labeling assay. Ther Apher Dial 2008; 12: 319-28. 3. Yamaguchi S, Yachiku S, Morikawa M. Analysis of proliferative activity of the parathyroid glands using proliferating cell nuclear antigen in patients with hyperparathyroidism. J Clin Endocrinol Metab 1997; 82: 2681-8. 333 4. Allendorf J, DiGorgi M, Spanknebel K, Inabnet W, Chabot J, Logerfo P. 1112 consecutive bilateral neck explorations for primary hyperparathyroidism. World J Surg 2007; 31: 2075-80. 5. Dowthwaite SA, Young JE, Pasternak JD, Yoo J. Surgical management of primary hyperparathyroidism. J Clin Densitom 2013; 16: 48-53. 6. Wang CA. Surgical management of primary hyperparathyroidism. Curr Probl Surg 1985; 22: 1-50. 7. Ruda JM, Hollenbeak CS, Stack BC, Jr. A systematic review of the diagno­sis and treatment of primary hyperparathyroidism from 1995 to 2003. Otolaryngol Head Neck Surg 2005; 132: 359-72. 8. Miccoli P, Pinchera A, Cecchini G, Conte M, Bendinelli C, Vignali E, et al. Minimally invasive, video-assisted parathyroid surgery for primary hyper­parathyroidism. J Endocrinol Invest 1997; 20: 429-30. 9. Goldstein RE, Blevins L, Delbeke D, Martin WH. Effect of minimally invasive radioguided parathyroidectomy on efficacy, length of stay, and costs in the management of primary hyperparathyroidism. Ann Surg 2000; 231: 732-42. 10. Sosa JA, Udelsman R. Minimally invasive parathyroidectomy. Surg Oncol 2003; 12: 125-34. 11. Grant CS, Thompson G, Farley D, van Heerden J. Primary hyperparathy­roidism surgical management since the introduction of minimally invasive parathyroidectomy: Mayo Clinic experience. Arch Surg 2005; 140: 472-479. 12. Coakley AJ, Kettle AG, Wells CP, O’Doherty MJ, Collins RE. 99mTc sestamibi­-a new agent for parathyroid imaging. Nucl Med Commun 1989; 10: 791-4. 13. Borley NR, Collins RE, O’Doherty M, Coakley A. Technetium-99m sestamibi parathyroid localization is accurate enough for scan-directed unilateral neck exploration. Br J Surg 1996; 83: 989-91. 14. Carneiro-Pla DM, Solorzano CC, Irvin GL, 3rd. Consequences of targeted parathyroidectomy guided by localization studies without intraoperative parathyroid hormone monitoring. J Am Coll Surg 2006; 202: 715-22. 15. Lavely WC, Goetze S, Friedman KP, Leal JP, Zhang Z, Garret-Mayer E, et al. Comparison of SPECT/CT, SPECT, and planar imaging with single- and dual-phase (99m)Tc-sestamibi parathyroid scintigraphy. J Nucl Med 2007; 48: 1084-9. 16. Berri RN, Lloyd LR. Detection of parathyroid adenoma in patients with pri­mary hyperparathyroidism: the use of office-based ultrasound in preopera­tive localization. Am J Surg 2006; 191: 311-4. 17. Haber RS, Kim CK, Inabnet WB. Ultrasonography for preoperative localiza­tion of enlarged parathyroid glands in primary hyperparathyroidism: com­parison with (99m)technetium sestamibi scintigraphy. Clin Endocrinol (Oxf) 2002; 57: 241-9. 18. Mapelli P, Busnardo E, Magnani P, Freschi M, Picchio M, Gianolli L, et al. Incidental finding of parathyroid adenoma with 11C-choline PET/CT. Clin Nucl Med 2012; 37: 593-5. 19. Quak E, Lheureux S, Reznik Y, Bardet S, Aide N. F18-choline, a novel PET tracer for parathyroid adenoma? J Clin Endocrinol Metab 2013; 98: 3111-2. 20. Lezaic L, Rep S, Jensterle SM, Hocevar M, Fettich J. 18F-fluorocholine PET/ CT for localization of hyper functioning parathyroid tissue in primary hyper­parathyroidism: a pilot study. Eur J Nucl Med Mol Imaging 2014; 41: 2083-9. 21. Nakayama M, Okizaki A, IshitoyaS, Sakaguchi M, Sato J, Aburano T. Dual­time-point F-18 FDG PET/CT imaging for differentiating the lymph nodes between malignant lymphoma and benign lesions. Ann Nucl Med 2013; 27: 163-9. 22. Cherry SR, Sorenson JA, Phelps ME. Physics in nuclear medicine. 4th edition Philadelphia: Elsevier/Saunders; 2012. p. 253-270. 23. Chien D, Jacene H. Imaging of parathyroid glands. Otolaryngol Clin North Am 2010; 43: 399-415. 24. Hellman P, Ahlström H, Bergström M, Sundin A, Langström B, Westerberg G, et al. Positron emission tomography with 11C-methionine in hyperparathy­roidism. Surgery 1994; 116: 974-81. 25. Otto D, Boerner AR, Hofmann M, Brunkhorst T, Meyer GJ, Petrich T, et al. Pre-operative localisation of hyperfunctional parathyroid tissue with 11C-methionine PET. Eur J Nucl Med Mol Imaging 2004; 31: 1405-12. 26. Oksüz MO, Dittmann H, Wicke C, Müssig K, Bares R, Pfannenberg C, et al. Accuracy of parathyroid imaging: a comparison of planar scintigraphy, SPECT, SPECT/CT, and C-11 methionine PET for the detection of parathyroid adenomas and glandular hyperplasia. Diagn Interv Radiol 2011; 17: 297-307. 27. Giussani A, Janzen T, Uusijarvi-Lizana H, Tavola F, Zankl M, Sydoff M, et al. A compartmental model for biokinetics and dosimetry of 18F-choline in prostate cancer patients. J Nucl Med 2012; 53: 985-93. 28. Tavola F, Janzen T, Giussani A, Facchinetti D, Veronese I, Uusijärvi-Lizana H, et al. Nonlinear compartmental model of 18F-choline. Nucl Med Biol 2012; 39: 261-8. 29. DeGrado TR, Coleman RE, Wang S, Baldwin SW, Orr MD, Robertson CN, et al. Synthesis and evaluation of 18F-labeled choline as an oncologic tracer for positron emission tomography: initial findings in prostate cancer. Cancer Res 2001; 61: 110-7. 334 research article Endobronchial ultrasound elastography strain ratio for mediastinal lymph node diagnosis Ales Rozman1, Mateja Marc Malovrh1, Katja Adamic1, Tjasa Subic1, Viljem Kovac2, Matjaz Flezar1 1 University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia 2 Institute of Oncology Ljubljana, Ljubljana, Slovenia Radiol Oncol 2015; 49(4): 334-340. Received 16 March 2015 Accepted 30 March 2015 Correspondence to: Assist. Prof. Matjaž Fležar, M.D., Ph.D., University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik 36, 4204 Golnik, Slovenia. Fax: +386 4 256 9117; Email: matjaz.flezar@klinika-golnik.si Disclosure: No potential conflicts of interest were disclosed. Background. Ultrasound elastography is an imaging procedure that can assess the biomechanical characteristics of different tissues. The aim of this study was to define the diagnostic value of the endobronchial ultrasound (EBUS) elastography strain ratio of mediastinal lymph nodes in patients with a suspicion of lung cancer. The diagnostic values of the strain ratios were compared with the EBUS brightness mode (B-mode) features of selected mediastinal lymph nodes and with their cytological diagnoses. Patients and methods. This prospective, single-centre study enrolled patients with an indication for biopsy and me­diastinal staging after a non-invasive diagnostic workup of a lung tumour. EBUS with standard B-mode evaluation and elastography with strain ratio measurement were performed before endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA). Results. Thirty-three patients with 80 suspicious mediastinal lymph nodes were included. Malignant infiltration was confirmed in 34 (42.5%) lymph nodes. The area under the receiver operating characteristic curve for the strain ratio was 0.87 (p < 0.0001). At a strain ratio . 8, the accuracy for malignancy prediction was 86.25% (sensitivity 88.24%, speci­ficity 84.78%, positive predictive value [PPV] 81.08%, negative predictive value [NPV] 90.70%). The strain ratio is more accurate than conventional B-mode EBUS modalities for differentiating between malignant and benign lymph nodes. Conclusions. EBUS-guided elastography with strain ratio assessment can distinguish malignant from benign medi­astinal lymph nodes with greater accuracy than conventional EBUS modalities. This new method may reduce the number of mediastinal EBUS-TBNAs and thus reduce the invasiveness and expense of mediastinal staging in patients with non-small lung cancer (NSCLC). Key words: cancer staging; elastography; endobronchial ultrasound; lung cancer; needle biopsy Introduction Mediastinal lymph node staging is essential for optimal treatment decisions in patients with non-small cell lung cancer (NSCLC) who do not have distant metastases.1,2 Current NSCLC guidelines recommend endosonographically guided needle biopsy of mediastinal lymph nodes as a reliable first-choice method, reducing the need for more invasive surgical staging.1 A combination of en­dobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) and endoscopic ultrasound-guided fine-needle aspiration (EUS­FNA) can define the mediastinal lymph node stage with a sensitivity of 91% and a negative predictive value (NPV) of 96%.1 Mediastinal lymph nodes can also be non-invasively characterised by conven­tional brightness mode (B-mode) EBUS.2,3 Power/ colour Doppler-mode image analysis of vascular patterns of lymph nodes can be helpful in predict­ing metastatic infiltration during the EBUS-TBNA procedure.4 Ultrasound elastography is an imaging proce­dure that can assess the biomechanical character­ 335 istics of different tissues and their deformation un­der compression.5 Malignant tissues are generally stiffer than native healthy tissues and can therefore be distinguished on the basis of decreased elastic­ity. Elastographic image analysis is based on quali­tative pattern analysis and/or on semi-quantitative histogram analysis. New EBUS software supports visualisation of the tissue elasticity modulus by colour-coding tissue deformability: the hardest tissues are shown in blue, intermediate tissues in green and soft tissues in red. Moreover, it is pos­sible to quantify the strain in two operator-selected areas. Comparing these different areas of tissue al­lows a numeric representation of the strain ratio between the two areas. Ultrasound elastography has previously been applied as an external procedure (for example, in the diagnosis of thyroid, breast and prostate tu­mours) and as an endoscopic procedure (for ex­ample, in the diagnosis of pancreatic tumours, and nodal involvement of rectal and oesophageal can­cer ).6-13 A meta-analysis of EUS elastography tri­als on the differentiation of benign and malignant lymph nodes reported a sensitivity of 88%, a speci­ficity of 85% and an area under the receiver operat­ing characteristic (ROC) curve of 0.95.14 However, only one of the included trials evaluated the strain ratio as a diagnostic standard.15 A recent prelimi­nary report on endobronchial ultrasound elastog­raphy suggests that this method may improve di­agnostic yield.16 The aim of this pilot study was to define for the first time the diagnostic value of the EBUS elastog­raphy strain ratio for mediastinal lymph node stag­ing in patients who were suspected to have lung cancer. Diagnostic values for the strain ratios were compared with EBUS B-mode features of selected mediastinal lymph nodes and with cytological di­agnosis. Patients and methods Patients This prospective single-centre study was con­ducted between August and December 2013. Evaluations were performed on consenting con­secutive patients who were at least 18 years old and been referred for bronchoscopy with a suspi­cion of lung cancer according to a chest CT scan. Eligible patients had enlarged but discrete N2/N3 lymph nodes, a centrally located tumour with nor-mal-sized mediastinal lymph nodes, or enlarged N1 lymph nodes with normal-sized mediastinal lymph nodes. Exclusion criteria were metastatic disease, severe co-morbidity that disqualified sur­gical treatment, mediastinal tumour infiltration, and small peripheral lung tumours with normal-sized mediastinal lymph nodes. Written informed consent was obtained from each patient prior to bronchoscopy. The study was approved by the National Medical Ethics Committee and was registered at ClinicalTrials. gov under the clinical trial number NCT02009319. Instruments and procedure All bronchoscopy procedures were performed under deep sedation that was carried out by an anaesthesiologist. EBUS procedures were per­formed with two BF-UC180F linear ultrasound bronchoscopes (Olympus Tokyo, Japan). Real-time EBUS B-mode and elastography with strain ratio measurements were performed using a pre­production model of the Endoscopic Ultrasound Center EU-Y0008 (Olympus Tokyo, Japan), which was later commercialised under the name EU-ME2 Premier Plus after minor modifications. Mediastinal lymph nodes were initially evalu­ated by B-mode EBUS as in Fujiwara et al. for ana­tomical location, size, shape, border distinction, echogenicity, central hilar structure and coagula­ Rozman A et al. / Elastography in mediastinal lymph nodes 336 EBUS = endobronchial ultrasound FIGURE 2. Flow diagram of lymph node confirmation. tion necrosis.3 A size of greater than 10 mm, round shape, distinct margin, heterogeneous echogenic­ity, absence of central echogenic hilum and coagu­lation necrosis were considered to be signs of ma­lignant infiltration of the lymph node. The region of interest (ROI) for the elastographic evaluation was selected using a trackball, avoid­ing large vessels because structures with very low elasticity might induce artefacts in the evaluation of stiffness distribution.13 The elastography pattern as the result of tissue compression was produced by vascular pulsations and respiratory movement and not by direct bronchoscope pressure on the bronchial wall. After obtaining an artefact-free im­age, the “freeze” function was used, and the larg­est possible area of the lymph node was outlined to determine the strain. As a reference, an area of normal-appearing soft tissue from the surrounding mediastinum was selected to determine the strain ratio by EBUS processor unit (Figure 1). We select­ed tissue between the lymph node and bronchial cartilage or lateral to the lymph node. The strain ratio was measured twice for each selected lymph node. Whenever possible, the EBUS bronchoscope was inserted into the oesophagus to determine the strain ratio of the same lymph nodes to assess the influence of tracheal cartilage on the transtracheal measurements. After non-invasive evaluation, EBUS-TBNA was performed using a 22-gauge needle. The patholo­gist who performed the cytological analysis was blinded to the strain ratio and other EBUS B-mode features. Definitive diagnosis EBUS-TBNA was performed at least twice per lymph node. Malignancy, where present, was ac­cepted as a definitive diagnosis. Patients with a be­nign outcome from EBUS-TBNA (i.e., lymphocytes) and proven cancer were sent for surgical treatment with lymph node dissection and histological ex­amination. In cases where malignant disease was excluded, those patients were followed up meticu­lously until a benign outcome for the course of the disease was also confirmed. Data analysis Data are presented as frequencies, ranges, means ± SD and percentages. The strain ratio for each lymph node was measured twice, and the mean of both measurements was accepted for further anal­ysis. Sensitivity, specificity, accuracy, positive pre­dictive values (PPVs), and NPVs were calculated. ROC analysis was performed to show the specific­ity/sensitivity for different strain ratio cut-off val­ues. The area under the ROC curve was calculated. The optimal cut-off value for the strain ratio was se­lected at the point with the highest sensitivity and specificity. A paired t-test was used to analyse tra­cheal and oesophageal measurements of the strain ratio. The analysis was performed using GraphPad Prism version 5.00 (GraphPad Software, San Diego, California, USA; www.graphpad.com). Results Patients and lymph nodes EBUS elastography was performed on 80 lymph nodes at different lymph node stations in 33 pa­tients (25 male and 8 female) with an average age of 67.5 (± 8.2) years. Twenty-seven patients had a final diagnosis of lung cancer (14 adenocarcinoma, 8 squamous cell carcinoma, 3 small cell carcinoma and 2 non-small cell carcinoma); six patients had benign diagnoses at the end of the study (2 post­tuberculotic changes, 2 pneumonia/lung abscesses, one sarcoidosis and 1 infectious mononucleosis). Cytological specimens were adequate from 75 (93.7%) lymph nodes and non-representative from five (6.3%) lymph nodes. Malignant infiltration was confirmed in 34 (42.5%) lymph nodes, primar­ily by EBUS-TBNA (sensitivity 97.1%, NPV 97.6%; Figure 2). The average size of the evaluated lymph nodes was 11.1 (± 4.8) mm, with a range of 4–26 mm. Radiol Oncol 2015; 49(4): 334-340. 338 TABLE 1. Sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) and accuracy of each endobronchial ultrasound image category for metastatic lymph nodes in comparison to strain ratio (%) Sensitivity 67.65 76.47 79.41 67.65 88.24 41.18 88.24 Specificity 78.26 78.26 56.52 84.78 69.57 91.30 84.78 PPV 69.70 72.22 57.44 76.67 68.18 77.78 81.08 NPV 76.60 81.82 78.79 78.00 88.89 67.74 90.70 Accuracy 73.75 77.50 66.25 77.50 77.50 70.00 86.25 CHS = central hilar structure; CNS = coagulation necrosis sign FIGURE 5. Sensitivity and specificity decision plot to determine the optimal cut-off for strain ratio. Curves cross at the strain ratio value 8. Eleven (28.21%) of 39 normal-sized lymph nodes sampled in patients with malignant disease were metastatic. The statistical data for the strain ratios in the group of normal-sized lymph nodes were as follows: acc. 89.74%, sensitivity 90.91%, specificity 89.29%, PPV 76.92%, and NPV 96.15%. Discussion The strain ratio determined by endobronchial ul­trasound elastography seems to be a promising new method for differentiating between malig­nant and benign lymph nodes in patients with lung cancer. This study confirmed the feasibility of elastography and strain ratio analysis using an EBUS bronchoscope in mediastinal lymph nodes. Moreover, the initial results showed a higher accu­racy in the differentiation of malignant and benign lymph nodes than the conventional non-invasive EBUS modalities evaluated in this study. The most accurate methods currently available for mediastinal staging in NSCLC patients are FNA under ultrasonic guidance and mediastinos­copy.1 These methods require tissue sampling and disrupt the integrity of mediastinal structures, car­rying the risk of complications such as bleeding, infection and pneumothorax.17 Although rapid on-site evaluation (ROSE) can reduce the number of punctures, many patients are nonetheless exposed to numerous sampling procedures at several dif­ferent lymph node stations and to subsequent mediastinoscopy.18,19 Less invasive techniques for accurate mediastinal lymph node diagnosis are therefore desirable for optimal mediastinal staging. Cervical mediastinoscopy, which was the gold­en standard for mediastinal staging, seems to have limited utility in the patients with T1 and T2 clini­cally staged N0 by positron emission tomography -computed tomography (PET-CT).20 However, in case of CT-enlarged or PET-positive mediastinal lymph nodes, tissue confirmation is indicated.2 Endosonography (EBUS/EUS) with fine needle as­piration is still the first choice since it is minimally invasive and has a high sensitivity to rule in me­diastinal nodal disease. If negative, surgical stag­ing with nodal dissection or biopsy is indicated. Video-assisted mediastinoscopy is preferred over mediastinoscopy.21 The elastography strain ratio as a semi-quantita­tive method has been evaluated as part of an endo­scopic procedure in gastroenterology, and favour-able results have been reported for mediastinal lymph node analysis in patients with oesophageal cancer.15 The conclusions from the staging of upper 340 vessels in the ROI, this was not entirely possible in all cases. The study design had several limitations. The first limitation was that this was a single-centre, single-operator scheme, and thus no inter-observ­er variations were taken into account. The gold standard for the determination of malignant in­volvement is a combination of several methods: a positive EBUS-TBNA result (assuming that there were no false-positive results), lymph node dis­section during surgery (with station evaluation rather than direct node-to-node comparison) and a final benign diagnosis in the remaining patients. Because the strain ratio was determined from “fro­zen” EBUS images, selection bias might be created during static image selection, especially during the selection of the reference area in the strain ratio evaluation. In conclusion, this study shows that EBUS-guided elastography with strain ratio assessment can distinguish malignant and benign mediastinal lymph nodes with greater accuracy than conven­tional EBUS modalities. The high NPV for normal-sized mediastinal lymph nodes in lung cancer patients is comparable to the NPV obtained with EBUS-TBNA. This new method may potentially reduce the number of mediastinal EBUS-TBNAs and thus reduce the invasiveness and expense of mediastinal staging in NSCLC patients. Further multicentre trials are needed to confirm these pre­liminary results. References 1. Silvestri GA, Gonzalez AV, Jantz MA, Margolis ML, Gould MK, Tanoue LT, et al. Methods for staging non-small cell lung cancer: Diagnosis and manage­ment of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2013; 143(5 Suppl): e211S-50S. 2. Cistaro A, Quartuccio N, Mojtahedi A, Fania P, Filosso PL, Campenni A, et al. Prediction of 2 years-survival in patients with stage I and II non-small cell lung cancer utilizing 18F-FDG PET/CT SUV quantification. Radiol Oncol 2013; 47: 219-23. 3. Fujiwara T, Yasufuku K, Nakajima T, Chiyo M, Yoshida S, Suzuki M, et al. The utility of sonographic features during endobronchial ultrasound-guided transbronchial needle aspiration for lymph node staging in patients with lung cancer: a standard endobronchial ultrasound image classification sys­tem. Chest 2010; 138: 641-7. 4. Nakajima T, Anayama T, Shingyoji M, Kimura H, Yoshino I, Yasufuku K. Vascular image patterns of lymph nodes for the prediction of metastatic disease during EBUS-TBNA for mediastinal staging of lung cancer. J Thoracic Oncol 2012; 7: 1009-14. 5. Saftoiu A, Vilman P. Endoscopic ultrasound elastography - a new im­aging technique for the visualization of tissue elasticity distribution. J Gastrointestin Liver Dis 2006; 15: 161-5. 6. Barr RG, Destounis S, Lackey LB 2nd, Svensson WE, Balleyguier C, Smith C. Evaluation of breast lesions using sonographic elasticity imaging: a multi­center trial. J Ultrasound Med 2012; 31: 281-7. 7. Brock M, von Bodman C, Palisaar RJ, Löppenberg B, Sommerer F, Deix T, et al. The impact of real-time elastography guiding a systematic prostate bi­opsy to improve cancer detection rate: a prospective study of 353 patients. J Urol 2012; 187: 2039-43. 8. Lyshchik A, Higashi T, Asato R, Tanaka S, Ito J, Mai JJ, et al. Thyroid gland tumor diagnosis at US elastography. Radiology 2005; 237: 202-11. 9. Giovannini M, Hookey LC, Bories E, Pesenti C, Monges G, Delpero JR. Endoscopic ultrasound elastography: the first step towards virtual biopsy? Preliminary results in 49 patients. Endoscopy 2006; 38: 344-8. 10. Saftoiu A, Vilmann P, Hassan H, Gorunescu F. Analysis of endoscopic ultra­sound elastography used for characterisation and differentiation of benign and malignant lymph nodes. Ultraschall Med 2006; 27: 535-42. 11. Giovannini M, Thomas B, Erwan B, Christian P, Fabrice C, Benjamin E, et al. Endoscopic ultrasound elastography for evaluation of lymph nodes and pancreatic masses: a multicenter study. World J Gastroenterol 2009; 15: 1587-93. 12. Janssen J, Dietrich CF, Will U, Greiner L. Endosonographic elastography in the diagnosis of mediastinal lymph nodes. Endoscopy 2007; 39: 952-7. 13. Saftoiu A, Vilmann P, Ciurea T, Popescu GL, Iordache A, Hassan H, et al. Dynamic analysis of EUS used for the differentiation of benign and malig­nant lymph nodes. Gastrointest Endosc 2007; 66: 291-300. 14. Xu W, Shi J, Zeng X, Li X, Xie WF, Guo J, et al. EUS elastography for the differentiation of benign and malignant lymph nodes: a meta-analysis. Gastrointest Endosc 2011; 74: 1001-9. 15. Paterson S, Duthie F, Stanley A. Endoscopic ultrasound-guided elastography in the nodal staging of oesophageal cancer. World J Gastroenterol 2012; 18: 889-95. 16. Trosini-Desert V, Jeny F, Taillade L, Vignot S, Zribi H, Capron F, et al. Bronchial endoscopic ultrasound elastography: preliminary feasibility data. Eur Respir J 2013; 41: 477-9. 17. Asano F, Aoe M, Ohsaki Y, Okada Y, Sasada S, Sato S, et al. Complications as­sociated with endobronchial ultrasound-guided transbronchial needle aspi­ration: a nationwide survey by the Japan Society for Respiratory Endoscopy. Respir Res 2013; 14: 50. 18. Lee HS, Lee GK, Lee HS, Kim MS, Lee JM, Kim HY, et al. Real-time endobron­chial ultrasound-guided transbronchial needle aspiration in mediastinal staging of non-small cell lung cancer: how many aspirations per target lymph node station? Chest 2008; 134: 368-74. 19. Trisolini R, Cancellieri A, Tinelli C, Paioli D, Scudeller L, Casadei GP, Parri et al. Rapid on-site evaluation of transbronchial aspirates in the diagnosis of hilar and mediastinal adenopathy: a randomized trial. Chest 2011; 139: 395-401. 20. Fernandez FG, Kozower BD, Crabtree TD, Force SD, Lau C, Pickens A, et al. Utility of mediastinoscopy in clinical stage I lung cancers at risk for occult mediastinal nodal metastases. J Thorac Cardiovasc Surg 2015; 149: 35-41. 21. De Leyn P, Dooms C, Kuzdzal J, Lardinois D, Passlick B, Rami-Porta R, et al. Preoperative mediastinal lymph node staging for non-small cell lung cancer: 2014 update of the 2007 ESTS guidelines. Transl Lung Cancer Res 2014; 3: 225-33. 22. Larsen MH, Fristrup C, Hansen TP, Hovendal CP, Mortensen MB. Endoscopic ultrasound, endoscopic sonoelastography, and strain ratio evaluation of lymph nodes with histology as gold standard. Endoscopy 2012; 44: 759-66. 341 research article Analysis of risk factors for perifocal oedema after endovascular embolization of unruptured intracranial arterial aneurysms Snezana Lukic1, Slobodan Jankovic2, Katarina Surlan Popovic3, Dragic Bankovic4, Peter Popovic3, Milan Mijailovic1 1 Department for Interventional Neuroradiology, Clinical Center, Kragujevac, University of Kragujevac, Serbia 2 Department for Clinical Pharmacology, Clinical Center, Kragujevac, Serbia 3 Clinical Radiology Institute, University Medical Centre Ljubljana, Ljubljana, Slovenia 4 Faculty of Natural Sciences and Mathematics, University of Kragujevac, Serbia Radiol Oncol 2015; 49(4): 341-346. Received 18 February 2015 Accepted 28 September 2015 Correspondence to: Prof. Slobodan Janković, M.D., Ph.D., Prim., Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia. E-mail: slobnera@gmail.com Disclosure: No potential conflicts of interest were disclosed. Background. Endovascular embolization is a treatment of choice for the management of unruptured intracranial aneurysms, but sometimes is complicated with perianeurysmal oedema. The aim of our study was to establish inci­dence and outcomes of perianeurysmal oedema after endovascular coiling of unruptured intracranial aneurysms, and to reveal possible risk factors for development of this potentially serious complication. Methods. In total 119 adult patients with endovascular embolization of unruptured intracranial aneurysm (performed at Department for Interventional Neuroradiology, Clinical Center, Kragujevac, Serbia) were included in our study. The embolizations were made by electrolite-detachable platinum coils: pure platinum, hydrophilic and combination of platinum and hydrophilic coils. Primary outcome variable was perianeurysmal oedema visualized by magnetic reso­nance imaging (MRI) 7, 30 and 90 days after the embolization. Results. The perianurysmal oedema appeared in 47.6% of patients treated with hydrophilic coils, in 21.6% of patients treated with platinum coils, and in 53.8% of those treated with mixed type of the coils. The multivariate logistic regres­sion showed that variables associated with occurrence of perianeurysmal oedema are volume of the aneurysm, hypertension, diabetes and smoking habit. Hypertension is the most important independent predictor of the perianeu­rysmal oedema, followed by smoking and diabetes. Conclusions. The results of our study suggest that older patients with larger unruptured intracranial aneurysms, who suffer from diabetes mellitus and hypertension, and have the smoking habit, are under much higher risk of having perianeurysmal oedema after endovascular coiling. Key words: intracranial aneurysms; endovascular embolization; perianeurysmal oedema; hypertension Introduction During the past decade method of endovascular embolization became a treatment of choice for the management of unruptured intracranial aneu­rysms, which are increasingly being diagnosed in the era of modern imaging methods. Endovascular embolization has been validated as a minimally invasive and effective treatment that can prevent rupture of an aneurysm and intracranial haemor­rhage, with shortening of hospital stay and faster patient recovery compared to the surgical treat­ment. Further step in development of this thera­peutic procedure was design of bioactive coils, which are very effective, but sometimes followed by perianeurysmal oedema. There are a few pub­ 342 lished case reports linking these coils with devel­opment of the perianeurysmal edema1-7, but we are still far away from complete understanding of this phenomenon. Almost identical post embolization reactions could be seen after use of bare platinum coils8,9, and it is not clear whether the per aneurys­mal oedema is an adverse reaction to the coils it­self, or just to specific type(s) of the coils. The perianeurysmal oedema could clinically present itself with headache, lethargy, confusion, meningismus, seizures, disorders of visual func­tion and/or cranial nerve(s) palsy.1,2,4,5,7 Majority of the reported perianeurysmal oedema cases ap­peared after coiling large aneurysms6, but oede­ma was also recorded after embolization of small aneurysms.9 From the cases described so far, we could conclude that the clinical appearance, the time frame of oedema occurrence and the final pa­tient outcome are highly variable. Perianeurysmal oedema is relatively rare com­plication that follows embolization of intracranial aneurysms. Most of the patients with this compli­cation are clinically inconspicuous, asymptomatic or with nonspecific symptoms, which leads to un­derestimation of the incidence and importance of this complication.5 We are currently unaware of any risk factors that could predetermine appear­ance or severity of the perianeurysmal oedema af­ter endovascular coiling.10-13 The aim of our study was to establish incidence and outcomes of perianeurysmal oedema after en­dovascular coiling of unruptured intracranial an­eurysms, and to reveal possible risk factors for de­velopment of this potentially serious complication. Material and methods Study design Our study was designed as case series, with an em­bedded case-control study. The case series consist­ed of patients who underwent endovascular em­bolization of unruptured intracranial aneurysm; those of them who developed perianeurysmal oedema were then considered to be the „cases“, and the rest of them were classified as „controls“. The cases and controls were not matched. The study was approved by the Clinical Center Kragujevac Medical Ethics Committee (No. 01/8642) and was carried out according to the Declaration of Helsinki as well as we have fol­lowed the relevant guidelines in this investigation. All patients signed informed consent. 343 Study population In total 119 patients (of both sex and older than 18 years) with endovascular embolization of unrup­tured intracranial aneurysm were included in our study. The embolizations were performed by expe­rienced interventional neuroradilogists (more than 200 performed embolizations each) at Department for Interventional Neuroradiology, Clinical Center, Kragujevac, Serbia, from January 2008 to December 2012. The embolizations were made by etachable platinum coils: pure platinum, hydrophilic and combination of platinum and hydrophilic coils. In order to prevent extrusion of the coils from aneu­rysms with broad neck, self-expanding stents were used. Study variables Primary outcome variable was perianeurysmal oedema visualized by magnetic resonance imag­ing (MRI) imaging 7, 30 and 90 days after the em­bolization, using standard T1 weighted (W), T2W and fluid attenuated inversion recovery (FLAIR) sequences (Figures 1–4). There are predictor vari­ables that were followed in the study: age of the patients, sex, arterial blood pressure, serum cho­lesterol level, size and location of the aneurysm, type of the coils, patient medication and smoking habits. Statistical analysis The study data were primarily described using medians, means and standard deviations for con­tinuous variables, and percentages and odds for categorical variables. Normality of the data dis­tribution was checked by Kolgomorov-Smirnof test. The differences in values of predictor vari­ables among the study groups (patients with and without perianeurysmal oedema) were tested for significance by Student’s T-test (for continuous variables) or by Chi-square test (for categorical variables). Univariate and multivariate logistic regression analyses were used to study the rela­tionship between occurrence of perianeurysmal oedema and the predictor variables. The differ­ences among the study groups were considered to be significant if probability of null hypothesis was less than 0.05. Lukic S et al. / Perifocal oedema after endovascular embolization 344 TABLE 1. The patients’ characteristics Results Age 46.30 ± 8.26* 56.87 ± 9.33* ***0.0005 Gender (F) 26 (35.1) 20 (44.4) 0.414 Hypertension 43 (58.1) 44 (97.8) ***0.0005 Diabetes 8 (10.8) 20 (44.4) ***0.0005 Smoking 39 (52.7) 34 (75.6) 0.022 Hypercholesterolemia 19 (25.7) 30 (66.7) ***0.0005 Corticosteroids before the embolization 29 (39.2) 13 (28.9) 0.346 Volume of an aneurysm 75 (33 – 154)** 518 (215 – 898)** ***0.0005 The coil type: ***0.006 - hydrophilic 22 (52.4%) 20 (47.6%) - platinum 40 (78.4%) 11 (21.6%) - mixed 12 (46.2%) 14 (53.8%) * = mean ± SD, ** = median (25-th percentile – 75-percentile); *** = significant difference categorical variables shown as n (%); FIGURE 5. The receiver-operator curve (ROC) for volume of the intracranial aneurysms and occurrence of the perianeurysmal oedema after endovascular embolization. The perianeurysmal oedema was observed by NMR imaging in 45 of 119 patients (37.8%). However, only 8 patients (6.7%) developed symp­tomatic oedema, with transient headache and malaise. The symptoms withdrew spontaneously in the next 48 hours. The characteristics of the patients with and without the perianeurysmal oedema are shown in Table 1. Location of the an­eurysms was similar in the both groups (p = 0.268), but type of the coils determined occurrence rate of the perianeurysmal oedema: it appeared in 47.6% of patients treated with hydrophilic coils, in 21.6% of patients treated with platinum coils, and in 53.8% of those treated with mixed type of the coils. However, the only significant difference in rate of perianurismal oedema was between the patients treated with platinum coils and the pa­tients treated with mixed type of coils. All of the patients with symptomatic oedema were treated with mixed type coils. From the Table 1 one could see that the higher the volume of an aneurysm, the perianeurysmal oedema is more frequent. If a Receiver-Operator Curve (ROC) is constructed for volume of the an­eurysms and occurrence of the oedema after endo­vascular embolization (Figure 5), it turns out that growth of volume of aneurysm for 1 mm3 increases risk of perianeurysmal oedema for 1.3% (AUROC = 0.908, p = 0.0005). If a cut-off value of 174 mm3 is taken into account, the volume of an aneurysm could predict occurrence of perianeurysmal oede­ma with sensitivity of 80.08%, and specificity of 83.8% (positive predictive value is 75.0%, and neg­ative predictive value is 87.3%). The results of univariate and multivariate bi­nary logistic regression for occurrence of perianu­rysmal oedema as dependent variable are shown in Table 2. After adjustment for other followed variables, the multivariate regression showed that variables associated with occurrence of peri- TABLE 2. Binary logistic regression for occurrence of perianeurysmal oedema as dependent variable Age 0.0005 1.146 (1.085 – 1.211) Hypertension 0.001 31.721 (4.144 – 242.784) 0.022 30.599 (1.624 – 576.504) Smoking 0.015 2.774 (1.223 – 6.291) 0.026 5.391 (1.226 – 23.710) Diabetes 0.0005 6.600 (2.577 – 16.901) 0.039 5.336 (1.091 – 26.099) Hyperholesterolemia 0.0005 5.789 (2.575 – 13.015) Volume of the aneurysm 0.0005 1.010 (1.006 – 1.015) 0.0005 1.013 (1.006 – 1.019) Radiol Oncol 2015; 49(4): 341-346. 345 aneurysmal oedema are volume of the aneu­rysm, hypertension, diabetes and smoking habit. Hypertension is the most important independent predictor of the perianeurysmal oedema (30-fold increased risk), followed by smoking and diabetes (about 5-fold increased risk with each of the vari­ables). Discussion Most of previous reports on perianeurysmal oede­ma after endovascular coiling described patients with developed clinical picture.4,5,9 However, peri­aneurysmal oedema occurs also in asymptomatic and patients with mild non-specific symptoms, so frequency and importance of this phenomenon could be underestimated. According to scarce published data, the oedema could be found in 9% of asymptomatic patients3, but our data showed much higher rate of 37.8%. Considering recently found link between occurrence of perianeurysmal oedema after endovascular embolization and re­currence of anurysm14, it becomes very important to identify risk factors for the oedema and to modify them with an aim to prevent its occur­rence. Mechanism of perianeurysmal oedema devel­opment remains unclear for the time being, but it seems that inflammation plays very important role. Several mechanisms have been proposed to explain perianeurysmal oedema. It may represent a normal healing response after coiling and is prob­ably related by the inflammatory changes. This in­flammatory response can be exaggerated in some cases, leading to peryaneurysmal oedema. In other cases, oedema is observed several months after an­eurysms treatment and is generally associated with massive recurrence. Oedema can develop also im­mediately after treatment due to thrombus forma­tion and secondary expansion of aneurysmal sac. It was described around both coiled and uncoiled aneurysms, but the uncoiled ones were invariably thrombosed. If the aneurysm was not thrombosed completely, continuous water-hammering effect against the residual lumen of the aneurysm (an­eurysm pulsing) could cause haemorrhage in the aneurysmal wall, triggering inflammation.15 This happens mostly in larger aneurysms with wide neck, which explains why in our study the aneu­rysms with larger volume were more frequently followed with perifocal oedema after emboliza­tion. Moreover, larger aneurysms have bigger en­dothelial surface, which when damaged by coils and blood jet produces larger quantity of autacoids that can initiate process of inflammation.1 As a confirmation of these hypotheses, both release of cytokines to cerebrospinal liquor13 and inflamma­tion of meninges12 were found in patients with per­ianeurysmal oedema after endovascular coiling. However, although clinical improvement of peri­aneurysmal oedema was demonstrated in some studies12,13, others did not confirm such beneficial effect.15 In our study pre-embolization administra­tion of corticosteroids did not affect the occurrence of perianeurysmal oedema. Older patients have higher risk for perianeu­rysmal oedema after embolization6,8,9,10; the pa­tients with oedema in our study were on average 10 years older than the patients without oedema. Hypertension, diabetes mellitus and smoking are also risk factors for perianeurysmal oedema, ac­cording to our and some other studies.6 This is not surprising, since all these factors adversely affect microcirculation16,17 leading to increased perme­ability of capillaries and propensity for inflamma­tion and oedema formation. Perianeurysmal oedema could be observed af­ter endovascular embolization with any type of coils.1-11 However, certain studies hypothesized that bioactive coils increase the risk for the oedema up to nine-fold.8,10,12 In our study bare platinum coils did have the smallest rate of perianeurysmal oedema, but the difference was not significant in comparison to the hydrophilic coils. This issue re­quires further research before any kind of recom­mendation about choice of coil type could be made. According to published data the incidence of perianeurysmal oedema is much lower, from a few percentages to 14, 3%.18,19 However all published cases were symptomatic, presented commonly with headache, malaise or cranial neuropathies. Perianeurysmal oedema is actually much more frequent, but mostly remains asymptomatic (as shown in our study), and is rarely followed by seri­ous complications. The results of our study suggest that older patients with larger unruptured intracranial an­eurysms, who suffer from diabetes mellitus and hypertension, and have the smoking habit, are under much higher risk of having perianeurysmal oedema after endovascular coiling. Such patients should be more strictly controlled by MRI imag­ing after the embolization, in order to reveal the oedema earlier, and adjust the care for the patient accordingly. 346 References 1. Pickett GE, Laitt RD, Herwadkar A, Hughes DG. Visual pathway compromise after hydrocoil treatment of large ophthalmic aneurysms. Neurosurgery 2007; 61: E873-4. 2. Schmidt GW, Oster SF, Golnik KC, Tumialán LM, Biousse V, Turbin R, et al. Isolated progressive visual loss after coiling of paraclinoid aneurysms. Am J Neuroradiol 2007; 28: 1882-9. 3. Fanning NF, Willinsky RA, Brugge KG. Wall enhancement, edema, and hydrocephalus after endovascular coil occlusion of intradural cerebral an­eurysms. J Neurosurg 2008; 108: 1074-86. 4. Cohen JE, Itshayek E, Attia M, Moscovici S. Postembolization perianeurys­mal edema as a cause of uncinate seizures. J Clin Neurosci 2012; 19: 474-6. 5. Vu Dang L, Aggour M, Thiriaux A, Kadziolka K, Pierot L. Post-embolization perianeurysmal edema revealed by temporal lobe epilepsy in a case of un­ruptured internal carotid artery aneurysm treated with bare platinum coils. J Neuroradiol 2009; 36: 298-300. 6. Horie N, Kitagawa N, Morikawa M, Tsutsumi K, Kaminogo M, Nagata I. Progressive perianeurysmal edema induced after endovascular coil em­bolization. Report of three cases and review of the literature. J Neurosurg 2007; 106: 916-20. 7. Stracke CP, Krings T, Möller-Hartmann W, Mahdavi A, Klug N. Severe inflam­matory reaction of the optic system after endovascular treatment of a supraophthalmic aneurysm with bioactive coils. Am J Neuroradiol 2007; 28: 1401-2. 8. White JB, Cloft HJ, Kallmes DF. But did you use HydroCoil? Perianeurysmal edema and hydrocephalus with bare platinum coils. Am J Neuroradiol 2008; 29: 299-300. 9. Craven I, Patel UJ, Gibson A, Coley SC. Symptomatic perianeurysmal edema following bare platinum embolization of a small unruptured cerebral aneu­rysm. Am J Neuroradiol 2009; 30: 1998-2000. 10. Killer M, Hauser T, Wenger A, Richling B, Ladurner G. Comparison of experi­mental aneurysms embolized with second-generation embolic devices and platinum coils. Acta Neurochir (Wien) 2009; 151: 497-505. 11. Nishino K, Ito Y, Hasegawa H, Shimbo J, Kikuchi B, Fujii Y. Development of cranial nerve palsy shortly after endosaccular embolization for asympto­matic cerebral aneurysm: report of two cases and literature review. Acta Neurochir (Wien) 2009; 151: 379-83. 12. Im SH, Han MH, Kwon BJ, Jung C, Kim JE, Han DH. Aseptic meningitis after embolization of cerebral aneurysms using hydrogel-coated coils: report of three cases. Am J Neuroradiol 2007; 28: 511-2. 13. Killer M, Arthur A, Al-Schameri AR, Barr J, Elbert D, Ladurner G, et al. Cytokine and growth factor concentration in cerebrospinal fluid from patients with hydrocephalus following endovascular embolization of un­ruptured aneurysms in comparison with other types of hydrocephalus. Neurochem Res 2010; 35: 1652-8. 14. Hayashi K, Kitagawa N, Morikawa M, Horie N, Kawakubo J, Hiu T, et al. Long-term follow-up of endovascular coil embolization for cerebral aneurysms using three-dimensional time-of-flight magnetic resonance angiography. Neurol Res 2009; 31: 674-80. 15. Tomokiyo M, Kazekawa K, Onizuka M, Aikawa H, Tsutsumi M, Ikoh M, et al. Mechanisms of perianeurysmal edema following endovascular emboliza­tion of aneurysms. Interv Neuroradiol 2007; 13(Suppl 1): 145-50. 16. Del Canizo Gómez FJ, Fernández Pérez C, Moreno Ruiz I, de Gorospe Pérez-Jáuregui C, Silveira Rodríguez B, González Losada T, et al. Microvascular complications and risk factors in patients with type 2 diabetes. Endocrinol Nutr 2011; 58: 163-8. 17. Mancini M, Di Donato O, Saldalamacchia G, Liuzzi R, Rivellese A, Salvatore M. Contrast-enhanced ultrasound evaluation of peripheral microcirculation in diabetic patients: effects of cigarette smoking. Radiol Med 2013; 118: 206-14. 18. Im SH, Han MH, Kwon BJ, Jung C, Kim JE, Han DH. A meningitis after embo­lization of cerebral aneurysms using hydrogelcoated coils: report of three cases. Am J Neuroradiol 2007; 28: 511-2. 19. Fanning NF, Wilingsky RA, Grugge KG. Wall enhancement, edema and hydrocephalus after endovascular coil occlusion on intradural cerebral aneurysms. J Neurosurgery 2008; 108: 1074-86. 347 research article Estimation of cell response in fractionation radiotherapy using different methods derived from linear quadratic model Safoora Nikzad1, Bijan Hashemi2, Golshan Mahmoudi3, Milad Baradaran-Ghahfarokhi4,5 1 Department of Medical Physics, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran 2 Department of Medical Physics, Tarbiat Modares University, Tehran, Iran 3 Department of Medical Physics, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran 4 Department of Medical Physics and Medical Engineering & Medical Student’s Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran 5 Department of Medical Radiation Engineering, Faculty of Advanced Sciences & Technologies, Isfahan University, Isfahan, Iran Radiol Oncol 2015; 49(4): 347-356. Received 5 May 2015 Accepted 12 August 2015 Correspondence to: Golshan Mahmoudi, Department of Medical Physics, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran. Phone: +98 913 803 9773; E-mail: golshan.mahmoudi@yahoo.com. Milad Baradaran-Ghahfarokhi, Medical Physics and Medical Engineering Department, Isfahan University of Medical Sciences (IUMS), Isfahan 81746-73461, Iran. Phone: +98 311 7922432; Fax: +98 311 6688597; E-mail: milad_bgh@yahoo.com Disclosure: No potential conflicts of interest were disclosed. Background. The aim of this study was to use various theoretical methods derived from the Linear Quadratic (LQ) model to calculate the effects of number of subfractions, time intervals between subfractions, dose per subfraction, and overall fraction time on the cells’ survival. Comparison of the results with experimental outcomes of melanoma and breast adenocarcinoma cells was also performed. Finally, the best matched method with experimental out­comes is introduced as the most accurate method in predicting the cell response. Materials and methods. The most widely used theoretical methods in the literature, presented by Keall et al., Brenner, and Mu et al., were used to calculate the cells’ survival following radiotherapy with different treatment schemes. The overall treatment times were ranged from 15 to 240 minutes. To investigate the effects of number of subfractions and dose per subfraction, the cells’ survival after different treatment delivery scenarios were calculated through fixed overall treatment times of 30, 60 and 240 minutes. The experimental tests were done for dose of 4 Gy. The results were compared with those of the theoretical outcomes. Results. The most affective parameter on the cells’ survival was the overall treatment time. However, the number of subfractions per fractions was another effecting parameter in the theoretical models. This parameter showed no significant effect on the cells’ survival in experimental schemes. The variations in number of subfractions per each frac­tion showed different results on the cells’ survival, calculated by Keall et al. and Brenner methods (P<0.05). Conclusions. Mu et al. method can predict the cells’ survival following fractionation radiotherapy more accurately than the other models. Using Mu et al. method, as an accurate and simple method to predict the cell response after fractionation radiotherapy, is suggested for clinical applications. Key words: fractionation radiotherapy; survival; dose per fraction; number of fractions; linear quadratic model Introduction Radiotherapy is one of the main procedures of cancer treatment. The goal of radiotherapy is to de­liver as much dose to the tumor site while keeping the dose to the surrounding normal tissues as low as possible.1, 2 In radiotherapy, in addition to the conventional techniques used in clinical practice, some state of the art specialized techniques such as Intensity Modulated Radiation Therapy (IMRT), 348 Respiratory-Gated, stereotactic, and Image Guide Radiotherapy (IGRT) have also been developed.3-7 These modern techniques optimize the radio­therapy dose distribution since they include more segments in the radiation field which are usu­ally shaped using more complicated equipment.3-7 These techniques enhance tumor local control and have lower radiation-induced toxicities in normal organs around the tumor compared to conven­tional techniques. Moreover, they vary in the dose delivery due to using more subfractions per each treatment fraction, different treatment times be­tween subfractions, and the prolonged treatment time of one fraction.4,8-15 The radiobiological efficiency of these tech­niques might be different from conventional one mainly due to the repair of sublethal damages.8-16 However, the rate and the mechanism of repair is a complicated function of different parameters such as dose per fraction, dose rate, repairs half time, and state and nature of the organs of interest (i.e. ./ß ratio of the organ).8-16 To predict the results of different radiation de­livery procedures on the cells’ survival, the basic theoretical model is the incomplete repair model of Thames17 generalized to multiple fractions by Nilsson et al.18 that is a developed form of Linear Quadratic (LQ) model. Some studies have inves­tigated the effects of prolonged time of radiation delivery on the survival of some cell lines and com­pared the results with theoretical methods derived from the LQ model.8-15 Although these theoretical methods are all de­rived from the basic LQ model, however, the rate of agreement between their results in researches and experiments was significantly different for di­verse dose schedules.8,9,19-21 Therefore, more investigations are needed in order to evaluate the effect of various treatment factors on the cells’ survival. In addition, it seems beneficial to compare the results of these methods theoretically and experimentally in order to find the best method that can be used to predict the cells’ survival after different fractionation radio­therapy schemes. The aim of this study was to compare various theoretical methods widely used in the litera­ture8,9,19-21 to estimate the effects of number of sub-fractions, time intervals between subfractions, dose per subfraction, and overall fraction time on the F10B16 skin melanoma and 4T1 breast adenocar­cinoma cells’ survival. Comparison of the results with experimental outcomes of melanoma and breast adenocarcinoma cells was also performed. Moreover, in this work, the best matched method with experimental outcomes is introduced as the most accurate one in predicting the cell response in fractionation radiotherapy. Materials and methods Theoretical methods Three methods of calculation derived from LQ model, presented by Keall et al., Brenner, and Mu et al.8,9,19-21, were compared to investigate the effect of different dose schemes (dose per subfraction, time intervals between subfractions, total treatment time of each fraction) on the survival of F10B16 skin melanoma and 4T1 breast adenocarcinoma cells. The basic idea of these methods is based on the completed LQ model as: [1] Which is a developed form of the basic LQ mod­el: [2] Where . and ß are cell parameters, D is the total dose delivered to the cells, S is the survival fraction of cells, and G parameter is defined in intermittent radiotherapy to investigate the effect of subfrac­tions. The G parameter has been formulated differ­ently by various investigators.8,9,19-21 The first method (method I) was presented by Keall et al.9 They have experimentally and theoreti­cally investigated the temporal effects of respirato­ry-gated and IMRT treatment delivery for dose of 2 Gy and in the total treatment times of 1.67 min (in conformal radiotherapy) to 15 min (in gated IMRT) on the cells’ survival. Keall et al. have used a simplified form of G to predict the cells’ survival and have compared the outcomes with experi­mental results.9 They have assumed negligible cell proliferation and unchanging radiosensitivity.9 According to Keall et al. study, the G parameter is calculated as9: [3] [4] In this method, µ is the rate constant for repair of sublethal damages, n is the number of subfrac­ 349 tions, . is the time of exposure and .t is the time between subfractions. This method assumes a constant value for both exposure time (t) and the time between exposures (.t).9 Keall et al. results showed no significant difference between the ex­perimental observations and theoretical calcula­tions.9 Moreover, this method indicated a good agreement with experimental results for the total dose of 2 Gy.9 The second method (method II) was utilized by Brenner.20 This method was also proposed in some review papers.19,21 Brenner has simplified the LQ model and experimentally and theoretically inves­tigated the temporal effects of fractionation treat­ment delivery on in vitro survival.20 In Brenner method, the G factor accounts for fraction protection and acts on the quadratic com­ponent as follow20: [5] In this method, the used parameters are the same as Keall et al. method.9 As this formula (equation 5) shows, the effects of time intervals between sub-fractions are ignored, however, Brenner has con­firmed that there was a good agreement between the outcomes of this formula and the experimental results.20 Therefore, it has been proposed that, this formula can be used to calculate the cell response after prolonged treatment delivery.20 In addition, this method can be employed to calculate the pro­traction effects in a single fractionation delivered at a constant rate, splitting dose, multi-fraction ir­radiation protocols and continuous low dose rates radiotherapy such as brachytherapy.20 The third theoretical method (method III) was reported by Mu et al.8 In Mu et al. study, the G pa­rameter is defined as below8: [6] All the used parameters in this method are ex­plained above. In this method, it is assumed that there is no recovery during actual irradiations but rather during the time between subfactions.8 Cell culture and assay The cells were cultured in plastic flasks at 37°C in a humidified atmosphere of 50 mL/L CO2 and 95% air with the RPMI1640 medium containing 10-15% fetal calf serum (FCS or FBS) with 100 U/mL peni­cillin and 100 µg/mL streptomycin. TABLE 1. The F10B16 and 4T1 cell parameters as input data for the used models . (Gy-1) Linear parameter of LQ model 0.0956 0.0424 ß (Gy-2) Quadratic parameter of LQ model 0.0177 0.0399 T1/2 (hour) Half time of sublethal repair 0.524±0.035 0.344±0.015 Due to tree shaped structure of these cell lines, complexity of counting their colonies, and sig­nificant number of samples used in this study, an automated and faster assay method was used. Therefore, instead of the clonogenic assay, the mul­ti 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetra­zoliumbromide (MTT) assay was used. This method was offered in other similar researches22-24 and all the details of experimental procedure are published in papers by our team for these two cell lines (F10B16 melanoma and 4T1 breast adenocar­cinoma) of interest.25,26 Theoretical schemes In this paper, . and ß parameters were calculated using the basic LQ model (equation 2). Hence, the cell survival fractions (S) following doses of 2, 4, 6, 8 and 10 Gy were experimentally determined for both melanoma and breast adenocarcinoma cells and then inserted in the basic formula of the LQ model. Using the S and D parameters and insert­ing them in the mentioned formula, the survival curves of cell lines were drawn and . and ß param­eters were derived using the MATLAB software (Version 7.11, R2010b, MathWorks, USA).8,25 In order to determine the time constant for re­pair of sublethal damage (T1/2), the cells were ex­posed in two fractions with different time intervals between the fractions. Then, the surviving fraction was plotted against the time between fractions and finally the half value of sublethal damage repair was investigated.8,25 All the cell’s parameters for both cell lines of in­terest, used in this study, are illustrated in Table 1. Different treatment schemes were designed in order to investigate the effects of the most im­portant radiobiological parameters including; the number of subfractions, time intervals between subfractions, subfraction doses, and overall treat­ment time, in complex radiotherapy practices. To investigate the effect of total treatment time, the survival fraction (SF) were calculated for dose of 2, 4 and 6 Gy in two subfractions of 1, 2 and 3 Gy, respectively. The overall treatment times were 350 ranged from 15 to 240 minutes. Although, total treatment time in complicated radiotherapy is about 1 hour and longer treatment time is not prac­tical, however, we followed the investigations for up to 4 hours to determine comprehensive results and investigate the ability of the developed models to predict the cells’ survival. To investigate the effects of increasing the num­ber of subfractions and dose per subfraction, the survival was calculated for total dose of 2, 4 and 6 Gy in 4 and 8 subfractions as follow: 4 fractions of 0.5 Gy and 8 fractions of 0.25 Gy (both for a total dose of 2 Gy), 4 fractions of 1 Gy and 8 fractions of 0.5 Gy (both for a total dose of 4 Gy) and 4 fractions of 1.25 Gy and 8 fractions of 0.75 Gy (both for a to­tal dose of 6 Gy). They all delivered through fixed overall treatment times of 30, 60 and 240 minutes. It should be noted that the theoretical methods presented by Keall et al., Brenner, and Mu et al. can be used in predicting survival in fractionation radi­otherapy and some of them have flaw in predicting survival when the dose is delivered continuously in one fraction.8,9 Therefore, in this work, the basic LQ model (equation 2) was used to predict the cell survival following continuous dose delivery. Experimental schemes The cells were picked out from the flasks when they reached to linear phase of exponential grow in the day before irradiation and were put in 96 well plates with density of 1000 cells in each well.22-25 There were 7 samples for each experiment and, to avoid the variability inherent to the assay used, all tests were performed for 3 independent experi­ ments. A Co-60 source with a dose rate of 0.81 Gy/ min was used for irradiation. The ionizing radia­tion was delivered in a 25×25 cm2 field size. All ir­radiations were performed at a distance of 20 cm between the radiation sources and plate. To measure the absorbed dose rate of the Cobalt-60 beam, a Farmer-type ionization chamber with a standard 60Co buildup cap, and positioned in air using a customized stand, was used. “For traceability to international standards, the ioniza­tion chamber was calibrated in comparison with the response of the Secondary Standard Dosimetry Laboratory (SSDL, Karaj Complex, Atomic Energy Organization of Iran) reference and working stand­ard ionization chambers in the 60Co gamma ray beam of a teletherapy unit. All of the SSDL ioniza­tion chambers used for calibrations are themselves calibrated at the International Atomic Energy Agency (IAEA) dosimetry laboratory”.27 To design the experimental tests, firstly, contin­uous radiation with doses of 2, 4 and 6 Gy, similar to conventional radiotherapy techniques, were de­livered to the cells. Next, to investigate the effect of overall treatment time on the cells’ survival, the same as the theoretical schedules, 6 groups from both of the studied cell lines were exposed to 2, 4 and 6 Gy in two subfractions with dose of 1, 2 and 3 Gy, respectively. In this step, the overall treatment time was 15 to 240 minutes. Then, to simulate the effects of the number of subfractions as well as dose per subfraction, 4 and 8 subfractions with dose of 1 and 0.5 Gy, respectively (total dose of 4 Gy), were delivered to the cells at overall treatment times of 30, 60 and 240 minute. After that, the results were compared with those of continuous radiation. It should be noted that, although the conven­tional treatment dose used in clinical situation is approximately 2 Gy per fraction8,9, however, the 351 effect of this low level of dose on the cell culture environment was negligible for the two cell lines of interest (Figure 1), and consequently the dose of 4 Gy was used in this experiment. Statistical analysis Statistical analysis was performed using the SPSS software version 14 (SPSS, Inc., Chicago, IL, USA). To assess the effects of different irradiation proto­ cols, the analysis of variance (ANOVA) was used. A significant level of 0.05 was considered to the tests. Results Figure 1 illustrates the survival curves for the two cells of interest as well as the calculated . and ß parameters. Figures 2, gives the survival of the cells in continuous radiation with dose of 2, 4 and 6 Gy and also in fractionation delivery in two subfrac­tions of 1, 2 and 3 Gy, during the overall treatment time of 15 to 240 minute. Figures 3 to 5, show the predicted survival using the theoretical methods of Keall et al., Brenner, and Mu et al., as well as the experimental results. For a total dose of 2 Gy and all irradiation times in both two cell lines of inter­est (4T1 and F10B16), there was no significant dif-For a dose of 4 Gy, there was no significant dif­ference (P<0.05) between the calculated survival by ference (P<0.05) between survivals calculated by the three used methods (Figure 3). three methods in total treatment time of up to 60 TABLE 2. Experimental and calculated survival using method III for total dose of 4 Gy 1×4 5 0.518±0.019 0.459±0.017 0.513±0.038 0.445±0.012 2×2 15 0.535±0.027 0.506±0.018 0.534±0.036 0.505±0.048 2×2 30 0.549±0.017 0.547±0.018 0.550±0.023 0.545±0.019 2×2 60 0.570±0.016 0.588±0.017 0.569±0.029 0.587±0.042 2×2 120 0.586±0.016 0.609±0.018 0.585±0.019 0.609±0.027 2×2 180 0.590±0.018 0.612±0.019 0.590±0.054 0.612±0.038 2×2 240 0.591±0.029 0.613±0.016 0.591±0.024 0.613±0.029 4×1 30 0.546±0.026 0.546±0.017 0.542±0.038 0.529±0.035 8×0.5 30 0.549±0.015 0.547±0.026 0.540±0.034 0.523±0.043 4×1 60 0.570±0.017 0.585±0.017 0.567±0.047 0.595±0.047 8×0.5 60 0.570±0.016 0.583±0.008 0.564±0.028 0.589±0.038 4×1 240 0.598±0.018 0.653±0.027 0.622±0.024 0.706±0.038 8×0.5 240 0.607±0.008 0.674±0.008 0.627±0.023 0.732±0.042 352 A B FIGURE 3. The survival fraction predicted by the used methods for F10B16 melanoma (A) and 4T1 breast adenocarcinoma (B) cell lines in different fraction numbers, dose per fractions, and total treatment times, for dose of 2 Gy. minute (Figure 4). After 60 minute, for the F1B16 cells, there was a significant difference between method I and the other two methods. While in the treatment time less than 240 minute there was no significant difference between methods I and III (P<0.05). For the 4T1 cells, there were significant differences (P<0.05) in calculated survival between method I and the two other methods. Considering total treatment time, these variations increased considerably after 60 minute. Small differences ob­served between methods II and III in groups with 4 or 8 subfractions (Figure 4). For a total dose of 6 Gy, the results of calcula­tions for the F10B16 cells were the same as dose of 4 Gy (Figure 5). For the 4T1 cells, a significant difference between the used methods observed, es­pecially between the method I and the other two methods (Figure 5). These results showed that, when the total treat­ment time increased, the survival of both two cell lines increased significantly according to all three methods. Increasing the number of subfractions showed different results according to the used methods. According to the method I, increasing the num­ber of subfractions in a fixed total treatment time reduced the survival in all three doses of 2, 4 and 6 Gy. The predicted survival according to the method II did not show any significant difference (P<0.05) due to the variations in number of subfractions. The calculated survival by the method III for F10B16 melanoma cell line, showed a significant decrease by increasing the number of subfractions from 2 to 4 and 4 to 8, for total dose of 2, 4 and 6 Gy and both treatment times of 30 and 60 minute. While, for the 240 minute treatment time, increas­ing the number of subfractions increased the sur­vival of the cells (Figures 3 to 5). For 4T1 cell line, increasing the number of sub-fractions decreased the survival of the cells in 30 minute treatment time. For the total treatment time of 60 minute, increasing the number of subfrac­tions from 2 to 4 fractions enhanced the survival of the cells, whereas, increasing the subfractions to 8 declined the cells survival. For the 240 minute treatment time, increasing the number of subfrac­tions raised the cells survival (Figures 3 to 5). Considering the three used methods, differ­ences between the exposed groups to 2 Gy was not significant for the F10B16 cells and was negligible for 4T1 cells. Therefore, to investigate the effects of number of subfractions and dose per subfraction in experimental investigations, a total dose of 4 Gy was used. The results of this experiment were 353 assessed in 2, 4 and 8 fractions during treatment times of 30, 60 and 240 min, for both F10B16 and 4T1 cell lines. Comparisons between the experimental results with those calculated by the three used methods showed that experimental results were in a good agreement with method III. The results of experi­mental investigations and the calculated survival by the method III are shown in Table 2. The results showed that, in a fixed overall treat­ment time, there was no statistical significant dif­ference (P<0.05) between the irradiated groups in different subfractions. Considering the overall treatment time, there was an agreement between experimental results and those predicted by the method III for the irradiated cells in total treatment time of 1 h, as opposed to the 4 h irradiated group. Discussion Recently, some researchers have shown the ef­fect of prolonged dose delivery time on the cell survivals.8-15 In this regard, several models have been offered to predict the effects of variations in the treatment procedures on the cells survival.8,9,20 One of these models is the developed LQ model by Thames and Dale.17 However, different theoreti­cal methods have been derived from this model in some researches in order to predict the survival of cells after prolonged dose delivery schemes.8,9,10,19 As stated earlier, these researches have just inves­tigated the effect of total treatment time and have not considered the effects of number of subfrac­tions, dose per subfarction and the time intervals between subfractions in detail. Therefore, more investigations were needed in order to determine the effect of different treatment factors on cells’ survival. In addition, it seems use­ful to compare the results of these methods theo­retically and experimentally in order to find the best method for clinical application in fractionation radiotherapy. In this study, three calculation methods derived from the basic LQ model proposed in different re­searches8,9,20 were used to evaluate the effects of different parameters such as total treatment time, number of subfractions, and subfractions interval on survival of cell lines with constant ., ß and µ parameters. Then, the results were compared with experimental outcomes of F10B16 skin melanoma and 4T1 breast adenocarcinoma cells. Comparison between the results of the used three methods with those of experimental results 354 showed that method III (Mu et al. model) was in a better agreement with experimental outcomes. Mu et al. proposed a method to calculate the effect of prolonged treatment time on the Chinese hamster fibroblasts (V79-379-A) cells’ survival for total treat­ment dose of 2 and 8 Gy. They have shown that, there is a good agreement between experimental and theoretical results for the total dose of 2 Gy and treatment time below 1 hour. While in our study, different mathematical methods presented by Keall et al., Brenner and Mu et al., were used to calculate the cells’ survival after different treatment schemes such as 2, 4, and 6 Gy continuous dose in two sub-fractions with dose of 1, 2, and 3 Gy, respectively. In this work, to investigate the effects of the number of subfractions and dose per subfraction, the cells’ survival after total doses of 2 Gy (4 subfractions of 0. 5 Gy and 8 subfractions of 0.25 Gy), 4 Gy (4 sub-fractions of 1 Gy and 8 subfractions of 0.5 Gy), 6 Gy (4 subfractions of 1.25 Gy and 8 subfractions of 0.75 Gy) were calculated through fixed overall treat­ment times of 30, 60 and 240 minutes. Considering the method III investigations in pre­ dicting the F10B16 cells survival (T1/2 = 30 minute), it is expected that increasing the number of subfrac­tions reduced the survival, in total treatment times of 30 and 60 minute. The reason was due to the repair of sublethal damages. For all defined sub-fractions (2, 4 and 8), the intervals between subfrac­ tions was lower than T1/2, therefore, after the first irradiation there was not enough time for the cells to repair their sublethal damages, hence the sur­vival reduced. This effect was found for 4T1 cells in total treatment time of 30 minute. However, for the 60 minute treatment time, considering the T1/2 of about 20 minute (significantly lower than F10B16) the time between 4 subfractions was higher than the repair time, and therefore, after irradiation in the first subfraction the damages were repaired be­fore starting the next exposure, consequently the survival increased. However, for the 8 subfractions in 60 minute treatment time, the results were the same as before. These explanations can justify the behavior of the used both two cell lines in 240 min­ute treatment time, too. Therefore, the survival of cells increased for this total treatment time. Experimental results showed that increasing the total treatment time, similarly occurred in new complicated methods such as IMRT, increased the cell survival in both cell lines and all three total dose of 2, 4 and 6 Gy in up to 2 hour treatment time. However, the extent of this effect was not consid­erable for F10B16 cells with shorter T1/2, and was negligible for the dose of 2 Gy for this cell line. 355 Moreover, the results of this research confirmed that a cell with lower ./ß ratio is considered to have a greater ability to undergo sublethal damage repair. The rate of sublethal damage repair may be represented by T1/2; therefore, cells with a shorter T1/2 have more repairs. In addition, the survival of 4T1 cell line with lower ./ß and T1/2 was dra­matically different than the F10B16, when the time interval between subfractions increased. In total treatment time of 4 hours, both theoreti­cal and experimental results showed an increase in survival with fractionated irradiation. Some studies have been performed to investigate the ability of LQ model in predicting the survival in low dose levels (< 1 Gy).21,28,29 Cherubini et al. and Jones et al. explained that in low doses (less than 1 Gy) the LQ model cannot predict the cell survival accurately.28,21 While, Smith et al. claimed that the LQ model calculates the survival precisely in such low doses.29 Brenner has shown that in total dose of 2 to 15 Gy, the LQ model can accurately predict the survival in in-vitro and in-vivo conditions.20 In this study, in line with Brenner, the results suggest that, in fractionation radiotherapy, the developed LQ model can potentially reach close agreement with reality in total treatment dose of 2 to 4 Gy. Compared with other studies, using small sub-fractions of 0.25–0.5 Gy, Marples et al.30, and Mu et al.8 investigated the phenomenon of hypersensitiv­ity to low doses per fraction. Marples et al. showed that, this would lead to a more effective cell killing than predicated by the LQ model.30 While Mu et al. study showed that there was no evidence for such effect since it should have resulted in lower surviv­al than expected and not higher.8 They explained that this effect is perhaps because of the effective dose rate in each fraction which is too high to avoid activating a possible repair.8 However, in our study which lower dose rate was used, cell killing reached close agreement to the amount predicted by the LQ model that is in an agreement with the Marples et al. result. The factors that influence the dose rate are radical recombination and sublethal damage repair.30 It should be noted that, at the dose levels and dose rates encountered in radiotherapy, the effect of radical recombination on cell killing is negligible.31 Ling et al.32 and Michaels et al.33 have compared the survival of CHO cells at dose rates of 0.6 Gy/min from a Co-60 unit, and their results showed that the obtained survival curves were ex­actly the same with up to 15 Gy/min dose rates. Hence, based on the results of our study and com­parisons with other works, an idea to reduce the effect of fractionation or prolonged treatment time is using higher dose rates or more treatment dose in one fraction. In other work by Keall et al.9, they have shown that both respiratory gating and IMRT delivery will decrease survival compared with continuous delivery of the same dose in the same overall time. Therefore, for a given treatment time, delivery method is another factor affecting the cell survival. Conclusions According to presented experimental and theo­retical results, in treatment of tumors in radio­therapy by new complicated methods, this should be noted that exceeding the treatment time will increase the survival of tumor cells and may de­crease tumor control. Increasing the number of subfractions in a course of treatment could reduce the cell survivals if the fractions time interval be lower than the repair time of sublethal damages. Although, this parameter has a negligible effect on the survival of the cell lines of interest in our ex­perimental study, this factor can be considered in compensating the increase in cell survival due to the time prolongation. It seems appropriate to use the method pro­posed by Mu et al. to predict the cell response fol­lowing fractionation radiotherapy, especially in new fractionation radiotherapy procedures with more number of subfractions and with prolonged total treatment times. This method can simply and accurately determine the cell survival after each radiotherapy assessment and can be used to cal­culate the compensating dose for these treatment schedules. Although the effect of fractionation dose delivery is negligible for one session (with dose of 2 Gy), and it seems that there is no need to compensate these effect, but it can be important for a radiotherapy period (30 or 35 session with 2 Gy in each fraction) because of the cumulative ef­fect of dose. References 1. Baskar R, Lee KA, Yeoh KW. Cancer and radiation therapy: current advances and future directions. Int J Med Sci 2012; 9: 193-9. 2. Begg CA, Stewart FA, Vens C. Strategies to improve radiotherapy with tar­geted drugs. Nat Rev Cancer 2011; 11: 239-53. 3. Stavrev P, Hristov D. Prostate IMRT fractionation strategies: two-phase treat­ment versus simultaneous integrated boost. Radiol Oncol 2003; 37: 115-26. 4. Chang SX, Cullip TJ, Deschesne KM. Intensity modulation delivery tech­niques: ‘step & shoot’ MLC auto-sequence versus the use of a modulator. Med Phys 2000; 27: 948-59. 356 5. Verhey LJ. Physical considerations in the use of intensity modulated radio­therapy to produce three-dimensional conformal dose distributions. J Jpn Soc Ther Radiol Oncol 2000; 12: 191-203. 6. Jeraj M, Robar V. Multileaf collimator in radiotherapy. Radiol Oncol 2004; 38: 235-40. 7. Ling CC, Burman C, Chui CS, Kutcher GJ, Leibel SA, LoSasso T, et al. Conformal radiation treatment of prostate cancer using inversely-planned intensity-modulated photon beams produced with dynamic multileaf collimation. Int J Radiat Oncol Biol Phys1996; 35: 721-30. 8. Mu X, Löfroth P-O, Karlsson M, Zackrisson B. The effect of fraction time in intensity modulated radiotherapy: theoretical and experimental evaluation of an optimization problem. Radiother Oncol 2003; 68: 181-7. 9. Keall PJ, Chang M, Benedict S, Thames H, Vedam SS, Lin P-S. Investigating the temporal effects of respiratory-gated and intensity-modulated radiotherapy treatment delivery on in vitro survival: an experimental and theoretical study. Int J Radiat Oncol Biol Phys 2008; 71: 1547-52. 10. Fowler JF, Welsh JS, Howard SP. Loss of biological effect in prolonged fraction delivery. Int J Radiat Oncol Biol Phys 2004; 59: 242-9. 11. Zheng XK, Chen LH, Wang WJ, Ye F, Liu JB, Li QS, et al. Impact of prolonged fraction delivery times simulating IMRT on cultured nasopharyngeal carci­noma cell killing. Int J Radiat Oncol Biol Phys 2010, 78: 1541-7. 12. Paganetti H. Changes in tumor cell response due to prolonged dose delivery times in fractionated radiation therapy. Int J Radiat Oncol Biol Phys 2005; 63: 892-900. 13. Wang X, Xiong XP, Lu J, Zhu GP, He SQ , Hu CS, et al. The in vivo study on the radiobiologic effect of prolonged delivery time to tumor control in C57BL mice implanted with Lewis lung cancer. Radiat Oncol 2011; 6: 4. 14. Wang JZ, Li XA, D’souza WD, Stewart RD. Impact of prolonged fraction delivery times on tumor control: a note of caution for intensity-modulated radiation therapy (IMRT). Int J Radiat Oncol Biol Phys 2003; 57: 543-52. 15. Shibamoto Y, Masato I, Sugie C, Ogino H, Hara M. Recovery from sublethal damage during intermittent exposures in cultured tumor cells: implications for dose modification in radiosurgery and IMRT. Int J Radiat Oncol Biol Phys 2004; 59: 1484-90. 16. Ling CC, Gerweck LE, Zaider M, Yorke E. Dose-rate effects in external beam radiotherapy redux. Radiother Oncol 2010; 95: 261-8. 17. Thames HD. An ‘incomplete-repair’ model for survival after fractionated and continuous irradiations. Int J Radiat Biol Relat Stud Phys Chem Med 1985; 47: 319-39. 18. Nilsson P, Thames HD, Joiner MC. A generalized formulation of the ‘incom­plete repair ’ model for cell survival and tissue response to fractionated low dose-rate irradiation. Int J Radiat Biol 1990; 57: 127-42. 19. Rourke SFCO, Mcaneney H, Hillen T. Linear quadratic and tumor control probability modeling in external beam radiotherapy. J Math Biol 2009; 58: 799-817. 20. Brenner DJ. The linear-quadratic model is an appropriate methodology for determining isoeffective doses at large doses per fraction. Semin Radiat Oncol 2008; 18: 234-9. 21. Jones L, Hoban P, Metcalfe H. The use of the linear quadratic model in radiotherapy: a review. Australas Phys Eng Sci Med 2001; 24: 132-46. 22. Price P, McMillan TJ. Use of the tetrazolium assay in measuring the response of human tumor cells to ionizing radiation. Cancer Res 1990; 50: 1392-6. 23. Buch K, Peters T, Nawroth T, Sanger M, Schmidberger H, Langguth P. Determination of cell survival after irradiation via clonogenic assay versus multiple MTT assay – a comparative study. Radiat Oncol 2012; 7: 1. 24. Sieuwerts AM, Klijn JGM, Peters HA, Foekens JA. The MTT Tetrazolium salt assay scrutinized: how to use this assay reliably to measure metabolic activity of cell cultures in vitro for the assessment of growth characteristics, IC50-values and cell survival. Eur J Clin Chem Clin Biochem 1995; 33: 813-23. 25. Nikzad S, Hashemi B, Hasan Z S, Mozdarani H. The cell survival of F10B16 melanoma and 4T1 breast adenocarcinoma irradiated to gamma radiation using the MTT assay based on two different calculation methods. J Biomed Phys Eng 2013; 3: 29-36. 26. Nikzad S, Hashemi B. MTT assay instead of the clonogenic assay in measur­ing the response of cells to ionizing radiation. J Radiobiol 2014; 1: 3-8. 27. Shanei A, Baradaran-Ghahfarokhi M. Evaluation of testicular dose and as­sociated risk from common pelvis radiation therapy in Iran. Physica Medica 2014; 30: 867-70. 28. Cherubini R, De Nadal V, Gerardi S, Guryev D. Status report of a system­atic investigation on low dose ionizing radiation effects in mammalian cells. Nuoro Cimento C 2008; 1: 57-67. 29. Smith LG, Miller RC, Richard SM, Brenner DJ, Hall EJ, Phil D. Investigation of hypersensitivity to fractionated low dose radiation exposure. Int J Radiat Oncol Biol Phys 1999; 45: 187-91. 30. Marples B, Joiner MC. The response of Chinese hamster V79 cells to low radiation doses: evidence of enhanced sensitivity of the whole cell popula­tion. Radiat Res 1993; 133: 41-51. 31. Ling CC, Gerweck LE, Zaider M, Yorke E. Dose-rate effects in external beam radiotherapy redux. Radiother Oncol 2010; 95: 261-8. 32. Ling CC, Spiro IJ, Mitchell J, Stickler R. The variation of OER with dose rate. Int J Radiat Oncol Biol Phys 1985; 11: 1367-73. 33. Michaels HB, Epp ER, Ling CC, Peterson EC. Oxygen sensitization of CHO cells at ultrahigh dose rates: prelude to oxygen diffusion studies. Radiat Res 1978; 76: 510-21. 357 research article Urokinase plasminogen activator (uPA) and plasminogen activator inhibitor type-1 (PAI-1) in breast cancer - correlation with traditional prognostic factors Maja Lampelj1, Darja Arko1, Nina Cas-Sikosek1, Rajko Kavalar2, Maja Ravnik1, Barbara Jezersek-Novakovic3, Sarah Dobnik1, Nina Fokter Dovnik1, Iztok Takac1,4 1 Department of Gynaecologic and Breast Oncology, Division of Gynaecology and Perinatology, University Medical Centre Maribor, Maribor, Slovenia 2 Division of Pathology, University Medical Centre Maribor, Maribor, Slovenia 3 Division of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia 4 Faculty of Medicine University of Maribor, Maribor, Slovenia Radiol Oncol 2015; 49(4): 357-364. Received 3 August 2014 Accepted 11 November 2014 Correspondence to: Sarah Dobnik, M.D., Department of Gynaecologic and Breast Oncology, Division of Gynaecology and Perinatology, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia. E-mail: sarah_dobnik@yahoo.com Disclosure: No potential conflicts of interest were disclosed. Background. Urokinase plasminogen activator (uPA) and plasminogen activator inhibitor type-1 (PAI-1) play a key role in tumour invasion and metastasis. High levels of both proteolytic enzymes are associated with poor prognosis in breast cancer patients. The purpose of this study was to evaluate the correlation between traditional prognostic fac­tors and uPA and PAI-1 expression in primary tumour of breast cancer patients. Patients and methods. 606 primary breast cancer patients were enrolled in the prospective study in the Department of gynaecological oncology and breast oncology at the University Medical Centre Maribor between the years 2004 and 2010. We evaluated the traditional prognostic factors (age, menopausal status, tumour size, pathohistological type, histologic grade, lymph node status, lymphovascular invasion and hormone receptor status), together with uPA and PAI-1. We used Spearman’s rank correlation, Mann Whitney U test and .2 test for statistical analysis. Results. Our findings indicate a positive correlation between uPA and tumour size (p < 0.001), grade (p < 0.001), his­tological type (p < 0.001), lymphovascular invasion (p = 0.01) and a negative correlation between uPA and hormone receptor status (p < 0.001). They also indicate a positive correlation between PAI-1 and tumour size (p = 0.004), grade (p < 0.001), pathohistological type (p < 0.001) and negative correlation between PAI-1 and hormone receptor status (p = 0.002). Conclusions. Our study showed a relationship between uPA and PAI-1 and traditional prognostic factors. Their role as prognostic and predictive factors remains to be further evaluated. Key words: urokinase plasminogen activator; plasminogen activator inhibitor; breast cancer; prognostic factor Introduction Urokinase plasminogen activator system (uPAS) consists of urokinase plasminogen activator (uPA), tissue plasminogen activator (tPA), urokinase plas­minogen activator receptor (uPAR), and plasmino­gen activator inhibitor type-1 (PAI-1) and type-2 (PAI-2). Proteolytic enzyme uPA converts the pro-enzyme plasminogen into proteolytically active form (plasmin), which takes part in physiologi­cal and pathophysiological processes on the basal membrane and inside the extracellular matrix, which are important for tumour growth and its me­tastases.1 Plasminogen activator inhibitor type-1, 358 which functions as a natural inhibitor of uPA, is the most important factor among fibrinolytic inhibi­tors for the development of vascular diseases and cancer. uPA and PAI-1 do not only have proteolytic characteristics, but also have impact on the funda­mental cellular processes, such as chemotaxis, mi­gration, invasion, adhesion, proliferation and an­giogenesis.2-6 uPA and PAI-1, as part of the fibrino­lytic system, are the first factors with a confirmed clinical role in breast cancer (level of evidence I).7,8 According to the conclusions of the meta-analysis by Look et al.7, uPA and PAI-1 are in addition to ax­illary lymph node involvement the most important independent prognostic factors. uPA and PAI-1 are supposed to be useful in deciding upon adjuvant systemic therapy in women with low-risk primary breast cancer.7 Use of PAI-1 for therapeutic purpos­es has shown promising results on tumour models; however, the results are yet to be confirmed.9 The increase of PAI-1 could represent a response to the increased proteolytic activity caused by uPA inside the tumour. It is also possible that PAI-1 has a direct effect on the development of the disease.10 Prognostic and predictive factors are clinically important for planning the treatment of breast can­cer, which improves disease-free survival, over­all survival and quality of life. Prognostic factors predict the course of the disease independently of treatment and are connected with disease-free survival and overall survival. Tumour size, axillary lymph node involvement, pathohistological tu­mour type, malignancy grade and lymphovascular invasion are prognostic factors in the case of breast cancer. To assess patients with a high risk of recur­rence, traditional prognostic factors do not suffice. Therefore, numerous studies are being conducted to discover better factors. uPA in PAI-1 are related to the course of breast cancer as statistically impor­tant independent prognostic factors.11-15 Numerous studies have shown that patients with low concen­trations of uPA and PAI-1 have better survival than patients with high concentrations.16-17 The prognos­tic roles of DNA ploidy and S-phase fraction are not clearly defined yet.18 Predictive factors are biological markers by means of which it is possible to predict response to a certain type of treatment. Status of the hormone receptors, which predicts the response to hormonal therapy, and human epidermal growth factor receptor 2 (HER2) expression, which predicts the response to anti-HER2 therapy in patients with HER2-positive breast cancer, were confirmed to be reliable predic­tive factors in breast cancer. High level of evidence supports the predictive significance of uPA and PAI­1, which are the subject of many studies.19 Protein over-expression and/or amplification of the HER2 gene are found in around 20% of all breast cancer patients. Pre-clinical studies show that HER2 accel­erates cellular adhesion and migration and therefore plays a key role in tumour cell invasion.20-23 Certain clinical studies indicate that in some cancer types HER2 stimulates the invasion of tumour cells with the effect on the accelerated release of proteolytic enzyme uPA and its inhibitor (PAI-1)24-27, whereas other studies did not confirm this assumption.28,29 The international coordinated guidelines, adopt­ed at the conference in St. Gallen in 2007, require knowledge of factors such as tumour size, malig­nancy grade, age, axillary node involvement, status of hormone receptors and HER2 expression as the basis for choosing adjuvant therapy.30 Despite excellent evidence about the prognos­tic value of uPA and PAI-1, determination of these markers is not yet routinely used for planning ad­juvant treatment. It is not completely clear if rou­tine determination of uPA and PAI-1 would add important new information as opposed to simply confirming what can already be deduced from the traditional prognostic factors. The aim of this study was to evaluate the correla­tion between uPA and PAI-1 and traditional prog­nostic factors in primary breast cancer. Statistically significant correlation between uPA and PAI-1 and traditional prognostic factors was expected. HER2 expression and its correlation with the traditional prognostic factors were also included. Patients and methods Patients Six hundred and six patients with primary breast cancer, treated at the Department of Gynaecologic Oncology and Breast Oncology of the Division of Gynaecology and Perinatology, University Medical Centre Maribor, between the years 2004 and 2010 were included in this prospective study. The study was conducted in accordance with good clinical practice and all applicable regulatory re­quirements, including Declaration of Helsinki. The study was approved by the institutional re­view board and registered at Slovenian Research Agency under the clinical trial number P3-0321. All patients had pathohistologically confirmed invasive breast cancer. None of the patients had clinically or radiologically registered metastatic disease at the beginning of primary treatment. The characteristics of patients and tumours are present­ 359 ed in Table 1. Traditional prognostic factors such TABLE 1. Characteristics of primary breast cancer patients and tumours (n = 606) as menopausal status, pathological tumour size, pathohistological tumour type, malignancy grade, axillary lymph node involvement and lymphovas- Age 136 22 cular invasion were assessed by means of clinical < 50 years 470 78 . 50 years examination and pathohistological examination of Menopausal status tumour tissue. Tumours were classified according Premenopausal 162 27 to the UICC-WHO criteria and malignancy grade Postmenopausal 444 73 Pathological tumour size according to Scarff-Bloom-Richardson (SBR) clas­ < 2 cm 282 46sification, modified by Elston.31 Lymphovascular . 2 cm 319 53 Unknown 5 1 invasion was evaluated as positive if tumour cell Pathohistological classification of emboli were present in the vascular space lined by tumours 496 82 endothelium. Hormone receptors were evaluated Invasive ductal 45 7Invasive lobular by means of immunohistochemical staining of par- 61 10 Other invasive 4 1 affin-embedded tumour tissue sections. Tumours Unknown in which at least 1% of tumour cells expressed Malignancy grade G1 126 21 oestrogen (ER) and/or progesterone (PR) receptors G2 212 35 were marked as hormone receptor positive. G3 235 39 Unknown 33 5 The study group was a cohort of women with Axillary lymph node involvement breast cancer primarily treated at our institution Negative 333 55 during a period of seven years. During this time, Positive 243 40 Unknown 30 5 recommendations for determination of some histo- Oestrogen receptors 119 20 logical parameters have changed. Progesterone re- Negative 478 79 Positive ceptors and HER2 were not routinely determined 9 1 Unknown in all patients throughout the study period and for Progesterone receptors 219 36 some early cases of HER2 determination in situ hy- Negative 337 56 Positive bridization was not performed in cases that were 50 8 Unknown immunohistochemically marked as 2+. Besides, tu- Hormone receptors mour grade was not reported for lobular histologi-Negative 101 17 Positive 492 81 cal subtypes in the past. Unfortunately, all this has Unknown 13 2 lead to a high rate of missing data in these fields. Lymphovascular invasion All patients were radically locally treated with Yes 103 17 No 481 79 a modified radical mastectomy or conservative Unknown 22 4 operation (tumorectomy, quadrantectomy) and HER2 Negative 127 21 postoperative radiation. They further received Positive 373 62adjuvant systemic therapy (chemotherapy and/ Unknown 106 17 or hormone therapy). Most patients with positive axillary lymph nodes and patients with a high risk and negative lymph nodes received adjuvant chemotherapy. All patients with positive hormone receptors received adjuvant hormone therapy. Laboratory measurements of uPA, PAI-1 and HER2 After histological examination of tissue sections, the tumour tissue obtained by surgery was stored for further analysis in liquid nitrogen. Samples of frozen tumour tissue were then pulverized with a Micro-dismembrator, dissolved in buffer (pH 5.5) composed of 0.02 M Tris-HCl, 0.125 M NaCl and 2% Triton X-100 and after 3 hours of stirring at 4oC centrifuged at 100,000 x g for 30 minutes. Protein content was measured with the bicinchoninic acid G = grade; HER2 = human epidermal growth factor receptor 2 (BCA) method (Pierce, Rockford, IL). Antigens uPA and PAI-1 were quantified with standardized im­munometric method using ELISA sets (American Diagnostica, Greenwich, CT, USA). Values of uPA and PAI-1 were expressed in ng/mg of proteins. Based on the assesed intensity of membrane reaction due to the overexpression of HER onco­protein, the tumour tissue was categorized into one out of three groups: negative (0, 1+), equivo­cal (2+) and positive (3+). The immunohistochemi­cally HER2 3+ result indicates positive HER2 status of the tumours. In all cases of the equivocal HER2 2+ results, the tumour tissue was retested with the fluorescence in situ hybridization (FISH) method 360 * ng / mg of proteins TABLE 2. Levels of urokinase plasminogen activator (uPA) and plasminogen activator inhibitor type-1 (PAI-1) in tumour tissue of primary breast cancer patients 2.34 < 3 319 59 uPA 0–24.95 (1.08 / 4.20) . 3 223 41 10.6 < 14 347 64 PAI-1 0–170.92 (6.93 / 18.27) . 14 195 36 using PathVysionTM HER-2 PROBE KIT, in order to determine the amplification of HER2 proto-onco­gene. All breast cancer patients with HER2 positive tumour status were treated with the monoclonal antibody trastuzumab. Statistical analysis Spearman Rank Correlation was used to test the re­lation between continuous variables. We used the non-parametric Mann-Whitney U test to compare continuous and categorical variables and chi-square test for the comparison of categorical variables. Continuous variables uPA and PAI-1 were convert­ed into binary variables to divide the patients into those with high risk and those with low risk by us­ing limit values of 3 ng uPA / mg of proteins and 14 ng PAI-1 / mg of proteins.32 Statistical analysis was performed by means of the SPSS 17.0 program. The value of p < 0.05 was considered statistically signifi­cant. uPA and PAI-1 values are shown in Table 2. Results 606 patients with primary breast cancer were in­cluded in the prospective study. Mean age of pa­tients was 60.1 ± 12.7 years. The youngest patient was 22 years old, and the oldest 95 years old. Correlation between uPA and PAI-1 values A strongly positive statistically significant correla­tion was established between uPA and PAI-1 val­ues (rs = 0.576, p < 0.001). The relation between uPA and PAI-1 values is shown in Figure 1. Correlation between uPA and PAI-1 and traditional prognostic factors Statistically significant correlation between uPA and PAI-1 and most of the traditional prognostic FIGURE 2. Correlation between urokinase plasminogen activator (uPA) and malignancy grade. Similar correlations were found for uPA and tumour size, pathohistological tumour type and lymphovascular invasion. Negative correlation was found between uPA and hormone and oestrogen receptor status. Analogous correlations between and plasminogen activator inhibitor type-1 (PAI-1) and all these factors except lymphovascular invasion were also determined. factors was established. They were related with tu­mour size, pathohistological tumour type, malig­nancy grade, status of oestrogen (ER) and hormone receptors. The relation between uPA and malig­nancy grade is shown in Figure 2. No statistically significant correlation between uPA and PAI-1 and age, menopausal status, pro­gesterone receptors (PR) and axillary lymph node involvement was found. Interestingly, only uPA’s correlation with lymphovascular invasion was sta­ 361 tistically significant. The correlation between uPA, TABLE 3. Correlations between urokinase plasminogen activator (uPA), plasminogen activator inhibitor type-1 (PAI-1), human epidermal growth factor receptor 2 (HER2) PAI-1 and traditional prognostic factors is shown and traditional prognostic factors in Table 3. Correlation between tumour HER2 Agepositive status and traditional prognostic < 50 years 136 p = 0.700 p = 0.402 p = 0.017 . 50 years 470 factors Menopausal status HER2 tumour status was statistically related to all Premenopausal 162 p = 0.896 p = 0.218 p = 0.008 Postmenopausal 444 the traditional prognostic factors, except to axillary Pathological tumour sizelymph node status (Table 3). < 2cm 282 p < 0.001 p = 0.004 p = 0.005 . 2cm 319 Pathological tumour typePatients with a high risk due to high Ductal invasive 541 p < 0.001 p < 0.001 p = 0.021 Other invasive 61 uPA and PAI-1 values and tumour HER2 Malignancy gradepositive status G1 + G2 338 p < 0.001 p < 0.001 p < 0.001 G3 235 High uPA levels were present in 223 patients (41%) Axillary lymph node involvement and high PAI-1 levels in 195 (36%) patients, as Negative 333 p = 0.052 p = 0.171 p = 0.385 Positive 243 shown in Table 2. High levels of one or both pro- Oestrogen receptorsteolytic enzymes, uPA and PAI-1, were identified Negative 119 p < 0.001 p < 0.001 p < 0.001 Positive 478 in 288 (53%) patients. HER2 overexpression was Progesterone receptors identified in 127 (25%) patients. Negative 219 p = 0.162 p = 0.960 p = 0.003Among 183 patients with high uPA, 50 patients Positive 337 (27%) had HER2-positive tumours. In the group of Hormone receptors Negative 101 p < 0.001 p = 0.002 p < 0.001 263 patients with low uPA, 60 patients (23%) had Positive 492 HER2-positive tumours. Out of 157 patients with Lymphovascular invasion (LVI) high PAI-1, 46 patients (29%) had HER2-positive Yes 103 p = 0.010 p = 0.292 p = 0.006 No 481 tumours. Among 289 patients with low PAI-1, 64 patients (22%) had HER2-positive tumours. In the * Due to missing values the number of patients is not always 606; G = grade group of 238 patients with high values of one or both proteolytic enzymes, uPA and PAI-1, 65 pa­tients (27%) had HER2-positive tumours and out of the 214 patients with low values of uPA and PAI-1 The purpose of the study was to evaluate the 47 patients (22%) had HER2-positive tumours. relation between uPA and PAI-1 and traditional prognostic factors in primary breast cancer. HER2 overexpression as a prognostic and predictive fac-Discussion tor and its correlation with traditional prognostic factors was also included in the study. Prognostic and predictive factors play a key role The sample size (606 patients) in our study is in the proper treatment of breast cancer patients. comparable with the reports by other authors.33-39 Knowledge about these factors enables the patients The study provides exact data on the character-with aggressive malignant tumours the possibility istics of patients with primary breast cancer and of adjuvant systemic therapy along with better sur-their tumours. More than a third of patients (78%) vival and spares the patients with less aggressive were aged 50 or more and 73% of patients were malignant tumours unnecessary systemic treat-postmenopausal. We determined that HER2 over­ment with numerous side effects while having the expression is more frequently present in younger same chance of recovery. Establishing prognostic and premenopausal patients. The same correla­factors that are independent of treatment became tion was not established with uPA and PAI-1. very challenging in modern medicine, as most pa-Similarly, Look et al.7 found no significant relation­tients with breast cancer receive adjuvant systemic ship between uPA and age or menopausal status. therapy after the primary treatment. It would be However, they reported a correlation between unethical to discontinue the adjuvant systemic PAI-1 and age and higher PAI-1 in postmenopausal therapy for research purposes regarding the char-women. They nevertheless considered these rela­acteristics of the disease. tionships not to be clinically meaningful. 362 Axillary lymph node involvement, tumour size, pathohistological tumour type, malignancy grade and lymphovascular invasion are the most im­portant prognostic factors of the clinical course of breast cancer. In numerous studies these tumour characteristics were proved as independent prog­nostic factors since the disease recurred more often and affected the survival in patients with affected axillary lymph nodes, larger tumours, invasive ductal carcinoma, higher malignancy grade and lymphovascular invasion. Numerous previous studies have shown that high levels of uPA and PAI-1 in primary tumour tissue negatively affect the outcome of breast can­cer. uPA enables the development of metastases through proteolytic degradation of the extracel­lular matrix. Furthermore, PAI-1 also has an im­portant role in invasion and metastasis, because it does not act only as inhibitor of uPA in plasmino­gen activator system but also affects most basic cell processes, such as adhesion, migration, invasion, proliferation and apoptosis of normal and malig­nant cells. Eljuga et al.17 even showed that PAI-1 de­termined immunohistochemically in tumour cells as opposed to the less available ELISA testing may carry important prognostic information in node-negative breast cancer patients. A strongly positive correlation between both proteolytic enzymes, uPA and PAI-1, was es­tablished, which is in line with the findings of other studies.7,28,29,34,36 Despite this correlation, it is clinically important to determine both factors. Establishing both values allows differentiation be­tween groups with high risk (with a high level of one or both factors) and making a decision on the proper individual adjuvant therapy.40 Size of the primary tumour is a known prognos­tic factor for the course of breast cancer. Patients with primary tumours equal to or larger than 2 cm, with a 53% share in our study, had more often high uPA and PAI-1 levels. A similar share of patients with large tumours (56%) was reported by Look et al.7 In larger tumours HER2 overexpression was also more frequently present. Axillary lymph node involvement is related to tumour size, as larger tumours more often devel­op regional lymph node metastases. Interestingly though, high uPA and PAI-1 values in our patients were not related to axillary lymph node involve­ment. The reason may lie in the role of uPA and PAI-1 as independent prognostic and predictive factors in patients with primary breast cancer without axillary lymph node involvement. uPA was proved to be a stronger prognostic factor than tumour size, axillary lymph node involvement and oestrogen receptor status. Furthermore, it was also proved to be the strongest predictive factor of dis-ease-free survival and overall survival in patients with primary breast cancer and no axillary lymph node involvement in numerous studies.7,8,11,15,41,42 In our study, 154 patients (40%) had a high uPA level and no axillary lymph node involvement. High PAI-1 levels were present in 136 patients (36%). High levels of both, uPA and PAI-1, were present in 88 patients (21%) with no axillary lymph node involvement. Individually or both, the uPA and PAI-1 levels were high in 202 patients (54%). De Cremoux et al.42 established high one or both levels of uPA and PAI-1 in 56% of patients with no axil­lary lymph node involvement, which is compara­ble to our results. Neither was axillary lymph node involvement related to the HER2 overexpression. Malignancy grade of primary tumour is a con­firmed prognostic factor in breast cancer. We have proved that patients with high levels of uPA and PAI-1 and HER2 overexpression more frequently have high-grade tumours. Poorly differentiated tumours (G2 and G3) were present in 74% of pa­tients. Evaluating the malignancy grade by means of studying the histological structure and cytologi­cal characteristics of malignant cells is a subjective method. Sotiriou et al.43 found that it was necessary in patients with grade 2 tumours (that is 30–60% of all tumours) to determine the genomic grade in­dex of the tumour. By doing so, the patients would be divided into those with a high and low risk for recurrence. G2 tumours were present in 35% of pa­tients in our study. We found that high levels of uPA and PAI-1 and HER2 overexpression are more often present in in­vasive ductal carcinoma than in lobular and other cancer types, which is in line with their different clinical outcomes. Invasive ductal type of breast cancer was present in 82% of our patients. Descotes et al.39 discovered 84% of invasive ductal breast cancer type. Oestrogen and progesterone receptor status is an important predictive factor of the response to hormone therapy. Patients with positive ER and PR in the tumour have better survival than patients with positive only one type of hormone receptors. Patients with hormone receptor negative tumours have the worst survival rate. ER are mostly predic­tive factors of the response to hormone therapy, whereas PR are prognostic factors of the disease course, therefore it is important to measure both in the primary breast cancer tissue. In our study, high levels of uPA and PAI-1 had a negative correlation 363 with ER, but no correlation with PR. HER2 overex­pression had a negative correlation with both types of hormone receptors. We counted 79% of patients with positive ER. Other authors report a similar share of ER-positive tumours.38,39 PR-positive tu­mours were present in only 56% of our patients. The cause for the lack of correlation between uPA and PAI-1 with PR may lie in the missing PR values. Lymphovascular invasion is an important mor­phological prognostic factor. Breast cancer devel­ops metastases into regional and distant lymph nodes by lymphatic dissemination, and metastases into other parenchymal organs by hematogenous spread. Lymphovascular invasion was more fre­quently present in the tumour in patients with high uPA levels. There was no such correlation with PAI-1. Čufer et al.36 did not find any correlation between uPA and PAI-1 and lymphovascular inva­sion. In cases of lymphovascular invasion, HER2 overexpression was more often present. High levels of one or both proteolytic enzymes were found in 53% of our patients and HER2 over-expression was present in 25%. A slightly higher percentage of HER2 overexpressing tumours was found in patients with high uPA, PAI-1 or both. A positive relationship between HER2 and proteo­lytic enzymes has been reported for other types of cancer24,25 but it has not been confirmed in breast cancer.28,29 This is the subject of our further re­search. The results of our study on the correlation be­tween uPA and PAI-1 and classic prognostic fac­tors in primary breast cancer are concordant with the results of a meta-analysis of 8377 patients on the prognostic effect of uPA and PAI-1 in primary breast cancer.7 Correlation between uPA and PAI­1 and tumour size, pathological tumour type and hormone receptors and between uPA and axillary lymph node involvement are in line. Further research of the characteristics of patients with primary breast cancer and their tumours as well as the definition of the role of proteolytic en­zymes uPA and PAI-1 as prognostic and predictive factors in breast cancer is required. Conclusions Urokinase plasminogen activator (uPA) and plas­minogen activator inhibitor 1 (PAI-1) play a key role in invasion and metastases of malignant tu­mours. High levels of both proteolytic enzymes are related to poor prognosis in patients with breast cancer. By conducting this study we established that primary breast cancer patients with high val­ues of uPA and PAI-1 usually have tumours that are larger, higher malignancy grade, invasive duct­al pathohistological type and hormone independ­ent. In cases of higher uPA lymphovascular inva­sion is more often present. We also established that HER2 overexpressing tumours occur more often in younger, premenopausal patients, are usually larger, hormone independent, of higher malignan­cy grade and invasive ductal histology, and they often show lymphovascular invasion. Despite these significant correlations, it seems that uPA and PAI-1 values may help to addition­ally stratify especially node-negative breast cancer patients into different prognostic subgroups. In or­der to form a solid recommendation for or against routinely performing uPA/PAI-1 testing in breast cancer patients, further research about the prog­nostic and predictive impact of these factors in pa­tients with primary breast cancer is required. The role of uPA and PAI-1 in survival of node-negative breast cancer patients is the subject of our ongoing research. Acknowledgement The authors would like to thank prof. Tanja Cufer, M.D., Ph.D., and Simona Borstnar, M.D., Ph.D., for their cooperation in the research programme P3­0321, as well as Ivan Vrhovec, Ph.D., and Barbara Mozina, M.Sc., for their long and successful coop­eration in the project of determining proteases. References 1. Ulisse S, Baldini E, Sorrenti S, D’Armiento M. The urokinase plasminogen activator system: a target for anti-cancer therapy. Curr Cancer Drug Targets 2009; 9: 32-71. 2. Reuning U, Magdolen V, Wilhelm O, Fischer K, Lutz V, Graeff H, et al. Multifunctional potential of the plasminogen activation system in tumor invasion and metastasis (review). Int J Oncol 1998; 13: 893-906. 3. Preissner KT, Kanse SM, May AE. Urokinase receptor: a molecular organizer in cellular communication. Curr Opin Cell Biol 2000; 12: 621-8. 4. Blasi F, Carmeliet P. uPAR: a versatile signalling orchestrator. Nat Rev Mol Cell Biol 2002; 3: 932-43. 5. Rosenberg S. The urokinase-type plasminogen activator system in cancer and other pathological conditions: introduction and perspective. Curr Pharm Des 2003; 9: 4p. 6. Reuning U, Sperl S, Kopitz C, Kessler H, Kruger A, Schmitt M, et al. Urokinase-type plasminogen activator (uPA) and its receptor (uPAR): development of antagonists of uPA/uPAR interaction and their effects in vitro and in vivo. Curr Pharm Des 2003; 9: 1529-43. 7. Look MP, van Putten WLJ, Duffy MJ, Harbeck N, Christensen IJ, Thomssen C, et al. Pooled analysis of prognostic impact of uPA and PAI-1 in 8377 breast cancer patients. J Natl Cancer Inst 2002; 94: 116-28. 364 8. Jänicke F, Prechtl A, Thomssen C, Harbeck N, Meisner C, Untch M, et al. for the German chemo N0 study group. Randomyzed adjuvant chemotherapy trial in high-risk, lymph node-negative breast cancer patients identified by urokinase-type plasminogen activator and plasminogen activator inhibitor type 1. J Natl Cancer Inst 2001; 93: 913-20. 9. Kwaan HC, McMahon B. The role of plasminogen-plasmin system in cancer. Cancer Treat Res 2009; 148: 43-66. 10. Takac I, Cufer T, Gorisek B, Cas-Sikosek N, Bali R, Bosilj D, et al. [The role of the urokinase-type plasminogen activator (uPA) and its inhibitor (PAI-1) in breast cancer]. [Slovenian]. Zdrav Vestn 2011; 80: 405-12. 11. Duffy MJ, O’Grady P, Devaney D, O’Siorain L, Fennelly JJ, Lijnen HJ. Urokinase-plasminogen activator, a marker for aggressive breast carcino­mas. Preliminary report. Cancer 1988; 62: 531-3. 12. Jänicke F, Schmitt M, Ulm K, Gossner W, Graeff H. Urokinase-type plasmi­nogen activator antigen and early relapse in breast cancer. Lancet 1989; 2: 1049. 13. Foekens JA, Schmitt M, Van Putten WL, Peters HA, Bontenbal M, Jänicke F, et al. Prognostic value of urokinase-type plasminogen activator in 671 primary breast cancer patients. Cancer Res 1992; 52: 6101-5. 14. Foekens JA, Schmitt M, van Putten WL, Peters HA, Bontenbal M, Jänicke F, et al. Plasminogen activator inhibitor-1 and prognosis in primary breast cancer. J Clin Oncol 1994; 12: 1648-58. 15. Foekens JA, Peters HA, Look MP, Portengen H, Schmitt M, Kramer MD, et al. The urokinase system of plasminogen activation and prognosis in 2780 breast cancer patients. Cancer Res 2000; 60: 636-43. 16. Jelisavac-Cosic S, Sirotkovic-Skerlev M, Kulic A, Jakic-Razumovic J, Kovac Z, Vrbanec D. Prognostic significance of urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor (PAI-1) in patients with primary invasive ductal breast carcinoma - a 7.5-year follow-up study. Tumori 2011; 97: 532-9. 17. Eljuga D, Razumovic JJ, Bulic K, Petrovecki M, Draca N, Bulic SO. Prognostic importance of PAI-1 in node negative breast cancer patients - results after 10 years of follow up. Pathol Res Pract 2011; 207: 290-4. 18. Gazic B, Pizem J, Bracko M, Cufer T, Borstnar S, Pohar-Marinsek Z, et al. S-phase fraction determined on fine needle aspirates is an independent prognostic factor in breast cancer - a multivariate study of 770 patients. Cytopathology 2008; 19: 294-302. 19. Borstnar S, Sadikov A, Mozina B, Cufer T. High levels of uPA and PAI-1 predict a good response to anthracyclines. Breast Cancer Res Treat 2010; 121: 615-24. 20. Xu FJ, Stack S, Boyer C, O’Briant K, Whitaker R, Mills GB, et al. Heregulin and agonistic anti-p185(c-erbB2) antibodies inhibit proliferation but in­crease invasiveness of breast cancer cells that overexpress p185(c-erbB2): increased invasiveness may contribute to poor prognosis. Clin Cancer Res 1997; 3: 1629-34. 21. Ignatoski KM, Maehama T, Markwart SM, Dixon JE, Livant DL, Ethier SP. ERBB-2 overexpression confers PI 3' kinase-dependent invasion capacity on human mammary epithelial cells. Br J Cancer 2000; 82: 666-74. 22. Adam L, Vadlamudi R, Kondapaka SB, Chernoff J, Mendelsohn J, Kumar R. Heregulin regulates cytoskeletal reorganization and cell migration through the p21-activated kinase-1 via phosphatidylinositol-3 kinase. J Biol Chem 1998; 273: 28238-46. 23. Arboleda J, Slamon DJ. Heregulin induced cell invasion of HER-2/neu trans­fected breast cancer cell line is mediated through activation of the phos­phatidylinositol 3-kinase/AKT2 pathway. Cold Spring Harbor Laboratory Meeting on Tyrosine Phosphorylation and Cell Signaling; 1999 May 12.-16. p. 11. 24. Berney CR, Yang J, Fisher RJ, Russell PJ, Crowe PJ. Correlates of urokinase-type plasminogen activator in colorectal cancer: positive relationship with nm23 and c-erbB-2 protein expression. Oncol Res 1998; 10: 47-54. 25. Allgayer H, Babic R, Gruetzner KU, Tarabichi A, Schildberg FW, Heiss MM. c-erbB-2 is of independent prognostic relevance in gastric cancer and is associated with the expression of tumor-associated protease systems. J Clin Oncol 2000; 18: 2201-9. 26. Dano K, Andreasen PA, Grondahl-Hansen J, Kristensen P, Nielsen LS, Skriver L. Plasminogen activators, tissue degradation and cancer. Adv Cancer Res 1985; 44: 139-66. 27. Schmitt M, Harbeck N, Thomssen C, Wilhelm O, Magdolen V, Reuning U, et al. Clinical impact of the plasminogen activation system in tumor inva­sion and metastasis: prognostic relevance and target for therapy. Thromb Haemostasis 1997; 78: 285-96. 28. Zemzoum I, Kates RE, Ross JS, Dettmar P, Dutta M, Henrichs C, et al. Invasion factors uPA/PAI-1 and HER2 status provide independent and complemen­tary information on patient outcome in node-negative breast cancer. J Clin Oncol 2003; 21: 1022-8. 29. Konecny G, Untch M, Arboleda J, Wilson C, Kahlert S, Boettcher B, et al. HER-2/neu and urokinase-type plasminogen activator and its inhibitor in breast cancer. Clin Cancer Res 2001; 7: 2448-57. 30. Decker T, Hungermann D, Böcker W. [Prognostic and predictive factors of invasive breast cancer: update 2009]. [German]. Pathologe 2009; 30: 49-55. 31. Elston CW. Grading of invasive carcinoma of the breast. In: Page DL, Anderson TJ, eds. Diagnostic histopathology of the breast. Edinburgh: Churchill Livingstone; 1987. p. 300-11. 32. Harbeck N, Dettmar P, Thomssen C, Berger U, Ulm K, Kates R, et al. Risk-group discrimination in node-negative breast cancer using invasion and pro­liferation markers: six-year median follow-up. Br J Cancer 1999; 80: 419-26. 33. Schmitt M, Thomssen C, Ulm K, Seiderer A, Harbeck N, Höfler H, et al. Time-varying prognostic impact of tumour biological factors urokinase (uPA), PAI-1 and steroid hormone receptor status in primary breast cancer. Br J Cancer 1997; 76: 306-11. 34. Harbeck N, Thomssen C, Berger U, Ulm K, Kates RE, Höfler H, et al. Invasion marker PAI-1 remains a strong prognostic factor after long-term follow-up both for primary breast cancer and following first relapse. Breast Cancer Res Treat 1999; 54: 147-57. 35. Cufer T, Vrhovec I, Borštnar S. Prognostic significance of plasminogen ac­tivator inhibitor-1 in breast cancer, with special emphasis on locoregional recurrence-free survival. Int J Biol Mark 2002; 17: 33-41. 36. Cufer T, Vrhovec I, Borštnar S. Prognostic and predictive value of the urokinase-type plasminogen activator (uPA) and its inhibitor PAI-1 and PAI-2 in operable breast cancer. Int J Biol Mark 2003; 18: 106-15. 37. Cufer T. Clinical utility of serine proteases in breast cancer. Radiol Oncol 2004; 38: 85-92. 38. Ferrero-Poüs M, Hacene K, Bouchet C, Le Doussal V, Tubiana-Hulin M, Spyratos F. Relationship between c-erbB-2 and other tumor characteristics in breast cancer prognosis. Clin Cancer Res 2000; 6: 4745-54. 39. Descotes F, Riche B, Saez S, De Laroche G, Datchary J, Roy P, et al. Plasminogen activator inhibitor type 1 is the most significant of the usual tissue prognostic factors in node-negative breast ductal adenocarcinoma independent of urokinase-type plasminogen activator. Clin Breast Cancer 2008; 4: 168-77. 40. Harbeck N, Kates R, Schmitt M. Clinical relevance of invasion factors uroki­nase-type plasminogen activator and plasminogen activator inhibitor type 1 for individualized therapy decisions in primary breast cancer is greatest when used in combination. J Clin Oncol 2002; 20: 1000-7. 41. Esteva FJ, Hortobagyi GN. Prognostic markers in early breast cancer. Breast Cancer Res 2004; 6: 109-18. 42. De Cremoux P, Grandin L, Diéras V, Savignoni A, Degeorges A, Salmon R, et al. Breast Cancer Study Group of the Institut Curie. Urokinase-type plasminogen activator and plasminogen-activator-inhibitor type 1 predict metastases in good prognosis breast cancer patients. Anticancer Res 2009; 29: 1475-82. 43. Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, et al. Gene Expression Profiling in Breast Cancer: Understanding the Molecular Basis of Histologic Grade To Improve Prognosis. J Natl Cancer Inst 2006; 98: 262-72. 365 research article Prognostic value of some tumor markers in unresectable stage IV oropharyngeal carcinoma patients treated with concomitant radiochemotherapy Erika Soba1, Marjan Budihna1, Lojze Smid2, Nina Gale3, Hotimir Lesnicar1, Branko Zakotnik1, Primoz Strojan1 1 Institute of Oncology Ljubljana, Ljubljana, Slovenia 2 University Department of Otorhinolaryngology and Cervicofacial Surgery, University Clinical Center, Ljubljana, Slovenia 3 Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia Radiol Oncol 2015; 49(4): 365-370. Received 13 October 2014 Accepted 15 November 2014 Correspondence to: Prof. Marjan Budihna, M.D., Ph.D., Na gmajni 50, 1133 Ljubljana, Slovenia. Phone: +386 1 512 7594; Mobile: +386 41 384 704; Email: budihna.marjan@gmail.com Disclosure: No potential conflicts of interest were disclosed. Background. The aim of the study was to investigate how the expression of tumor markers p21, p27, p53, cyclin D1, EGFR, Ki-67, and CD31 influenced the outcome of advanced inoperable oropharyngeal carcinoma patients, treated with concomitant radiochemotherapy. Patients and methods. The pretreatment biopsy specimens of 74 consecutive patients with inoperable stage IV oropharyngeal squamous cell carcinoma treated with concomitant radiochemotherapy were in retrospective study processed by immunochemistry for p21, p27, p53, cyclin D1, EGFR, Ki-67, and CD31. Disease-free survival (DFS) was assessed according to the expression of tumor markers. Results. Patients with a high expression of p21 (.10%), p27 (>50%), Ki-67 (>50%), CD31 (>130 vessels/mm2) and low expression of p53 (<10%), cyclin D1 (<10%) and EGFR (<10%) (favorable levels - FL) had better DFS than patients with a low expression of p21 (<10%), p27 (.50%), Ki-67 (.50%), CD31 (<130 vessels/mm2) and high expression of p53 (.10%), cyclin D1 (.10%) and EGFR (.10%) (unfavorable levels - UL). However, statistical significance in survival between FL and UL was achieved only for p27 and cyclin D1. DFS significantly decreased with an increasing number of markers with an unfavorable level per tumor (1–4 vs. 5–7) (78% vs. 32%, respectively; p = 0.004). The number of markers per tumor with UL of expression retained prognostic significance also in multivariate analysis. Conclusions. Statistical significance in survival between FL and UL emerged only for p27 and cyclin D1. The number of markers per tumor with UL of expression was an independent prognostic factor for an adverse outcome. Key words: oropharynx; radiochemotherapy; tumor markers Introduction Prognostic evaluation of patients with unresect­able squamous cell carcinoma in the head and neck (SCCHN) is currently based on the perfor­mance status of the patient and the tumor stage. Recently, the presence of human papillomavirus (HPV) DNA in tumor cells has also been identified as a strong predictor of survival in patients with oropharyngeal primaries.1 However, these classi­cal prognostic factors often do not provide suffi­cient information for the selection of the optimal therapy. The changes in the genes or their products which can be traced biochemically or immunohis­tochemically may serve to estimate the aggressive­ness of the tumor and, consequently, to adapt treat­ment accordingly. On the basis of the favorable results of our pro­ 366 TABLE 1. Antibodies and preconditioning applied for immunohistochemistry p21 SX118 DAKO p27 1B4 Novocastra p53 DO7 DAKO Ciklin D1 P2D11F11 Novocastra EGFR H11 DAKO Ki-67 MIB1 DAKO CD31 JC/70A DAKO 1:10 1:10 1:50 1:10 1:10 1:20 1:15 MW*, 6 min, EDTA buffer, pH 8.0 cooling 10 min MW*, 6 min, EDTA buffer, pH 8.0 cooling 10 min MW*, 6 min, EDTA buffer, pH 8.0 cooling 10 min MW*, 6 min, EDTA buffer, pH 8.0 cooling 10 min Proteinase 1 (Ventana) 12 min MW*, 6 min, EDTA buffer, pH 8.0 cooling 10 min MW**, 7 min, 96°C, citr. buffer, pH 6.0 + Proteinase 1 (Ventana), 2 min EGFR = epidermal growth factor receptor; EDTA = ethylene-diamine-tetraacetic acid; MW* = common microwave oven; MW** = microwave oven Polar Patent spective randomized clinical study2, concomitant radiochemotherapy with mitomycin C and bleo­mycin was introduced in the 1990s as a routine treatment for patients with unresectable squamous cell carcinoma of the oropharynx (SCCOP) in our department. Because the treatment morbidity was rather severe also in those patients who were not cured3, the identification of patients from a ho­mogenous group (in regard to the primary tumor origin, stage and treatment), who will or will not respond to aggressive radiochemotherapy would help in sorting patients into various treatment programs of different intensity, to spare some of them from unnecessary toxicity. Therefore, the pri­mary aim of our study was to investigate how the expression of growth promoting (cyclin D1, epi­dermal growth factor receptor [EGFR], Ki-67) and growth suppressing (p21, p27, p53) tumor markers and CD31 in the primary tumor tissue influenced the outcome of patients with unresectable SCCOP. Patients and methods Patients The 95 consecutive patients with previously un­treated, technically inoperable SCCOP were treat­ ed with curative intent in the period 1991.1998 with irradiation and concomitant application of mitomycin C and bleomycin. To achieve maximal possible homogeneity of the studied group, 21 pa­tients with disease of UICC TNM stage III were excluded for further evaluation. Patients were ir­radiated five times weekly with one fraction of 2 Gy/day with a planned total dose of 66.70 Gy. The physical dose was converted into a biologically ef­fective dose (BED) according to the formula: BED = TD x (1 + d/./ß) – K x (Tt – Td), where TD = total tumor dose in Gy, Tt = total treatment time in days, K = daily dose equivalent of repopulation in units of Gy./ß per day (K = 0.6, and ./ß = 10), and Td = lag time in days to the onset of effective repopulation during the treatment (4). It was assumed that Td = 28 days. The chemotherapy regimen consisted of intra­muscular applications of bleomycin 5 mg twice a week with the planned dose being 70 mg and one application of mitomycin C 15 mg/m2 applied in­travenously after delivery of 9.10 Gy of irradia­tion. Radiotherapy was considered intensive if the BED was .65Gy10. Chemotherapy was considered intensive if the dose of mitomycin C was .14.1 mg/ m2 and of bleomycin .35mg. The whole treatment was considered intensive, if chemotherapy or ra­diotherapy or both were intensive. Methods Immunohistochemistry The pretreatment biopsy specimens of the pri­mary tumors were in retrospective clinical study processed by immunochemistry for p21, p27, p53, Cyclin D1, EGFR, Ki-67, and CD31. Immunohistochemistry was performed on 4 µm paraffin sections mounted on silicon-coated glass slides. The antibodies and preconditioning ap­plied for immunohistochemistry are presented in Table 1. To determine the level of expression of the test­ed markers, semi-quantitative scoring of immune reactivity was performed according to the percent­age of positivity in the tumor cells as follows: 0 = less than 10%, 1 = 10.50%, 2 = more than 50% of tu­mor cells with a positive reaction (for p21, p27, p53, Ki-67, cyclin D1 and EGFR). Microvascular density (MVD) was assessed quantitatively with the CD31 antibody. Stained microvessels were counted and expressed as the number of microvessels per mm2 in the areas of maximal neovascularization of the tumor stroma. Statistical methods Disease free survival (DFS) was defined as the time interval from the beginning of the treatment to the appearance of local and/or regional progression 367 TABLE 2. The distribution of patients according to the expression of tumor markers < 10% 10%–50% 20 26 22 23 28 9 31 21 13 15 13 33 Microvascular density in stroma: Median 118 (48–206) per mm2 < 130 (n = 40) > 50% 13 14 22 7 31 13 > 130 (n = 19) Total 59 59 59 59 59 59 59 EGFR = epidermal growth factor receptor and/or distant metastases. The survival curves were plotted by using the method of Kaplan-Meier5 and a log rank test was used to test the differences in survival between subgroups.6 Survival was calcu­lated in subgroups defined by the level of expres­sion of tumor markers, performance status (PS) and intensity of treatment. For multivariate analysis, a Cox proportional hazards model was used.7 Ethical consideration The study was carried out according to the Helsinki Declaration (1964, with later amendments) and of the European Council Convention on Protection of Human Rights in Bio-Medicine (Oviedo 1997). It was approved by the Institutional Review Board Committee and by the National Committee for Medical Ethics, Ministry of Health, Republic of Slovenia Results Because of severe mucositis, some patients could not be irradiated to the planned dose or receive the full dose of bleomycin. Some patients had mild liver damage and the dose of mitomycin had to be below the planned dose. So, the intensity of the treatment was not the same for all patients. Only in 59 out of 74 patients with UICC stage IV of SCCOP was there sufficient biopsy tissue in par­affin blocks for the analysis of all seven tumor mark­ers under investigation. Among them, there were 2 females and 57 males with a median age of 52 years (39–67 years). In 42 patients the WHO PS was 0, and in 17 it was assessed as 1–2. The treatment intensity was low in 13 patients and high in 46. The distribution of patients according to the expression of markers in the tumor tissue is pre­sented in Table 2 The median follow-up time of patients was 6 years (3–10 years). Twenty nine (49%) patients had a local, regional or distant relapse; at the last fol­low-up 15 (25%) were still alive and 44 (75%) were dead. The probability for DFS at five years was 48%. Intensive treatment (high vs. low) and good PS (0 vs. .1) were associated with statistically sig­nificantly better DFS: 59% vs. 9%, p = 0.000; 58% vs. 19%, p = 0.005, respectively. In the subgroup of in­tensively treated patients and good PS, DFS was 65 %, while for those with a poor performance status it was 30% (p = 0.08). Patients with a favorable expression profile (FL) of studied markers, i.e. a high expression of p21 (. 10%), p27 (> 50%), Ki-67 (> 50%), CD31 (> 130 ves­sels/mm2) and low expression of p53 (< 10%), cyc­lin D1 (< 10%) and EGFR (< 10%), had better DFS than patients with an unfavorable expression of these markers, i.e. p21 (< 10%), p27 (. 50%), Ki-67 (. 50%), CD31 (< 130 vessels/mm2) and a high expres­sion of p53 (. 10%), cyclinD1 (. 10%) and EGFR (. 10%). However, statistical significance in DFS between FL and UL was achieved only in the case of p27 and cyclin D1. DFS significantly decreased with an increasing number of markers with UL per tumor (1–4 vs. 5–7): 78% vs. 32%, p = 0.004 (Table 3, 368 TABLE 3. Disease-free survival at 5 years according to the expression of tumor treated with concomitant radiochemotherapy ismarkers scarce. Out of several known tumor markers we choose some growth promoting (cyclin D1, EGFR, Ki-67) and growth suppressing (p21, p27, p53) UL < 10% 20 34 markers, and CD31. In addition to PS, which is p21 0.204 FL . 10% 39 55 one of the stronger prognostic factor in different UL . 50% 45 40 p27 0.040 malignant diseases8, and treatment intensity, only FL > 50% 14 77 FL < 10% 28 59 the number of markers with FL of expression in a p53 0.177 UL . 10% 31 38 particular tumor and p27 (as an individual marker ) FL < 10% 31 66 Cyclin D1 0.020 were recognized as independent prognosticators UL . 10% 28 30 for DFS. FL < 10% 13 77 EGFR 0.093 UL . 10% 46 41 The published results on the value of these tumor UL . 50% 46 42 markers in SCCHN vary. For example, in the case Ki-67 0.131 FL > 50% 13 68 of p27, a negative cell-cycle regulator that blocks UL < 130 40 40 CD31* FL > 130 19 69 0.100 progression from late G1 to S phase9, its protein ex-Number of UL 1–4 23 78 pression was found to positively correlate with dis­ 0.004 per patient 1-4 vs 5-7 5–7 36 32 ease outcome in some studies9-12, although a nega-Number of UL 0–1 39 65 0.002 tive relationship was also described.13 Probably the only for P27 & cyclin D1 2 20 17 most widely studied marker is the nuclear tran-DFS = disease free survival; UL = unfavorable level of expression of tumor marker; FL = favorable scription factor p53, playing a role in the control level of expression of tumor marker; EGFR = pidermal growth factor receptor; n = number of patients; * in micro-vessels per mm2. of cell proliferation, apoptosis and maintenance of the fidelity of DNA duplication.14,15 The results of a meta-analysis on the role of upregulated p53 in Figure 1). Considering the expression of only p27 patients with SCCHN were inconclusive, mainly and cyclin D1, apart from other markers, DFS was due to large heterogeneity across the studies.16 significantly worse for those patients whose tumor Interestingly, in three rather homogeneous studies had unfavorable expression levels of both mark-with SCCOP17-19, meta-analysis indicates that p53 ers: (0–1 vs. 2): 65% vs. 17%, respectively (p = 0,002) overexpression/mutation confers a survival advan­(Table. 3). Intensively treated patients in poor PS tage.16 Discrepancies in the results of different stud-with FL of expression of p27 survived significantly ies can also be found in the case of p21 (G1-phase better than those with a low expression of p27 (75% blocker )20-23, cyclin D1 (promotes progression of vs. 0%, p = 0.017). In the case of cyclin D1, corre-cells throughout the cycle)24-26, Ki-67 (a measure of sponding analysis was not possible due to a small the proliferative capacity of the tumor )27-29, EGFR number of intensively treated patients with FL of (cell growth promoter )30,31, and CD31 (microvas­this marker. cular density indicator ).32-34 In our study, the HPV In a Cox proportional regression model, PS, testing was not performed. However, because 90% intensity of treatment and p27 retained statistical of the patients from our series were heavy smokers significance (HR = 2.363, 95% CI = 0.999–5.589, p = (i.e. with lifetime tobacco exposure of one pack of 0.050; HR = 2.550, 95% CI = 1.105–5.886, p = 0.028; cigarettes per day for .10 years) - a fact that nega-HR = 3.743, 95% CI =1.064–13.169, p = 0.040, re-tively influences immune system activity, which spectively) while cyclin D1 was marginally signifi-is crucial for the favorable outcome observed in cant (HR = 1.070, 95% CI = 1.000–1.145, p = 0.051). HPV-positive tumors - the tumor HPV status in Compared to p27, the number of all markers per our patients would be less likely to play a signifi­tumor with UL of expression had a stronger statis-cant role.35,36 tically significant influence on the prognosis (HR = Survival of our patients, who represent a rather 3.614, p = 0.009), along with the intensity of treat-homogeneous group regarding histology, primary ment (HR = 3.150, p = 0.005) and PS (HR = 2.352, tumor localization, stage, and treatment, depended p = 0.031). primarily on the intensity of the applied therapies, their PS and also on the biological characteristics of the tumor. The latter was determined by study-Discussion ing dysregulation in the expression of seven tumor markers, and was influenced also by several other Information on the prognostic value of different tu-pathologic processes taking place in the tumor, mor markers in patients with unresectable SCCOP, not considered in our study. It was found that a 369 low expression of p21, p27, Ki-67, CD31 and high expression of p53, cyclin D1 and EGFR negatively influenced DFS. In general, when analyzing sepa­rately the expression of each of the seven markers, the difference in their expression showed no sta­tistically significant correlation with survival prob­ability; the two exceptions were p27 and cyclin D1. In addition to the lack of prognostic potential as an intrinsic characteristic of an individual marker, another reason for negative results could be the low number of patients in our series. However, the influence of studied markers on survival increased above the level of statistical significance, when the sum of only those markers with UL per tumor was taken into account (Figure1). By ranking the pa­tients according to this criterion, we found a signif­icantly lower DFS in the group with the increased number of markers with UL of expression. In the multivariate analysis, the sum of UL of expression of markers per tumor remained an independent prognostic factor for DFS, along with PS and inten­sity of treatment. It appears that the prediction of the outcome of the disease on the basis of expres­sion of only one marker, even in a homogeneous group of patients, is not necessarily successful. The expression profiles of different genes are inter­dependent and none of the known tumor mark­ers can play independently inside this network. Accordingly, the expression level of a particular maker resulted from the sum of influences exerted by a variety of other markers and vice versa. This might also explain why the opinions on the prog­nostic value of individual markers in the literature differ to such a great extent. As expected, the survival of our patients with poor PS was low. Among them we tried to identify those who did not benefit from rather toxic con­comitant radiochemotherapy. Intensively treated poor PS patients with . 4 UL markers and those who had FL of expression of one or both of p27 and cyclin D1 had comparable DFS (67% and 60% respectively) to intensively treated patients with good PS. On the other hand, in spite of intensive treatment, patients with poor PS with > 4 UL mark­ers and those with UL of expression of one or both of p27 and cyclin D1 had poor DFS (17% and 0% respectively). It seems that these patients should not be treated that vigorously and are candidates for palliative treatment programs. In conclusion, in a series of 59 SCCOP patients, uniformly treated with concomitant radiochemo­therapy with mitomycin C and bleomycin, a set of seven markers, determined immunohistochemical­ly, was recognized as a significant predictor of DFS only when the number of markers per tumor with UL of expression was considered. Of the individ­ual markers, only p27 correlated with survival on multivariate analysis, in addition to the PS of the patients and the intensity of the applied therapies. Despite intensive treatment, poor PS patients with > 4 markers with UL of expression as well as those with UL of expression of p27 and cyclin D1, had unfavorable survival rates: these patients should be treated with palliative intent. References 1. Genden EM, Sambur IM, de Almeida JR. Human papillomavirus and oro­pharyngeal squamous cell carcinoma: what the clinician should know. Eur Arch Otorhinolaryngol 2013; 270: 405-16. 2. Smid L, Lesnicar H, Zakotnik B, Soba E, Budihna M, Furlan L, et al. Radiotherapy, combined with simultaneous chemotherapy wit h mitomycin C and bleomycin for inoperable head and neck cancer - preliminary report. Int J Radiat Oncol Biol Phys 1995; 32: 769-75. 3. Budihna M, Šoba E, Šmid L, Zakotnik B, Strojan P, Čemažar M, et al. Inoperable oropharyngeal carcinoma treated with concomitant irradia­tion, mitomycin C and bleomycin - long term results. Neoplasma 2005; 52: 165-74. 4. Fowler, JF. The linear quadratic formula and progress in fractionated radio­therapy. Br J Radiol 1998; 62: 679-94. 5. Kaplan EL, Meier P. Nonparametric estimation from incomplete observa­tions. JASA 1958; 53: 457-89. 6. Peto R, Pike MC, Armitage P, Breslow NE, Cox DR, Howard SV, et al. Design and analysis of randomized clinical trials requiring prolonged observation of each patient. II. Analysis and examples. Br J Cancer 1977; 35: 1-39. 7. Cox DR. Regression models and life tables. J Royal Stat Soc 1972; 34: 187­ 220. 8. Kovac V, Zwitter M, Zagar T. Improved survival after introduction of chemo­therapy for malignant pleural mesothelioma in Slovenia: Population-based survey of 444 patients. Radiol Oncol 2012; 46: 136-44. 9. Kudo Y, Takata T, Yasui W, Ogawa I, Miyauchi M, Takekoshi T, et al. Reduced expression of cyclin-dependent kinase inhibitor p27Kip1 is an indicator of malignant behaviour in oral squamous cell carcinoma. Cancer 1998; 83: 2447-55. 10. Shimada M, Kitagawa K, Dobashi Y, Isobe T, Hattori T, Uchida C, et al. High expression of Pirh2, an E3 ligase for p27, is associated with low expression of p27 and poor prognosis in head and neck cancers. Cancer Sci 2009; 100: 866-72. 11. Moreno-Galindo C, Hermsen M, García-Pedrero JM, Fresno MF, Suárez C, Rodrigo JP. p27 and BCL2 expression predicts response to chemotherapy in head and neck squamous cell carcinomas. Oral Oncol 2014; 50: 128-34. 12. Fillies T, Woltering M, Brandt B, Van Diest JP, Werkmeister R, Joos U, et al. Cell cycle regulating proteins p21 and p27 in prognosis of oral squamous cell carcinomas. Oncol Rep 2007; 17: 355-9. 13. Zhang M, Li J, Wang L, Tian Z, Zhang P, Xu Q , et al. Prognostic significance of p21, p27 and survivin protein expression in patients with oral squamous cell carcinoma. Oncol Lett 2013; 6: 381-6. 14. O‘Rourke RW, Miller CW, Kato GJ, Simon KJ, Chen DL, Dang CV, et al. A potential transcriptional activation element in the p53 protein. Oncogene 1990; 5: 1829-32. 15. Wu X, Zhong D, Lin B, Zhai W, Ding Z, Wu J. p38 MAPK regulates the expres­sion of ether a go-go potassium channel in human osteosarcoma cells. Radiol Oncol 2013; 47: 42-9. 16. Tandon S, Tudur-Smith C, Riley RD, Boyd MT, Jones TM. A systematic review of p53 as a prognostic factor of survival in squamous cell carcinoma of the four main anatomical subsites of the head and neck. Cancer Epidemiol Biomarkers Prev 2010; 19: 574-87. 370 17. Awwad S, Jaros E, Somes J, Lunec J. P53 overexpression in head and neck carcinoma and radiotherapy results. Int J Radiat Oncol Biol Phys 1996; 34: 323-32. 18. Grabenbauer GG, Muhlfriedel C, Roedel F, Niedobitek G, Hornung J, Roedel C, et al. Squamous cell carcinoma of the oropharynx: Ki-67 and p53 can identify patients at high risk for local recurrence after surgery and postop­erative radiotherapy. Int J Radiat Oncol Biol Phys 2000; 48: 1041-50. 19. Otero-Garcia J E, Youssef E, Enamorado I, Du W, Yoo G H, Merati K, et al. Prognostic significance of p53 and FHIT in advanced oropharyngeal carci­noma Am J Otolaryngol 2004; 25: 231-9. 20. Ribeiro DA, Nascimento FD, Fracalossi ACC, Gomes TS, Oshima CT, Franco MF. Expression profiling of cell cycle regulatory proteins in oropharyngeal carcinomas using tissue microarrays. In Vivo 2010; 24: 315-20. 21. Erber R, Klein W, Andl T, Enders C, Born AI, Conradt C, et al. Aberrant p21 (CIP1/WAF1) protein accumulation in head-and-neck cancer. Int J Cancer.1997; 74: 383-9. 22. Hafkamp HC, Mooren JJ, Claessen SM, Klingenberg B, Voogd AC, Bot FJ, et al. P21 Cip1/WAF1 expression is strongly associated with HPV-positive tonsillar carcinoma and a favorable prognosis. Mod Pathol 2009; 22: 686-98. 23. Chung YL, Lee MY, Horng C, Jian JJ, Cheng SH, Tsai SY, et al. Use of combined molecular biomarkers for prediction of clinical outcomes in locally advanced tonsillar cancers treated with chemoradiotherapy alone. Head Neck 2009; 3: 9-20. 24. Perisanidis C, Perisanidis B, Wrba F, Brandstetter A, El Gazzar S, Papadogeorgakis N, et al. Evaluation of immunohistochemical expression of p53, p21, p27, cyclin D1, and Ki67 in oral and oropharyngeal squamous cell carcinoma. J Oral Pathol Med 2012; 41: 40-6. 25. Yu Z, Paul PM,. Haffty BG, Sasaki C, Zerillo C, Joe J, et al. Cyclin D1 is a valuable prognostic marker in oropharyngeal squamous cell carcinoma. Clin Cancer Res 2005; 11: 1160-6. 26. Rahimi AS, Wilson DD, Saylor DK, Stelow EB, Thomas CY, Reibel JF, et al. p16, cyclin D1, and HIF-1. predict outcomes of patients with oropharyn­geal squamous cell carcinoma treated with definitive intensity-modulated radiation therapy. Int J Otolaryngol. 2012 (2012); Article ID 685951, 9 p. doi: 10.1155/2012/685951. 27. Koelbl O, Rosenwald A, Haberl M, Müller J, Reuther J, Flentje M. P53 and KI-67 as predictive markers for radiosensitivity in squamous cell carcinoma of the oral cavity. An immunohistochemical and clinicopathologic study. Int J Radiat Oncol Biol Phys 2001; 49: 147-54. 28. Watanabe S, Watanabe R, Oton-Leite AF, Alencar Rde C, Oliveira JC, Leles CR, et al. Analysis of cell proliferation and pattern of invasion in oral squa­mous cell carcinoma. J Oral Sci 2010; 52: 417-24. 29. Pereira CS, Oliveira MV, Fraga CA, Barros LO, Domingos PL, Roy A, et al. Impact of the epithelial dysplasia grading and Ki67 proliferation index in the adjacent non-malignant mucosa on recurrence and survival in head and neck squamous cell carcinoma. Pathol Res Pract 2012; 208: 651-6. 30. Ang KK, Berkey BA, Tu X, Zhang HZ, Katz R, Hammond EH, et al. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res 2002; 62: 7350-6. 31. Semrau R, Duerbaum H, Temming S, Huebbers C, Stenner M, Drebber U, et al. Prognostic impact of human papillomavirus status, survivin, and epidermal growth factor receptor expression on survival in patients treated with radiochemotherapy for very advanced nonresectable oropharyngeal cancer. Head Neck 2013; 35: 1339-44. 32. Ataman OU, Bentzen SM, Wilson GD, Daley FM, Richman PI, Saunders MI, et al. Molecular biomarkers and site of first recurrence after radiotherapy for head and neck cancer. Eur J Cancer 2004; 40: 2734-41. 33. Chuang HC, Su CY, Huang HY, Chien CY, Chen CM, Huang CC. High Expression of CD105 as a prognostic predictor of early tongue cancer. Laryngoscope 2006; 116: 1175-9. 34. Foote RL, Weidner N, Harris J, Hammond E, Lewis JE, Vuong T, et al. Evaluation of tumor angiogenesis measured with microvessel density (MVD) as a prognostic indicator in nasopharyngeal carcinoma: results of RTOG 9505. Int J Radiat Oncol Biol Phys 2005; 61: 745-53. 35. AngKK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tan PF, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 2010; 363: 24-35. 36. Spanos WC, Nowicki P, Lee DW, Hoover A, Hostager B, Gupta A, et al. Immune response during therapy with cisplatin of radiation for human papillomavirus related head and neck cancer. Arch Otolaryngol Head Neck Surg 2009; 135: 1137-46. 371 research article Clinical results of proton beam therapy for twenty older patients with esophageal cancer Takashi Ono1, Tatsuya Nakamura1, Yusuke Azami1, Hisashi Yamaguchi1, Yuichiro Hayashi1, Motohisa Suzuki1, Yoshiomi Hatayama1, Iwao Tsukiyama1, Masato Hareyama1, Yasuhiro Kikuchi1, Kenji Nemoto2 1 Department of Radiation Oncology, Southern Tohoku Proton Therapy Center, Koriyama, Fukushima, Japan 2 Department of Radiation Oncology, Yamagata University Faculty of Medicine, Yamagata, Japan Radiol Oncol 2015; 49(4): 371-378. Received 15 March 2015 Accepted 20 June 2015 Correspondence to: Takashi Ono, Department of Radiation Oncology, Southern Tohoku Proton Therapy Center, 7-172, Yatsuyamada, Koriyama, Fukushima, 963-8052, Japan. Phone: +81-24-934-3888; Fax: +81-24-934-5393, E-mail: abc1123513@gmail.com. Disclosure: No potential conflicts of interest were disclosed. Background. In an aging society, increasing number of older patients are diagnosed with esophageal cancer. The purpose of this study was to assess the clinical efficacy and safety of proton beam therapy for older patients with esophageal cancer. Patients and methods. Older patients (age: . 65 years) newly diagnosed with esophageal cancer between January 2009 and June 2013 were enrolled in this study. All patients underwent either proton beam therapy alone or proton beam therapy with initial X-ray irradiation. Toxicities were evaluated using the Common Terminology Criteria for Adverse Events version 4.0. Results. Twenty patients were eligible for this study and all completed the treatment. The median age was 78 years (range: 65–89 years) and the median follow-up time was 26.5 months (range: 6–62 months). Seven patients had lymph node metastases and 10 had stage II/III cancer. The median dose of proton beam therapy was 72.6 Gy relative bio­logical dose effectiveness (RBE) (range: 66–74.8 Gy [RBE]) for proton beam therapy alone and 33 Gy (RBE) (range: 30.8–39.6 Gy [RBE]; total dose range: 66.8–75.6 Gy [RBE]) for proton beam therapy with initial X-ray irradiation. The 2-year overall survival rate was 81.8% (95% confidence interval [CI]: 62.4%–100%), and the 2-year local control rate was 89.4% (95% CI: 75.5%–100%). Grade 2 or 3 toxicities occurred in some cases; however, no grade 4 or 5 toxicity was observed. Conclusions. High-dose (66–75.6 Gy [RBE]) proton beam therapy without chemotherapy was an efficacious and safe treatment for older patients with esophageal cancer. Key words: proton therapy; aged; esophageal neoplasms; radiotherapy Introduction Esophageal cancer is the sixth leading cause of cancer death and the eighth most common cancer worldwide.1 In eastern Asia, esophageal cancer is the fourth most common cause of cancer death.2 Surgery remains the main treatment choice for resectable esophageal cancer. However, follow­ing reports from the Radiation Therapy Oncology Group3,4 and studies of the efficacy of chemoradio­therapy (CRT)5-7, CRT has become another choice for the treatment of esophageal cancer. In an aging society, an increasing number of older patients are diagnosed with esophageal can­cer. Not all of these patients can be treated with CRT or surgery because of their age, general con­dition and/or complications, although there are some reports regarding the use of CRT or surgery in older patients with esophageal cancer.8,9 Other studies have reported the efficacy of radiotherapy alone for older patients.9,10 However, compared with CRT or surgery, X-ray irradiation alone has not shown satisfactory results for the treatment of esophageal cancer.3,9 372 New radiotherapy treatments, such as intensity-modulated radiotherapy and proton beam therapy (PBT), deliver concentrated doses to the target vol­ume, avoiding the organs at risk.11-13 These thera­pies may thus be suitable for treating older patients with esophageal cancer. Despite the increased use of PBT for esophageal cancer14-18, few data are avail­able regarding the efficacy of PBT in older patients with esophageal cancer. In this study, we treated older patients with esophageal cancer using PBT without chemotherapy. We retrospectively evalu­ated the efficacy and safety of PBT in these older patients. Patients and methods Patients Patients newly diagnosed with esophageal cancer treated with PBT without chemotherapy between January 2009 and June 2013 at the Southern Tohoku Proton Therapy Center were recruited from our database retrospectively. All patients were his­tologically confirmed to have esophageal cancer based on a biopsy before each treatment. Every patient was assessed, and the clinical stage of es­ophageal cancer was determined using endoscopy, computed tomography (CT) and positron emission tomography (PET)-CT. Written informed consent was obtained from all patients and the investiga­tors followed recommendations of the Helsinki Declaration. The inclusion criteria were as follows: a histo­logically confirmed diagnosis of esophageal can­cer, age of . 65 years, World Health Organization performance status of 0–2 and no distant organ me­tastasis or other sites of uncontrolled cancer. Proton beam therapy Treatment planning for PBT was based on three-di­mensional CT images taken at 2 mm intervals in the exhalation phase while using a respiratory gating system (Anzai Medical, Tokyo, Japan). The gross tumor volume (GTV) included the primary tumor and lymph node metastases. The primary tumor volume was determined from markers implanted using endoscopy at the cranial and caudal ends of the tumor. Lymph nodes over 1.0 cm in the short axis or exhibiting a high 18F-fluorodeoxyglucose uptake on PET-CT were considered metastases. The clinical target volume (CTV) was defined as GTV plus longitudinal margins of . 2.5 cm and lateral margins of 0.5 cm. The planning target volume (PTV) was CTV plus 0.5 cm margins. The daily PBT fraction was 2.2 Gy relative biological dose effectiveness (RBE). Proton energy levels of 150 MeV or 210 MeV for 1–2 portals, and spread- out Bragg peak were tuned as much as possible until the PTV was exposed to a 90% isodose of the prescribed dose (Figure 1). The PBT system at our institute (Proton beam system, Mitsubishi, Tokyo, Japan) used synchrotron, and scattering methods. Treatment was administered during the exhalation phase using a respiratory gating system. Daily front and lateral X-ray imaging was used for positioning. The PBT schedule was 33.0 Gy (RBE) in 15 fractions over 3 weeks in the combination therapy group and 72.6 Gy (RBE) in 33 fractions over 7 weeks in the PBT-only group. The PBT dose was modulated ap­propriately considering the response of the primary tumor as determined using endoscopy and PET-CT images. If the reduction in the maximal diameter of the primary tumor was < 50%, 1 to 3 fractions of PBT were performed without replanning. On the other hand, PBT was stopped without administer­ 373 ing 1 to 3 fractions if the degree of tumor reduction TABLE 1. Patient characteristics was adequate and the patient had esophageal ulcer. Initial X-ray irradiation Treatment planning for X-ray irradiation was also based on three-dimensional CT images taken at 2.5 mm intervals. The patients received PBT along with the initial X-ray irradiation (combination ther­apy) if they had . T2 disease or lymph node me­tastases. However, patients were treated with PBT without initial X-ray irradiation if they had severe cardiopulmonary complications, such as intersti­tial pneumonitis or myocardial infarction, their performance status was 2 or they refused X-ray ir­radiation. The cephalic and caudal borders of the initial X-ray irradiation fields included the bilateral supr­aclavicular nodes and cephalic plexus for thoracic or abdominal esophageal cancer. For cephalic es­ophageal cancer, we irradiated the region from the laryngopharynx to the carina. 10-MV X-ray irradia­tion was used with anteroposterior fields. The field number was generally two, whereas three fields were used for the field within a field technique when there was a large hot area. The daily X-ray irradiation fraction was 1.8 Gy, and the irradiation schedule was 36.0 Gy in 20 fractions delivered over 4 weeks. Evaluation and follow-up All patients underwent endoscopy and PET to evaluate the initial tumor response within three months of the completion of treatment. The follow-up interval was every 2–3 months for the first year and every 3–6 months thereafter. Endoscopy, CT and PET-CT were performed if necessary. Complete response was defined as the complete disappearance of all detectable tumors, partial response was defined as a . 50% reduction in the maximal diameter of the tumor and stable disease was defined as no decreases or increases in the tu­mor diameter. Progressive disease was defined as enlargement of the primary tumor or the appear­ance of new lesions, including lymph node and distant metastases. Toxicities were evaluated us­ing the Common Terminology Criteria for Adverse Events version 4.0.19 Statistical analysis The statistical tests were performed using the IBM SPSS Statistics version 22 software package (SPSS Age (years) Median 78 Range 65–89 65–69 2 (10%) 70–74 5 (25%) 75–79 6 (30%) 80–89 7 (35%) Gender Male 14 (70%) Female 6 (30%) Performance status 0 7 (35%) 1 11 (55%) 2 2 (10%) Follow up time (months) Median 26.5 Range 6–62 T category* T1 8 (40%) T2 5 (25%) T3 6 (30%) T4 1 (5%) N category* N0 13 (65%) N1 4 (20%) N2 3 (15%) Stage* I 10 (50%) II 5 (25%) III 5 (25%) Tumor location Cervical 3 (15%) Upper thoracic 4 (15%) Mid thoracic 9 (45%) Lower thoracic 4 (25%) Histopathology Squamous cell 19 (95%) carcinoma Adenocarcinoma 1 (5%) Proton dose in PBT with initial X-ray irradiation (n=9) (Gy (RBE)) Median 33.0 (total dose: 69.0) 30.8–39.6 (total dose: Range 66.8–75.6) Proton dose in PBT alone (n=11) (Gy (RBE)) Median 72.6 Range 66.0–74.8 PBT = proton beam therapy; RBE = relative biological dose effectiveness; * Numbers correspond to the tumor-node-metastasis system of classification (International Union Against Cancer criteria) 374 FIGURE 3. (A) Overall survival rate of the patients with stage I and II/III esophageal cancer. The 2-year overall survival rate was statistically different between the two groups (p = 0.041). (B) Overall survival rate of the patients in T 1/2 and T 3/4. The 2-year overall survival rate was statistically different between the two groups (p = 0.010). (C) Overall survival rate of the patients receiving proton beam therapy alone or proton beam therapy with initial X-ray irradiation. The 2-year overall survival rate was not statistically different between the two groups (p = 0.890). NS = not significant; PBT = proton beam therapy Inc., Chicago, IL, USA). The overall survival (OS) time was defined as the time between the start of treatment and the last follow-up. The local control time was defined as the time between the start of treatment and the date on which tumor recurrence was found or the last follow-up. The Kaplan–Meier method and log-rank test were applied to estimate survival probabilities and compare the survival rates, respectively. Results Patients Twenty-six older patients were treated for esopha­geal cancer using PBT with or without initial X-ray irradiation between January 2009 and June 2013. All of these subjects were treated without any con­current treatments, including chemotherapy. Of these 26 patients, 2 were excluded from the anal­ysis because of distant metastasis and 4 were ex­cluded for uncontrolled cancer at other sites. The characteristics of the remaining 20 patients, includ­ ing 9 with inoperable cancer, are summarized in Table 1. All 20 patients completed their treatment. The cohort comprised 14 men and 6 women, with a median age of 78 years (range: 65–89 years). The median follow-up time was 26.5 months (range: 6–62 months). Comorbidities included interstitial pneumonitis owing to collagen disease (2 patients), chronic obstructive pulmonary disease (3 patients), myocardial infarction (5 patients), chronic heart failure (3 patients), chronic renal failure (2 patients) and diabetes mellitus (1 patient). Lymph node me­tastasis was present in 7 patients, and 10 patients had stage II/III cancer. Eleven patients were treated with PBT alone, and 9 patients were treated with combination therapy. The median dose of PBT FIGURE 4. Local control rate for the patients with esophageal cancer after proton beam therapy. The 2-year local control rate was 89%. 375 TABLE 2. Toxicities Esophagitis 3 (15%) 3 (15%) 14 (70%) 0 0 0 Esophageal ulcer 13 (65%) 0 6 (30%) 1 (5%) 0 0 Esophageal stenosis 18 (90%) 0 2 (10%) 0 0 0 Esophageal fistula 19 (95%) 0 1 (5%) 0 0 0 Pneumonitis 6 (30%) 11 (55%) 2 (10%) 1 (5%) 0 0 Pleural effusion 12 (60%) 6 (30%) 2 (10%) 0 0 0 Pericardial effusion 17 (85%) 3 (15%) 0 0 0 0 was 72.6 Gy (RBE) (range: 66.0–74.8 Gy (RBE)) for PBT alone and 33.0 Gy (RBE) (range: 30.8–39.6 Gy (RBE)) for the combination therapy. With regard to the dose of X-ray irradiation, all patients received 36.0 Gy (RBE), except for one patient who received 32.4 Gy. Survival and local control All patients were followed for at least 13 months or until death. Six patients died, 4 from esopha­geal cancer and 2 from other causes (1 from bac­terial pneumonia and 1 from another cancer ). The 1- and 2-year OS rates were 90.0% (95% confidence interval [CI]: 76.9%–100%) and 81.8% (95% CI: 62.4%–100%), respectively (Figure 2). There was a significant difference in the 2-year OS rate between the patients with stage I (100%) and stage II/III (60.0%) cancers (p = 0.041) (Figure 3A). There was also a significant difference in the 2-year OS rate between the patients with T category 1/2 (100%) and T category 3/4 (47.6%) (p = 0.010) lesions (Figure 3B). On the other hand, the OS rate with or without initial X-ray irradiation was not signifi­ cantly different (p = 0.890) (Figure 3C). Seventeen (85%) patients achieved a complete response and 3 (15%) achieved a partial response. Three patients (1 treated with PBT alone and 2 treated with the com­bination therapy) had local recurrence. The 2-year local control rate was 89.4% (95% CI: 75.5%–100%) (Figure 4). Failure patterns Seven patients had recurrence. One patient had lymph node recurrence within the PBT field, 3 had distant metastases and 3 had local recurrence. There were no primary tumors or sites of lymph node recurrence outside the irradiation field in the PBT-only group. Toxicities There were no grade 4 or 5 toxicities after treatment (Table 2). Of the 20 patients, 6 (30%) had grade 2 esophageal ulcers, 2 (10%) had grade 2 pneumoni­tis and 2 (10%) had grade 2 pleural effusion. One patient (5%) with an esophageal ulcer required in­travenous hyperalimentation (grade 3 esophageal ulcer ), and the ulcer healed one month later. Two patients (10%) with esophageal stenosis were treat­ed with dilation using endoscopy (grade 2 esopha­geal stenosis). One of these patients developed an esophageal fistula (grade 2 esophageal fistula) just after dilation and was treated with the insertion of a stent in the esophagus. Neither patient required surgery. One patient (5%) with pneumonitis was treated with oxygenation and steroid administra­tion two years after PBT because of dyspnea (grade 3 pneumonitis). In that case, the dyspnea was re­lieved 3 days later, and the dose of steroids was gradually reduced. Discussion We herein demonstrated that PBT without chemo­therapy is efficacious and safe for the treatment of older patients with esophageal cancer. To the best of our knowledge, this is the first report on the use of PBT without chemotherapy in older patients with esophageal cancer. Radiotherapy alone is one choice for treating older patients with esophageal cancer who cannot receive CRT or surgery. Kawashima et al.10 report­ed the results of 66 Gy X-ray irradiation without chemotherapy in 51 older patients with no lymph node metastasis: the 1- and 2-year OS rates were 71% and 53%, respectively. Additionally, Cooper et al.3 reported 1- and 2-year OS rates after 64 Gy radiotherapy alone of 34% and 10%, respectively, 376 TABLE 3. Previous results of radiation therapy without chemotherapy for esophageal cancer and our result Nemoto et al., 200020 78 T1 N0 65.5 Gy 88% 73% Kawashima et al., 51 T1–3 N0 66 Gy 71% 53% 200610 Cooper et al., 19993 62 T1–3 N0–1 64 Gy 34% 10% Smith et al., 20099 623 T1–4 N0–1 none 16% 7% 69.0 Gy (RBE) (PBT with X-ray) Oto et al., 2015 20 T1–4 N0–2 90% 81.8% 72.6 Gy (RBE) (PBT only) OS = overall survival; PBT = proton beam therapy; RBE = relative biological dose effectiveness Smith et al.9 reported 1-year and 2-year OS rates for older esophageal cancer patients treated with X-ray irradiation alone of 16% and 7%, respectively and Nemoto et al.20 reported 1- and 2-year OS rates after radiotherapy alone (median total dose: 65.5 Gy) for superficial esophageal cancer (stage I) of 88% and 73%, respectively. Our results, including those for the 7 patients with lymph node metastases, showed superior 1- and 2-year OS rates to those observed in these studies (Table 3). These results may differ because we were able to administer higher CTV doses with less exposure to organs at risk, such as the lungs and heart, although a previous report in­dicated that higher doses do not improve outcomes in cases of CRT.4 Studies of esophageal cancer treated with CRT have reported 5-year OS rates of 11%–75.7%; these cohorts included older patients.3,5,6,8 In a compara­tive study of CRT versus surgery alone for the treatment of esophageal cancer, Ariga et al.5 report­ed a 3-year OS rate of 69.1% for CRT patients in stage II/III and 47.9% for surgery patients in stage II/III; for patients with stage I cancer, the 2-year OS rate was 100% in the CRT group and 90% in the surgery alone group. Ishikura et al.7 reported long-term toxicities after CRT in a study of 139 patients, with grade 4 or 5 esophagitis (7 patients), pneu­monitis (4 patients) and pericardial effusion (1 pa­tient). Our results showed an equivalent 2-year OS rate for patients with stage I cancer but an inferior OS rate for patients with stage II/III cancer. This result suggests that PBT without chemotherapy is sufficient for treating stage I esophageal can­cer, although patients with stage II/III esophageal cancer have a higher OS when treated with con­comitant chemotherapy. However, older patients receiving platinum-based chemotherapy develop significantly more grade 3–5 toxicities than young­er patients.21,22 In addition, patients treated with CRT for esophageal cancer experience more life-threatening acute toxicities than those treated with radiotherapy alone (CRT: 8%; radiotherapy alone: 2%).3 Higher grade toxicities are more common in patients receiving concomitant CRT, particularly older patients. Therefore, the administration of concomitant chemotherapy is not possible in all older patients; PBT without chemotherapy may be a feasible treatment choice for older patients with stage II/III esophageal cancer, particularly for older patients who have cardiopulmonary comorbidi­ties, renal failure or a bad performance status. No broad consensus has been established re­garding the optimal CTV protocol for elective nodal irradiation in cases of esophageal cancer. Zhao et al.23 evaluated the results of late-course ac­celerated hyperfractionated involved-field confor­mal radiotherapy for locally advanced esophageal squamous cell carcinoma, reporting OS rates of 77% at 1 year and 56% at 2 years, although both T4 and N1 patients were included. In addition, the rate of out-field node recurrence alone was only 8%. Similarly, Kawaguchi et al.24 observed a rate of out-field lymph node recurrence alone of 11%. Ji et al.25 reported that lymph nodes located near esophageal cancer lesions receive considerable in­cidental doses of irradiation to the involved field, which may eliminate subclinical lesions. Zhang et al.26 reported the results of involved-field irradia­tion for esophageal cancer, including patients with lymph node metastasis (73.4%); they observed that the rate of out-field lymph node recurrence was as high as 30%. In our study, we observed no re­currence outside the irradiation field in patients receiving PBT alone and found no significant differences in the OS rates between the patients treated with PBT alone and those treated with the combination therapy. These results suggest that in­volved-field irradiation is a sufficient treatment for 377 esophageal cancer without lymph node metastasis. Furthermore, PBT has advantages over other treat­ments for esophageal cancer because higher radia­tion doses can be administered without increasing the toxicity. High radiation doses reportedly do not improve the OS rate in cases of CRT4; however, the optimal dose for radiotherapy alone has not been deter­mined. The OS rate may increase if patients receive a higher dose of radiation. In Japan, even older patients receive 66 Gy for radiotherapy alone10; most patients receiving PBT can tolerate doses higher than 66 Gy (RBE). In the current study, our patients who underwent PBT experienced no severe or fatal toxicities within the follow-up pe­riod, although they received doses higher than 66 Gy. There were 2 patients who had esophageal stenosis in the current study, however, the esopha­geal stenosis was severe in both cases because of esophageal cancer before starting treatment and the stenosis remained even after they achieved a complete response. Kawashima et al.10 reported that 3 (5.9%) patients receiving 66 Gy X-ray irra­diation developed grade 5 pneumonitis within 90 days of the start of radiotherapy. Mizumoto et al.15 reported the results of PBT (median total dose of combined X-ray and proton beam: 80.0 Gy (RBE); median dose of PBT alone: 79.0 Gy (RBE) without chemotherapy for locally advanced T1-4 N0/1 M0 esophageal cancer. The only toxicity observed was non-healing ulcers in 4 (8%) patients. These results suggest that, compared with X-ray therapy alone (dose: > 60 Gy (RBE)) or PBT (dose: > 80 Gy (RBE)), PBT is a safe and feasible treatment for esophageal cancer when the dose is 66.0 to 75.6 Gy (RBE). We used initial X-ray irradiation for elective nodal irradiation, because the available field size of PBT at our institute is 15 cm × 15 cm. Some re­searchers have also reported using combination therapy.14,15 When the patients received the initial X-ray irradiation at our institute, PBT was per­formed as shown in Figure 1. Although the proton beam was stopped when it reached a location close to the spinal cord (Figure 1A), the irradiation dose for the spinal cord was adequately reduced and no patients with radiation myelopathy were observed as of the last follow-up. On the other hand, the lung regions received a high dose (Figure 1B), however, the irradiation dose for the lung of PBT was less than the oblique opposed X-ray irradiation follow­ing anteroposterior irradiation for elective nodal irradiation. As a result, we think that combination therapy is therefore a practical and safe technique for treatment with esophageal cancer. There are two limitations associated with this study. First, the number of patients was very small and we only included patients from a single institu­tion. However, the current study revealed the high 1- and 2-year OS rates with following survivors for at least 13 months. Second, the follow-up time was short, as we started using PBT only in 2008. Therefore, longer follow-up is needed to ascertain the long-term OS rate and toxicities. The high 1- and 2-year OS rates with acceptable toxicity observed in this study indicated that high-dose 66.0–75.6 Gy (RBE) PBT without chemothera­py was an efficacious and safe treatment for older patients with esophageal cancer. References 1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 2010; 127: 2893-917. 2. Soerjomataram I, Lortet-Tieulent J, Parkin DM, Ferlay J, Mathers C, Forman D, et al. Global burden of cancer in 2008: a systematic analysis of disability-adjusted life-years in 12 world regions. Lancet 2012; 380: 1840-50. 3. Cooper JS, Guo MD, Herskovic A, Macdonald JS, Martenson JA Jr, Al-Sarraf M, et al. Chemoradiotherapy of locally advanced esophageal cancer: long-term follow-up of a prospective randomized trial (RTOG 85-01). Radiation Therapy Oncology Group. JAMA 1999; 281: 1623-7. 4. Minsky BD, Pajak TF, Ginsberg RJ, Pisansky TM, Martenson J, Komaki R, et al. INT 0123 (Radiation Therapy Oncology Group 94-05) phase III trial of combined-modality therapy for esophageal cancer: high-dose versus stand­ard-dose radiation therapy. J Clin Oncol 2002; 20: 1167-74. 5. Ariga H, Nemoto K, Miyazaki S, Yoshioka T, Ogawa Y, Sakayauchi T, et al. Prospective comparison of surgery alone and chemoradiotherapy with selective surgery in resectable squamous cell carcinoma of the esophagus. Int J Radiat Oncol Biol Phys 2009; 75: 348-56. 6. Nishimura Y, Mitsumori M, Hiraoka M, Koike R, Nakamatsu K, Kawamura M, et al. A randomized phase II study of cisplatin/5-FU concurrent chemo­radiotherapy for esophageal cancer: Short-term infusion versus protracted infusion chemotherapy (KROSG0101/JROSG021). Radiother Oncol 2009; 92: 260-5. 7. Ishikura S, Nihei K, Ohtsu A, Boku N, Hironaka S, Mera K, et al. Long-term toxicity after definitive chemoradiotherapy for squamous cell carcinoma of the thoracic esophagus. J Clin Oncol 2003; 21: 2697-702. 8. Uno T, Isobe K, Kawakami H, Ueno N, Kobayashi H, Shimada H, et al. Efficacy and toxicities of concurrent chemoradiation for elderly patients with es­ophageal cancer. Anticancer Res 2004; 24: 2483-6. 9. Smith GL, Smith BD, Buchholz TA, Liao Z, Jeter M, Swisher SG, et al. Patterns of care and locoregional treatment outcomes in older esophageal cancer patients: The SEER-Medicare Cohort. Int J Radiat Oncol Biol Phys 2009; 74: 482-9. 10. Kawashima M, Kagami Y, Toita T, Uno T, Sugiyama M, Tamura Y, et al. Prospective trial of radiotherapy for patients 80 years of age or older with squamous cell carcinoma of the thoracic esophagus. Int J Radiat Oncol Biol Phys 2006; 64: 1112-21. 11. Isacsson U, Lennernäs B, Grusell E, Jung B, Montelius A, Glimelius B. Comparative treatment planning between proton and x-ray therapy in esophageal cancer. Int J Radiat Oncol Biol Phys 1998; 41: 441-50. 12. Zhang X, Zhao KL, Guerrero TM, McGuire SE, Yaremko B, Komaki R, et al. Four-dimensional computed tomography-based treatment planning for intensity-modulated radiation therapy and proton therapy for distal esopha­geal cancer. Int J Radiat Oncol Biol Phys 2008; 72: 278-87. 378 13. Pan X, Zhang X, Li Y, Mohan R, Liao Z. Impact of using different four-dimen­sional computed tomography data sets to design proton treatment plans for distal esophageal cancer. Int J Radiat Oncol Biol Phys 2009; 73: 601-9. 14. Sugahara S, Tokuuye K, Okumura T, Nakahara A, Saida Y, Kagei K, et al. Clinical results of proton beam therapy for cancer of the esophagus. Int J Radiat Oncol Biol Phys 2005; 61: 76-84. 15. Mizumoto M, Sugahara S, Nakayama H, Hashii H, Nakahara A, Terashima H, et al. Clinical results of proton-beam therapy for locoregionally advanced esophageal cancer. Strahlenther Onkol 2010; 186: 482-8. 16. Shohei K, Hirohiko T. Proton Beam Therapy with High-Dose Irradiation for Superficial and Advanced Esophageal Carcinomas. Clin Cancer Res 2003; 9: 3571-7. 17. Mizumoto M, Sugahara S, Okumura T, Hashimoto T, Oshiro Y, Fukumitsu N, et al. Hyperfractionated concomitant boost proton beam therapy for esophageal carcinoma. Int J Radiat Oncol Biol Phys 2011; 81: e601-6. 18. Lin SH, Komaki R, Liao Z, Wei C, Myles B, Guo X, et al. Proton beam therapy and concurrent chemotherapy for esophageal cancer. Int J Radiat Oncol Biol Phys 2012; 83: e345-51. 19. Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0. Published May 28, 2009; Revised Version 4.03 June 14, 2010. U. S. Department of Health and Human Sevices [citated 2015 Mar 14]. Available at http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_ QuickReference_8.5x11.pdf 20. Nemoto K, Matsumoto Y, Yamakawa M, Jo S, Ito Y, Oguchi M, et al. Treatment of superficial esophageal cancer by external radiation therapy alone: results of a multi-institutional experience. Int J Radiat Oncol Biol Phys 2000; 46: 921-5. 21. Blanchard EM, Moon J, Hesketh PJ, Kelly K, Wozniak AJ, Crowley J, et al. Comparison of platinum-based chemotherapy in patients older and young­er than 70 years: an analysis of Southwest Oncology Group Trials 9308 and 9509. J Thorac Oncol 2011; 6: 115-20. 22. Liu M, Shi X, Guo X, Yao W, Liu Y, Zhao K, et al. Long-term outcome of ir­radiation with or without chemotherapy for esophageal squamous cell carcinoma: a final report on a prospective trial. Radiat Oncol 2012; 7: 142. 23. Zhao KL, Ma JB, Liu G, Wu KL, Shi XH, Jiang GL. Three-dimensional conformal radiation therapy for esophageal squamous cell carcinoma: is elective nodal irradiation necessary? Int J Radiat Oncol Biol Phys 2010; 76: 446-51. 24. Kawaguchi Y, Nishiyama K, Miyagi K, Suzuki O, Ito Y, Nakamura S. Patterns of failure associated with involved field radiotherapy in patients with clini­cal stage I thoracic esophageal cancer. Jpn J Clin Oncol 2011; 41: 1007-12. 25. Ji K, Zhao L, Yang C, Meng M, Wang P. Three-dimensional conformal radia­tion for esophageal squamous cell carcinoma with involved-field irradiation may deliver considerable doses of incidental nodal irradiation. Radiat Oncol 2012; 7: 200. 26. Zhang X, Li M, Meng X, Kong L, Zhang Y, Wei G, et al. Involved-field irra­diation in definitive chemoradiotherapy for locally advanced esophageal squamous cell carcinoma. Radiat Oncol 2014; 9: 64. 379 research article Cutaneous melanoma frequencies and seasonal trend in 20 years of observation of a population characterised by excessive sun exposure Serena Bonin1, Antonio Albano1, Nicola di Meo1, Alessandro Gatti2, Giuseppe Stinco3, Fabrizio Zanconati4, Giusto Trevisan1,2 1 Department of Medical Sciences, Unit of Dermatology, University of Trieste, Trieste, Italy 2 Unit of Dermatology; Azienda Ospedaliero-Universitaria Ospedali Riuniti di Trieste; Ospedale Maggiore, Trieste, Italy 3 Department of Experimental and Clinical Medicine, Institute of Dermatology, University of Udine, Italy 4 Department of Medical Sciences, Unit of Surgical Pathology, University of Trieste, Trieste, Italy Radiol Oncol 2015; 49(4): 379-385. Received 9 June 2015 Accepted 8 August 2015 Correspondence to: Serena Bonin, Ph.D., DSM Depatment of Medical Sciences, University of Trieste, University Hospital of Cattinara, Surgical Pathology Blg, Strada di Fiume 447, 34149 Trieste, Italy. Phone: +39 399 6266; Fax: +39 3996247; E-mail: sbonin@units.it Disclosure: No potential conflicts of interest were disclosed. Background. Cutaneous melanoma is an aggressive form of skin cancer. It has become an increasingly common neoplasm in the most developed countries, especially among individuals of European origin. Patients and methods. Anonymous data of patients with cutaneous melanoma were collected from the diag­nostic database of the University Hospital of Trieste from 1 January 1990 to 10 December 2013. Our study is based on a population which was constant over the period of observation; it was also well-defined and characterised by unrestrained sun exposure. Results. The number of cutaneous melanomas increased during the period of observation with a seasonality trend and gender related differences both for anatomical sites distribution and stage of the disease. Moreover, 6% of our cohort developed multiple melanomas. Conclusions. In a well-defined population devoted to excessive sun exposure the frequencies of skin melanomas roughly doubled from 1990 to 2013 following a seasonal trend. In that population, prevention efforts according to gender specific risk behaviour, as well as follow-up programmes both for evaluation of metastatic spreading and for early diagnosis of additional skin melanomas, are crucial due to gender specific differences and to the occurrence of multiple melanomas. Key words: cutaneous melanoma; sun exposure; frequencies; multiple melanomas; gender related differences Introduction Cutaneous melanoma has become an increasingly common neoplasm in most developed countries, especially among individuals of European origin.1,2 Different patterns referred to patients’ sex and age have been observed worldwide; most recent esti­mates indicate wide North–South and East–West variation of melanoma incidence in Europe, with the lowest rates in Southern and Eastern countries.3 The main possible reason for the general increas­ing melanoma incidence over the last 40 years is greater exposure of pale Caucasian skin to natural ultraviolet (UV) radiation.2 Epidemiological stud­ies suggest a relationship between suntan habits and high risk of melanoma. Sun exposure is highly prevalent in all age groups, especially among the young; it is influenced by certain convictions and attitudes towards suntan, and it is stimulated by peer pressure and beauty reasons. Although the 380 TABLE 1. Frequencies by age range tic database of the University Hospital of Trieste. Inclusion criteria were a diagnosis of cutaneous melanoma and to be resident in the Italian prov­ince of Trieste. Data include gender, age at diag- In situ 20-29 30-39 40-49 50-59 60-69 70-79 80+ nosis, date of diagnosis and anatomical site of Women melanoma onset. Data were collected separately 1990-1995 0 3 4 6 5 1 4 1 for in situ and invasive melanoma from 1 January 1996-2001 2002-2007 0 0 2 1 8 8 13 12 7 5 11 10 9 3 1 1 1990 to 10 December 2013. For patients submitted to BRAF mutation analysis for therapeutic issues also BRAF mutational status was retained. For 2008-2013 1 9 16 25 17 13 11 4 survival and analyses related to melanoma thick- Men ness, histological type and disease progression, we 1990-1995 0 0 0 3 6 0 4 0 retained those patients who were followed up by 1996-2001 1 1 1 4 9 4 11 1 the Dermatology unit of the University Hospital 2002-2007 2008-2013 0 0 0 1 2 13 5 22 2 19 5 16 2 21 1 3 of Cattinara. Clinical data criteria as well as re­sults are reported in the Supplementary file. The study was conducted according to the Declaration Total in situ 2 17 68 90 70 60 65 12 of Helsinki protocols. Moreover, clinical data were Invasive 0-19 20-29 30-39 40-49 50-59 60-69 70-79 80+ available only for patients who signed an informed Women consent for research use of their data. 1990-1995 4 10 14 17 28 21 20 13 1996-2001 0 11 20 19 36 31 29 17 Statistical analyses 2002-2007 0 9 21 35 30 35 24 23 The distribution of the clinical, histological, and epi­ 2008-2013 0 7 28 38 36 62 39 29 demiological categorical variables was compared by Men the chi-squared test. ANOVA and t-test were per­ 1990-1995 3 5 11 12 21 30 26 17 formed for continuous variables depending on the 1996-2001 2002-2007 0 0 3 2 10 8 16 21 35 35 32 54 40 42 20 34 number of categories. Seasonality test was evalu­ated by means of the Walter and Elwood test.8 To estimate trend across groups for non-parametric 2008-2013 1 6 16 34 39 71 78 38 data the Cuzick’s test was used. All p-values are Total invasive Overall (%) 8 10 (1) 53 70 (4) 112 180 (10) 192 282 (15) 260 330 (18) 336 396 (21) 298 363 (20) 191 203 (11) two-sided with values <0.05 regarded as statistically significant. Statistical analyses were performed with the Stata/SE 12 package (Stata, College Station, TX). general public is now aware that sunlight exposure has been leading to increased risk of skin cancer over the decades, most people still believe that a tanned person looks healthier.4 Moreover, also en­vironmental risks referred to climate changes5 and increased reporting of in situ melanomas6 seem to contribute to the rise of melanoma rates. The aim of this study is to analyse the frequen­cies and characterise melanoma patients in one of the Italian cities with the highest incidence of cuta­neous melanoma7 and whose inhabitants sunbathe excessively. Patients and methods Anonymous data from patients with cutane­ous melanoma were collected from the diagnos- Results Frequencies From 1990 to 2013, 1834 patients had a diagnosis of cutaneous melanoma. They were all Caucasian residents of the province of Trieste between 14 and 98 years of age. Data on in situ as well as invasive cutaneous melanoma by age group and gender are reported in Table 1. Overall, mean age at diagnosis was 59 years (± 16.3 years) with 50% gender distri­bution. Age at diagnosis for men was significantly higher (average age 62 years) than for women (av­erage age 57 years) (p < 0.001). The number of mela­noma patients has increased over the years both for in situ and invasive melanomas and that number was more pronounced in the last 6 years (Figure 1). The diagnosis of melanoma was more frequent for middle-aged and older patients, although an incre­ 381 ment for younger patients (aged < 40) over years has been observed. When we divided the 24 years of observation into 4 periods of 6 years, we did not find any vari­ation of age at diagnosis over the years (p = 0.1), even by sex stratification (p = 0.06 for men and p = 0.5 for women). However for invasive melano­mas a significant increment of age at diagnosis was observed over the 4 intervals of observation (p = 0.02) (Figure 2). This information was main­tained for men (p = 0.05), but not for women (p = 0.3) (Figure 2). Age at diagnosis for men increased significantly from 61 (± 18 years) years to 64 (± 15 years) years. Age at diagnosis for in situ melano­mas did not vary across years (p = 0.3), for both genders (p = 0.6 for men and p = 0.4 for women). Anatomical sites The distribution of primary cutaneous melanomas by anatomical sites is reported in detail in Table 2. Hereafter only significant results are reported. Overall, melanomas of the trunk were more fre­quent in men (54%) than women (35%) (p < 0.001). This result was maintained considering separately invasive and in situ melanomas (p < 0.001 for both); therefore 53% of men and 36% of women developed an invasive melanoma of the trunk. With regard to in situ neoplasm the proportion of melanoma of the trunk was 60% for men and 32% for women. Melanomas of the head and neck were also more frequent in men (p = 0.003), who had that localiza­tion in 12% of cases compared to 8% of women. The distribution of invasive melanomas of head and neck was different between genders (p = 0.02), al­though it was comparable for in situ (p = 0.07). The percentage of melanomas of the lower limbs was higher in women (29% vs. 10%) (p < 0.001), both for invasive melanomas (p < 0.001) and in situ ones (p < 0.001) (Table 2). Invasive melanomas of hands and foot were more frequent in women (p = 0.05) with a propor­tion of 4.4% of cases in comparison to 2.5% of men; this observation was not confirmed for in situ mela­noma (p = 0.7). When we divided the 24 years of observation into 4 periods of 6 years the proportions of melanomas by anatomical sites remained essentially the same (p = 0.1), both for men (p = 0.2) and women (p = 0.4). Seasonality The overall monthly diagnosis of invasive melano­mas showed significant excess from the cyclic vari- FIGURE 1. Frequencies of cutaneous melanoma over years of observation: (A) invasive melanomas; (B) in situ melanomas; (C) invasive and in situ melanomas. ation with the maximum around the month of June (represented by q = 171°) and another lower peak in October (p < 0.001) (Figure 3). The same pat­tern was present for males (q = 162°- early June, p < 0.001) and females (q = 181°- late June, p = 0.0002) (Figure 3). No statistically significant cyclic trend was evidenced for in situ melanomas (p = 0.2), ei­ther for men (p = 0.5) or women (p = 0.3). According to the anatomical site of invasive mel­anoma onset, head and neck did not show cyclic trend (p=0.3), but all the other sites did so: invasive melanomas of the hands and foot peaked in late May (q = 147°, p = 0.002), those that were developed on the trunk and lower limbs peaked in June (q = 157°, p = 0.03; q = 178°, p = 0.0001, respectively) and invasive melanomas of the upper limbs peaked in July (q = 205°, p = 0.004). 382 TABLE 2. Distribution of melanomas by sex and anatomical sites, percentage is included in brackets In situ melanoma Head and neck 10 (4.7) 15 (9.6) 25 (6.8) Hands and feet 2 (1.0) 2 (1.3) 4 (1.1) Back Chest Trunk (back+chest) 21 (10.0) 48 (22.7) 69 (32.7) 35 (22.3) 60 (38.2) 95 (60.5) 56 (15.2) 108 (29.3) 164 (44.5) Upper limbs 50 (23.7) 20 (12.7) 70 (19.0) Lower limbs 64 (30.3) 18 (11.5) 82 (22.3) NOS 16 (7.6) 7 (4.5) 23 (6.3) Total 211 (100) 157 (100) 368 (100) Invasive melanoma Head and neck 60 (8.5) 92 (12.1) 152 (10.4) Hands and feet 31 (4.4) 19 (2.5) 50 (3.4) Back Chest Trunk (back+chest) 86 (12.2) 165 (23.4) 251 (35.6) 156 (20.5) 244 (32.1) 400 (52.6) 242 (16.5) 409 (27.9) 651 (44.4) Upper limbs 107 (15.2) 125 (16.4) 232 (15.8) Lower limbs 201 (28.5) 74 (9.7) 275 (18.8) Other 2 (0.3) 0 (0.0) 2 (0.1) NOS 54 (7.6) 50 (6.6) 104 (7.1) Total 706 (100) 760 (100) 1466 (100) NOS = not otherwise specified Multiple melanomas In our cohort 112 patients developed multiple mel­anomas: 26 of them had synchronous melanomas and 86 had metachronous melanomas. Synchronous With respect to synchronous melanomas all pa­tients, except one, had diagnosis of in situ melano­mas for both neoplasms. There was no difference between genders for age at diagnosis. Distribution of anatomical sites was significantly different between genders (p = 0.04), mainly because melanomas of the lower limbs were more frequent among women (p = 0.03; 21% vs. 0); the other anatomical sites were similarly distributed between genders (p = 0.3). No patient belonging to this group of multiple melanomas de­veloped the neoplasm on hands and foot. Metachronous Of the 86 patients who developed metachronous melanomas 53 were males and 33 were females, therefore men developed multiple metachronous melanomas (p = 0.02) more frequently. The appear­ance of the second melanoma did not follow any seasonal trend (p = 0.07). Most patients had a di­agnosis of invasive melanomas for both neoplasms. On average, the second primary melanoma devel­oped 3.7 years after the first one (min-max = 0–17 years), without any significant difference in time of appearance among genders (p = 0.8) and anatomical sites of the primary and secondary melanomas (p = 0.8 and p = 0.5, respectively). Overall, the location of primary melanomas was not significantly different between genders (p = 0.07), although a higher fre­quency of primary melanomas of the trunk was ob­served for men (62% vs. 33%, p = 0.02) and a higher frequency of primary melanoma of the lower limbs was observed in women (24% vs. 9%, p = 0.05) in agreement with data obtained from the entire co­hort. In 30 patients (35%) the second melanoma de­veloped in the same anatomical region as the first one, more frequently on the back (40%) and lower limbs (23%). The site of second primary melanomas differed between genders (p = 0.03) with a preva­lence of the melanoma of the trunk (62% vs. 36%; p = 0.01) for men and lower limbs for women (27% vs. 4%; p = 0.002). Breslow’s depth of primary mela­nomas was significantly higher than that of second­ary melanomas (p = 0.001) (Figure 4). Similarly, the mean number of mitoses of primary melanomas was higher than that of the second ones (1.5 vs. 0.6, p = 0.01) and on average the stage of the first lesion was also higher than the second one (p = 0.01). BRAF mutational status: BRAF mutational status was assessed in 40 patients for therapeutic issues due to disease progression. Of those 25 had mutations at BRAF gene while 15 were wild type (see supplementary file for details). Patients with mutant BRAF gene were significantly younger (average age 52 years) than those with wild type BRAF melanoma (average age 64 years; p = 0.03), in particular this observation was con­firmed in females (p = 0.04), but not in males (p = 0.2) as shown in supplementary Table 2. BRAF mutational status was also related to ana­tomical sites (p = 0.03): in our sub-group of patients BRAF mutations prevail in melanoma of trunk compared to melanoma of the hand and foot that were all wild type for BRAF. Clinical data and cancer specific survival Elaboration of clinical data and cancer specific sur­vival did not add anything new to cutaneous mela­ 383 noma research, because they confirm already pub­lished evidence. Therefore, they are reported in the supplementary file. Discussion This is a population-based study referred on data collection from cutaneous melanoma patients resi­dent in Trieste, a seaside town of about 250.000 residents in North-eastern Italy. This population is stable and well-defined, and it was constant over the years of observation.9 Residents in this area are mainly fair skinned and blue eyes because of their Celtic.10 and Austro-Hungarian origins. The pecu­liarity of the inhabitants of this town referred to cutaneous melanoma is their unrestrained sun ex­posure, mainly for traditional and cultural reasons. For that particular reason any increment in cutane­ous melanoma frequencies over the years is mainly due to environmental changes and suntan habits. The frequencies of cutaneous melanoma retrieved in this study are in agreement with the data report­ed by the Cancer registry of Friuli-Venezia Giulia region.11 The number of cutaneous melanoma in­creased during the period of observation, showing higher rates particularly for middle-aged and for elderly residents, who probably had not used any sun protection in their childhood and adult life, as reported by other authors.12 During our period of observation, a steep increment in cutaneous mela­noma cases occurred; in addition in the last six years cutaneous melanoma frequencies doubled in comparison to 1990-1995 in a homogeneous and stable population. That increment may be due in part to improved registration of melanoma as well as to over-diagnosis12, but it’s unlikely that the fre­quency growth could be solely ascribed to those factors. Although there are no data supporting this hypothesis, a possible explanation could be the cu­mulative effect of air pollution and sun exposure, since the air pollution index has been correlated with skin cancer.13 Nonetheless, other individual factors such as the use of artificial sunbed, cosmet­ics including sunscreen, photosensitising drugs, and exogenous hormones could be additional risk factors for the development of cutaneous melano­ma.14 In our cohort seasonality of cutaneous mela­noma diagnosis was detected with a higher peak in June probably as a consequence of increased patient awareness and self-detection of suspected lesions due to summer clothing.15 The presence of a lower seasonality peak in October could find a reasonable possible explanation in the effect of in­tense sun exposure on the visibility of melanocytic proliferation after intense ultraviolet exposure. Consequently, in summer the highlighted pig­mented lesions may alert the patients themselves or the physician.15 No seasonality has been detect­ed for in situ lesions in agreement with Asken et al. as different explanations seem to work for invasive and in situ melanomas.12 Six percent of our cohort developed multiple melanomas, which are not uncommon in cutane­ous melanoma patients.16 In agreement with Savoia et al. in our series of patients there are no differ­ences in clinical characteristics or histopathological features of the first cutaneous melanoma between patients with single or multiple metachronous 384 FIGURE 3. Seasonality of invasive cutaneous melanoma. FIGURE 4. Box plot representing the Breslow’s depth variation between 1st and 2nd cutaneous melanoma in patients with me­tachronous melanomas. Median values are reported in boxes melanomas; the distribution of the first melanoma sites also follows the same pattern as single mel­anomas.16 The significant decrease in the mean Breslow’s thickness as well as in the number of mitoses and stage for the second metachronous melanoma is mainly due to the follow-up in those patients and to their increased awareness of pig­mented lesion after having a melanoma.16 Even for early melanoma lesions the importance of sched­uled and well defined follow-up procedures was stressed. Regarding melanomas mutated at BRAF gene we observed a correlation between BRAF muta­tions and being young at the time of primary mela­noma diagnosis in agreement with other authors17, and our observation was confirmed particularly in women. BRAF mutations seem to be associated also to anatomical sites of the primary lesion, with melanomas on the trunk presenting higher rate of mutations at BRAF gene, as already shown.18 Gender-related differences in the anatomical distribution and stage of cutaneous melanoma were found in our cohort. At diagnosis women present with thinner lesions and show up be­fore men as shown by the lower age at diagnosis and lower Breslow’s depth for female (Results in Supplementary file). Women show in situ or stage I lesions, while men have stage II and lo­cally advanced cutaneous melanoma as shown by an Austrian report which found similar results.19 Moreover, as already pointed out20, cutaneous mel­anoma predominated at lower limb and hand-foot for women and trunk for men. In our cohort inva­sive melanomas of head and neck were significant­ly more frequent in men as observed in England after the early 1990s.21 In agreement with others22 men also tended to develop multiple metachro­nous melanomas more frequently. Overall those observations underline that pre­vention efforts should be increased taking into account gender-specific risk behaviour. As it was shown in the female population, less aggressive cutaneous melanoma are most likely diagnosed due to prevention. Conclusions In a well-defined population excessively exposing to sunlight the frequencies of cutaneous melanoma have roughly doubled from 1990 to 2013. In that population gender specific differences as well as a seasonality trend have been observed, stressing the importance of prevention efforts taking into ac­count gender-specific risk behaviour. The fact that multiple melanomas are not really uncommon, especially in men, highlights the need for follow-up programmes not only for evaluation of metastatic spread but also for early diagnosis of additional cutaneous melanoma. Acknowledgements The authors would like to thank Dr. Valentina Melita for her professional assistance in the lan­guage revision of the manuscript and Dr Davide Brunetti for his critical review. References 1. Jemal A, Saraiya M, Patel P, Cherala SS, Barnholtz-Sloan J, Kim J, et al. Recent trends in cutaneous melanoma incidence and death rates in the United States, 1992-2006. J Am Acad Dermatol 2011; 65(5 Suppl 1): S17-25 e1-3. 385 2. MacKie RM, Hauschild A, Eggermont AM. Epidemiology of invasive cutane­ous melanoma. Ann Oncol 2009; 20(Suppl 6): vi1-7. 3. Forsea AM, Del Marmol V, de Vries E, Bailey EE, Geller AC. Melanoma inci­dence and mortality in Europe: new estimates, persistent disparities. Br J Dermatol 2012; 167: 1124-30. 4. Bolanca Z, Bolanca I, Buljan M, Blajic I, Penavic Zeljko J, Situm M. Trends, habits and attitudes towards suntanning. Coll Antropol 2008; 32(Suppl 2): 143-6. 5. Diffey B. Climate change, ozone depletion and the impact on ultraviolet exposure of human skin. Phys Med Biol 2004; 49: R1-11. 6. Mocellin S, Nitti D. Cutaneous melanoma in situ: translational evidence from a large population-based study. Oncologist 2011; 16: 896-903. 7. Melanoma. In Veneto la maggiore incidenza. Ma e Torino la citta italiana dove ci si ammala di piu: QuotidianoSanita.it; 2013. Available from: http://www.quotidianosanita.it/scienza-e-farmaci/articolo.php?articolo_ id=17059. 8. Walter SD, Elwood JM. A test for seasonality of events with a variable popu­lation at risk. Br J Prev Soc Med. 1975; 29: 18-21. 9. TuttItalia. Statistiche demografiche: popolazione provincia di Trieste 2001­2013. Available from: http://www.tuttitalia.it/friuli-venezia-giulia/provincia­di-trieste/statistiche/popolazione-andamento-demografico/. 10. The Celtic Tribes of Roman Gaul. Available from: http://www.celtnet.org.uk/ gaulish-tribes.html. 11. Serraino D, Bidoli E, De Paoli A, de Dottori M, Zucchetto A, Zanier L. Cancer in Friulia venezia Giulia incidence, survival and prevalence: Udine: Agenzia Regionale della Sanita; 2009. 12. Akslen LA, Hartveit F. Cutaneous melanoma - season and invasion? A pre­liminary report. Acta Derm Venereol 1988; 68: 390-4. 13. Grant WB. Air pollution in relation to U.S. cancer mortality rates: an eco­logical study; likely role of carbonaceous aerosols and polycyclic aromatic hydrocarbons. Anticancer Res 2009; 29: 3537-45. 14. Volkovova K, Bilanicova D, Bartonova A, Letasiova S, Dusinska M. Associations between environmental factors and incidence of cutaneous melanoma. Environ Health 2012; 11(Suppl 1): S12. 15. Crocetti E, Guzzinati S, Paci E, Falcini F, Zanetti R, Vercelli M, et al. Strong seasonality in the diagnosis of skin melanoma in Italy: the Italian Network of Cancer Registries (AIRTUM) study. Tumori 2009; 95: 665-8. 16. Savoia P, Osella-Abate S, Deboli T, Marenco F, Stroppiana E, Novelli M, et al. Clinical and prognostic reports from 270 patients with multiple primary melanomas: a 34-year single-institution study. J Eur Acad Dermatol Venereol 2012; 26: 882-8. 17. Pracht M, Mogha A, Lespagnol A, Fautrel A, Mouchet N, Le Gall F, et al. Prognostic and predictive values of oncogenic BRAF, NRAS, c-KIT and MITF in cutaneous and mucous melanoma. J Eur Acad Dermatol Venereol 2015; 29: 1530-8. 18. Bauer J, Buttner P, Murali R, Okamoto I, Kolaitis NA, Landi MT, et al. BRAF mutations in cutaneous melanoma are independently associated with age, anatomic site of the primary tumor, and the degree of solar elastosis at the primary tumor site. Pigment Cell Melanoma Res 2011; 24: 345-51. 19. Duschek N, Skvara H, Kittler H, Delir G, Fink A, Pinkowicz A, et al. Melanoma epidemiology of Austria reveals gender-related differences. Eur J Dermatol 2013; 23: 872-8. 20. Chevalier V, Barbe C, Le Clainche A, Arnoult G, Bernard P, Hibon E, et al. Comparison of anatomical locations of cutaneous melanoma in men and women: a population-based study in France. Br J Dermatol 2014; 171: 595­ 601. 21. Wallingford SC, Alston RD, Birch JM, Green AC. Increases in invasive mela­noma in England, 1979-2006, by anatomical site. Br J Dermatol 2011; 165: 859-64. 22. Slingluff CL Jr, Vollmer RT, Seigler HF. Multiple primary melanoma: incidence and risk factors in 283 patients. Surgery 1993; 113: 330-9. 386 research article Release of growth factors after mechanical and chemical pleurodesis for treatment of malignant pleural effusion: a randomized control study Aljaz Hojski1, Maja Leitgeb2, Anton Crnjac1 1 Department of Thoracic Surgery, University Medical Centre Maribor, Slovenia 2 Biochemistry Division, Medical Faculty, University of Maribor, Maribor Slovenia Radiol Oncol 2015; 49(4): 386-394. Received 16 October 2014 Accepted 12 November 2014 Correspondence to: Prof. Anton Crnjac, M.D., Ph.D., Department of Thoracic Surgery, University Medical Centre Maribor, Ljubljanska 5, SI-2000 Maribor, Slovenia. Phone: +386 2 3211413, Fax: +386 2 3211417; E-mail: anton.crnjac@ukc-mb.si Disclosure: No potential conflicts of interest were disclosed. Background. Growth factors are key inducers of fibrosis but can also mediate inflammatory responses resulting in in­creasing pleural effusion and acute respiratory distress syndrome. The primary aim of the study was to analyse growth factors release after performing chemical and mechanical pleurodesis in the first 48 hours at the patients with malig­nant pleural effusion. The secondary endpoints were to evaluate the effectiveness of the both pleurodeses, symptoms release and the quality of life of patients after the treatment. Patients and methods. A prospective randomized study included 36 consecutive female patients with breast car­cinoma and malignant pleural effusion in an intention-to-treat analysis. We treated 18 patients by means of thoraco­scopic mechanical pleurodesis and 18 patients by chemical pleurodesis with talcum applied over a chest tube. We gathered the pleural fluid and serum samples in the following 48 hours under a dedicated protocol and tested them for growth factors levels. A quality of life and visual analogue pain score surveys were also performed. Results. Median measured serum vascular endothelial growth factor (VEGF) level after chemical pleurodesis was 930.68 pg/ml (95% CI: 388.22–4656.65) and after mechanical pleurodesis 808.54 pg/ml. (95% CI: 463.20-1235.13) (p = 0.103). Median pleural levels of transforming growth factor (TGF) ß1 were higher after performing mechanical pleu­rodesis (4814.00 pg/ml [95% CI: 2726.51–7292.94]) when compared to those after performing chemical pleurodesis (1976.50 pg/ml [95% CI: 1659.82–5136.26]) (p = 0.078). We observed similar results for fibroblast growth factor (FGF) ß; the serum level was higher after mechanical pleurodesis (30.45 pg/ml [95% CI: 20.40–59.42]), compared to those after chemical pleurodesis (13.39 pg/ml [95% CI: 5.04 – 74.60]) (p = 0.076). Mechanical pleurodesis was equally effective as chemical pleurodesis in terms of hospital stay, pleural effusion re-accumulation, requiring of additional thoracentesis, median overall survival, but, it shortened the mean thoracic drainage duration (p = 0.030) and resulted in a higher symptoms release and in a better quality of life (p = 0.047). Conclusions. We recorded an increase in serum VEGF levels after chemical pleurodesis, however on the contrary, an increase in the pleural fluid level of TGFß1 and FGFß] after mechanical pleurodesis with respect to compared group. Although the differences did not reach statistical significance, VEGF, TGFß1 and FGFß remain the most inter­esting parameters for future research. Considering the mechanisms of growth factors action, we conclude that in our study group mechanical pleurodesis might be more efficient in terms of growth factors release, thoracic drainage duration and resulted in a higher symptoms release and in a better quality of life than chemical pleurodesis. Key words: malignant pleural effusion; pleurodesis; growth factors; quality of life 387 Introduction A large number of different methods for pleurode­sis used throughout the world tells us that an ideal procedure is still undetermined. Defining the best palliative treatment for malignant pleural effusion has been elucidated many times.1-3 In our clinical practice, we use two distinct pleurodesis proce­dures. The most common choice is chemical pleu­rodesis with talc. We have also established thora­coscopic mechanical pleurodesis as an alternative method to treat malignant pleural effusions.4 By observation, we speculate that chemical pleu­rodesis causes a systemic inflammatory response whereas mechanical pleurodesis only local tissue response with fewer side effects. Considering the known facts of tissue regeneration, scaring, and healing5 we see growth factor release as an impor­tant link in these processes. The primary aim of the study was to analyse growth factors release after performing chemical and mechanical pleurodesis in the first 48 hours. The secondary endpoints were to evaluate the ef­fectiveness of the both pleurodeses, symptoms release and the quality of life of patients after the treatment. Growth factors in pleurodesis Inflammatory cells are the main source of growth factors.6 At the same time, we find them attached to the glycoproteins of the extracellular matrix. Different events in the healing process are trig­gered by their action and interaction.7-9 Their role in the process of pleurodesis was studied many times.10-12 A research for their differential diag­nostic power has exposed the vascular endothelial growth factor (VEGF) being typically higher in ma­lignant pleural effusion.13 We must also take into account and explore the impact of their systemic effects, particularly the role of VEGF in relation to the acute respiratory distress syndrome.14,15 VEGF induces angiogenesis as well as increases vascular permeability and stimulates the tumour growth and metastasis.16 It promotes the formation of pleural effusion and is a crucial factor for the growth of malignant tissue and formation of me­tastases.17-19 A study on an animal model showed the transforming growth factor (TGF) ß1 to be an ideal inductor of pleurodesis with a better perfor­mance than talc but minimal to no side effects.12 Fibroblast growth factor (FGF) ß is clearly linked to the success of pleurodesis.10 After damaging the integrity of extracellular matrix, it is released from the binding sites and serves as the first inductor of fibroblast proliferation and collagen synthesis. Patients and methods The study included 36 female patients with breast carcinoma and cytologically confirmed malig­nant pleural effusion whose lungs re-expanded after thoracic drainage and were eligible for sur­gery. The patients had the Eastern Cooperative Oncology Group (ECOG) performance status 0-2. The study was approved by the National Ethics Committee with number 40/09/09 and conduct­ed at the Department of Thoracic Surgery at the University Medical Centre Maribor, Slovenia, between July 2010 and August 2013. All patients signed a written consent to participate in the study. Laboratory tests and statistical analysis were carried out at the same centre in cooperation with the Biochemistry Division of Medical Faculty, University of Maribor. Overall, 81 female patients with breast carcino­ma and malignant effusion were presented by the oncologists, of whom 36 met the inclusion criteria. We excluded from the study patients who were due to an underlying disease or concomitant diseases not fit to undergo surgery under general anaes­thesia or had a trapped lung. Their demographic data and history as well as prognostic factors that could affect the treatment outcome in different on­cological patients (age20,21, type of tumour16,22, time interval from previous surgical treatment till pleu­rodesis, specific systemic oncological therapy22,23, performance status22,24, maximal volume of previ­ous thoracentesis25) were collected (Table 1). A random numbers were assigned to the pa­tients at admission and they were divided into two groups. Group with chemical pleurodesis with talc (n = 18) and group with thoracoscopic mechanical pleurodesis (n = 18). Patients with chemical preurodesis were treat­ed with a 5 g of talcum (Ph.Eur.7.0, Caesar Loretz GmbH, Austria, EU) and 100 ml of 0.9% NaCl slur­ry over the chest drain. We administered 40 ml of 1% lidocaine intrapleuraly, 20 to 30 minutes prior to talk application. Next, we clamped the drain for 2 hours and then reattached it to the active suction (-15 cm H2O) drainage system for 24 hours. After the first day, the drainage system was switched to underwater seal gravity drainage until the daily amount of drained fluid was less than 200 ml. A fa­vourable chest x-ray report was a requirement for chest tube removal. 388 TABLE 1. Demographical and specific data related to preoperative conditions for chemical pleurodesis with talc (CPT) and thoracoscopic mechanical pleurodesis (TMP) group group Age (years) [±SD] 66 [± 12] 63 [±11] 0.418 Positive hormone receptors 55.6% 50.0% 0.757 (estrogen/progesterone) HER2-positive 31.4% 23.9% 0.741 Time interval from surgery till 63 [± 45] 66 [± 53] 0.884 pleurodesis (months) [±SD] Time interval from chemotherapy 6.5 [± 4.0] 7.0 [± 5.0] 0.837 till pleurodesis (months) [±SD] Received chemotherapy before 94.4% 95.0% 1.000 pleurodesis Received radiotherapy before 33.3% 55.5% 0.210 pleurodesis Median ECOG performance status 1 1.5 0.635 Time from diagnosis of malignant pleural effusion to pleurodesis 3 [± 1] 5 [± 2] 0.112 (months) [±SD] Max. volume of previous 1297 [±354) 1375 [±441] 0.555 thoracocentesis (ml) [±SD] Pleural dissemination (median value) not measurable 3 ­ pH [±SD] 7.27 [±0.09] 7.22 [±0.06] 0.072 ECOG = Eastern Cooperative Oncology Group; HER2 = human epidermal growth factor receptor 2; SD = standard deviation TABLE 2. Median values of growth factors in serum and pleural fluid after chemical pleurodesis with talc (CPT) and thoracoscopic mechanical pleurodesis (TMP) Maximum value of serum 930.68 808.54 0.103 VEGF (pg/ml) [95% CI] [388.22 - 4656.65] [463.20 – 1235.13] Median value of pleural TGFß1 (pg/ml) [95% CI] 1976.50 [1659.82 – 5136.26] 4814.00 [2726.51 – 7292.94] 0.078 Median value of pleural FGF. (pg/ml) [95% CI] 108.35 [73.29 – 162.62] 66.265 [60.71 – 153.12] 0.364 Median value of pleural FGFß (pg/ml) [95% CI] 13.39 [5.04 – 74.60] 30.45 [20.40 – 59.42] 0.076 CI = confidence interval; FGF = fibroblast growth factor; TGF = transforming growth factor; VEGF = vascular endothelial growth Patients with mechanical pleurodesis were treated with a two-port video-assisted thoraco­scopic surgery (VATS)26 during which we per­formed the mechanical abrasion of parietal pleura using A.M.I.-dock reusable applicator for dispos­able (DLU) tips (A.M.I. GmbH, Austria, EU). During VATS, we assessed the coverage of the pleura with malignant tissue: 0 = no obvious lesions; 1 = isolated lesions; 2 = diffuse (covering the majority of the pleura) lesions; 3 = massive (normal pleura could not be seen). The result was calculated by adding the data from visceral, parietal and diaphragmal pleura and ranged from 0 to 9 (Table 1). After 24 hours of active suction (-15 cm H2O) drainage, we disconnected the drains to allow un­derwater seal gravity drainage of effusion and re­moved them after a favourable chest x-ray when the daily amount of drained fluid was less than 200 ml. Post procedural analgesia was kept the same for both study arms. We used a combination of paren­teral metamizole, piritramide and after the first post procedural day, we switched to per orally adminis­tered combination of paracetamol and tramadol. To assess the growth factors release we used a specially designed protocol based on research da­ta of our own unpublished pilot study. The time frame for sampling the pleural fluid and blood samples was: prior to the procedure (time 0) and 3, 12, 24, 36 and 48 h after procedure. For both groups we used only one 20F silicone chest drainage cath­eter (Portex, Smiths Medical, USA) oriented pos­teriorly and caudally, through which we collected pleural fluid samples. Blood samples taken at 24 and at 48 hours were tested for two general indica­tors of inflammation: leukocyte count, C-reactive protein (CRP) level. All pleural samples were also tested for lactate dehydrogenase (LDH) level and pH level of the effusion was determined before the pleurodesis. The supernatant prepared from the pleural fluid samples was centrifuged for 10 min at 3000 revolutions per minute (RPM) at 4°C. Alongside with that of the blood serum it was fro­zen at -70°C and stored for later ELISA testing to determine FGF. and ß, VEGF and TGFß1 levels. To evaluate the effectiveness of both methods of pleurodesis we used chest X-ray and pleural cavity ultrasound examination. The length of tho­racic drainage, hospital stay and pleural effusion re-accumulation were recorded (Table 2). A need for additional procedures due to the recurrence of dispnoic problems and pleural effusion accounted for an unsuccessful pleurodesis. The overall sur­vival was calculated from the day of pleurodesis till the death for any causes. Follow-up lasted up to 12 months. To describe better the immediate effects of pleurodesis, we measured the visual analogue pain score (VAS) at 0, 12, 24, and 48 hours after pleurodesis. Additionally, we used the question­naire of European Organisation for Research and Treatment of Cancer (EORTC) QLQ-C30, version 3.027 to determine the impact of treatment on pa­tients’ quality of life. Global health status, physical functioning, fatigue, pain and dyspnoea measure­ FIGURE 1. Comparison of serum VEGF values during 48 hours after chemical (CPT) and mechanical (TMP) pleurodesis (p = 0.103). ments were selected. Patients completed them with the data for the week before the procedure and for the week and month thereafter. These question­naires served us as a reflection of patient tolerance towards pleurodesis. Statistical analysis Statistical analysis was performed by using IBM SPSS Statistics for Windows, Version 19.0. Armonk, NY: IBM Corp. When comparing groups of patients, .2 test was used to test the frequency counts, t-test was used to test the means of con­tinuous variables and when the value distribution was not normal, Mann-Whitney U test was used to test their median values. Kolmogorov-Smirnov test was used for assessing the normality of data. When dealing with repeated measures, mixed ANOVA was used to compare the mean values between and within groups. Kaplan-Meier survival analysis was used for estimating the survival function for both groups. When performing multiple comparisons Bonferroni correction was used to adjust p-values. Statistical significance was set at p < 0.05. Results Patients The two observed groups did not differ signifi­cantly with respect to demographic data and prog­nostic factors (Table 1). The age, hormonal status of tumours, HER2 status, and time interval from pre­vious surgical treatment, last chemotherapy, time from diagnosis of malignant pleural effusion until pleurodesis, specific systemic oncological therapy, FIGURE 2. Comparison of median values of pleural TGF ß1 during 48 hours after chemical (CPT)-1 and after mechanical (TMP) pleurodesis – 2 (p = 0.078). performance status and maximal volume of previ­ous thoracentesis were well balanced (Table 1). The average coverage of the pleura in the group with mechanical pleurodesis was 3.4 out of 9 points and the median value was 3. The pH values of pleu­ral effusions did not statistically differ (Table 1). Growth factors release after performing chemical and mechanical pleurodesis The focal point of our study was serum VEGF, spe­cifically the maximum value for a given patient. Median serum VEGF level, measured after chemi­cal pleurodesis, was 930.68 pg/ml and after me­chanical pleurodesis, it was 808.54 pg/ml (Table 2, Figure 1). Maximal serum VEGF level after chemi­cal pleurodesis was 11930 pg/ml and after mechan­ical it was 2687 pg/ml (Figure 1). Other key values that showed intergroup differ­ence were that of the pleural TGFß1 and FGF. and ß. Median pleural levels of FGF. and ß and TGFß1 were higher after performing mechanical pleu­rodesis, when compared to those after performing chemical pleurodesis (Table 2, Figure 2). Levels of VEGF in pleural fluid samples and TGFß1 in serum were similar in both groups. We have not recorded any specific impact of any technique of pleurodesis on the serum levels of FGF. and ß, most of the time they were unde­tectable being below the lower limit of sensitivity of ELISA. We found the average largest increase in serum VEGF, TGFß1 pleural value, FGF. and ß pleural values between the 24th and 36th hour post pleu­ 390 TABLE 3. Serum inflammatory parameters and biochemical parameters of pleural effusion before and after chemical pleurodesis with talc (CPT) and thoracoscopic mechanical pleurodesis (TMP) Mean increase in serum leucocyte value 3.8 2.9 0.456 (x109/l) [± SD] (median value of increase) [±3,3] (3.0) [±3.1] (2.3) Mean increase in serum CRP value (mg/l) 54 61 0.977 [± SD] (median value of increase) [±46] (36.5) [±59] (34.5) Mean increase in pleural LDH value (µkat/l) 5.3 4.6 0.826 [± SD] (median value of increase) [±7.6] (2.8) [±18.5] (3.5) CRP = C-reactive protein; LDH = lactate dehydrogenase; SD = standard deviation TABLE 4. Results of post pleurodesis (PD) outcomes for chemical pleurodesis with talc (CPT) and thoracoscopic mechanical pleurodesis (TMP) group Mean thoracic drainage duration (days) 4.5 3.8 0.030 Median hospital stay (days) 5 5 0.126 Pleural effusion re-accumulation 38.9% 20.0% 0.288 Additional thoracentesis required 11.1% 5.0% 0.595 Median survival post PD (months) [95% CI] 6.8 [5-9] 7.0 [4-10] 0.060 CI = confidence interval TABLE 5. Median visual analogue scale (VAS) score for pain before and after chemical pleurodesis with talc (CPT) and thoracoscopic mechanical pleurodesis (TMP) Median VAS before pleurodesis [95% CI] 2 [2-3] 3 [2-3] 0.311 Median VAS 12 hour [95% CI] 5 [3-6] 4 [2-4] 0.039 Median VAS 24 hour [95% CI] 5 [4-6] 4 [3-5] 0.085 Median VAS 48 hour [95% CI] 3 [3-5] 3,5 [3-4] 0.881 CI = confidence interval rodesis. VEGF levels in the serum were still grow­ing up to 48th hour post pleurodesis when we stopped taking samples. There was a significant elevation of blood leuko­cytes in the first 24 hours, it was more pronounced for the group with chemical pleurodesis. The same was established for the CRP levels. The LDH levels in serum and pleura correlated well with the levels of inflammation and they show a tendency to be lower after mechanical pleurodesis compared to those after chemical pleurodesis (Table 3). We made no relevant correlation between pH value of pleural effusion and success of pleurode­ sis or level of growth factors. The effectiveness of pleurodesis Both methods were successful in preventing the re­currence of malignant pleural effusion. One patient in the group with mechanical pleurodesis and two patients in the group with chemical pleurodesis needed additional interventions due to the re-accu­mulation of MPE. The difference in the pleurodesis success between the groups was not significant as in other previous studies on small samples, ex­cept in terms of length of post procedural thoracic drainage (Table 4). Mean thoracic drainage dura­tion after mechanical pleurodesis was significantly shorter than after chemical pleurodesis (p = 0.030). We found no relevant correlation between pleu­rodesis success and the level of growth factors in our study groups. One patient in the group with mechanical pleu­rodesis had a larger drainage of bloody effusion. Haemoglobin in the pleural fluid sample was 4.6 g/dL - 800 ml on the first postoperative day. The volume of drainage has significantly decreased on the post-operative day one. One patient from the group with chemical pleurodesis formed an empy­ema by the end of the first week after pleurodesis. We treated her successfully using negative-pres­sure wound therapy (V.A.C. freedom, K.C.I., USA) over the thoracostomy at the site of the chest drain for 25 days. The median survival of patients was 6.8 months after performing chemical and 7.0 months after performing mechanical pleurodesis (p = 0.060) These patients were evenly distributed between both observation groups and Kaplan-Meier surviv­al analysis showed similar survival curves for both groups. Five patients survived for 1 year or more, which shows more than 10% of one-year survival. There was also no mortality associated with the re­ported procedures (Table 4). Symptoms release and the quality of life The measured score through a pain visual analogue scale (VAS) showed lower pain-load for patients in the TMP group (Table 5). There was significant less pain load after mechanical pleurodesis compared to chemical pleurodesis 12 hours post procedure (p = 0.039). This difference was lost after 48 hours. We got similar results for quality of life (Table 6). The quality of life questioners were filed at the three time points: (1) “pre” – one week before pleuodesis, (2) “post” -1 week after pleurodesis, and (3) “end” – 1 month after pleurodesis. Tables 6 and 7 summarize the important results to express 391 TABLE 6. Comparison of quality of life questionnaire results for global health, physical functioning, fatigue, pain, and dyspnoea between groups at 3 different points in time according to the pleurodesis (PD) - chemical pleurodesis with talc (CPT) and thoracoscopic mechanical pleurodesis (TMP) Global health status (mean ± SD/median) Physical functioning (mean ± SD/median) Fatigue (mean ± SD/median) Pain (mean ± SD/median) Dyspnoea (mean ± SD/median) 15.7 ± 9.0/16.7 38.9 ± 11.5/40.0 77.2 ± 15.9/77.8 67.6 ± 20.2/66.7 68.5 ± 21.3/66.7 20.4 ± 10.0/16.7 47.0 ± 20.6/46.7 68.5 ± 18.4/66.7 63.0 ± 14.6/66.7 61.1 ± 26.2/66.7 28.7± 14.1/25.0 48.9 ± 21.1/53.3 64.2 ± 20.4/66.7 52.8 ± 21.6/58.3 63.0 ± 25.3/66.7 20.0 ± 11.6/16.7 45.0 ± 14.8/40.0 70.6 ± 17.0/77.8 64.2 ± 16.5/66.7 76.7 ± 21.9/66.7 31.3 ± 7.1/33.3 51.3 ± 12.3/53.3 56.7 ± 16.5/55.6 55.8 ± 13.5/50.0 65.0 ± 20.2/66.7 43.8± 12.9/41.7 64.0 ± 13.4/66.7 41.1 ± 19.8/38.9 35.0± 17.0/33.3 40.0 ± 20.5/33.3 pre PD = 1 week prior to pleurodesis; post PD = 1 week after the pleurodesis; end PD = 1 month after pleurodesis TABLE 7. Comparison of the quality of life questionnaire results between the chemical pleurodesis with talc and thoracoscopic mechanical pleurodesis group Global health status* < 0.001 0.001 0.032 0.047 Physical functioning < 0.001 0.057 0.742 0.083 Fatigue < 0.001 0.008 0.271 0.014 Pain < 0.001 0.040 0.477 0.057 Dyspnoea < 0.001 0.536 0.631 < 0.001 * Mauchly’s test indicated that the assumption of sphericity had been violated for global health status; therefore, Greenhouse-Geiser correction was used how the observed interventions effect patients and the impact of the treatment on the quality of life when the effect of pleurodesis is maximal. For specific indicators we took global health, physical functioning, fatigue, pain, and dyspnoea. In both groups, we saw an improvement in qual­ity of life over time. For the majority of targeted questions, the comparison between the two groups showed a statistically significant improvement of quality of life in the group with mechanical pleu­rodesis. A distinctly beneficial effect of mechanical pleurodesis was present as a reduction in the form of dyspnoea (Table 7). Discussion It is our strong belief that studies oriented towards pleural space can help us understand many prob­lems we encounter in our daily work. An ideal agent that would produce effective pleurodesis in the shortest possible time with little side effects and would be affordable remains vague. Light wrote in an article summarizing his research on pleural space that it is the lack of interest of industry that turns researchers away from dedicated research.28 Our wish was to set up new, additional stand­ards on how to determine the best agent for this palliative procedure. We are focused on the cul­prits for small but significant differences between two methods of pleurodesis and try to explain the reasons for rare but serious side effects after chemical pleurodesis with talcum. For patients with malignant pleural effusion, a pleurodesis ef­ficiency of more than 90% achieved by the chemical pleurodesis is satisfactory and does not necessar­ily require further studies. However, the process of healing and scarring is extremely important for understanding the impact of surgical procedure on the body and opens up many opportunities for improvement. Progress in methods of pleurode­sis can be used as an advantage for patients with recurrent primary pneumothorax and other cases where chemical pleurodesis is often used although 392 many thoracic surgeons believe that this is not the best choice of treatment. Besides the usual indicators of inflammation, such as leukocyte number, LDH or CRP14, we are establishing growth factors as additional param­eters to determine the safety and efficiency of dif­ferent methods of pleurodesis. VEGF is the most extensively studied cytokine related to pleural effusion and pleurodesis out­come; it has numerous actions crucial to our un­derstanding of the effects of pleurodesis, central being the fact that it is 50,000 times more potent vasodilator than histamine.29 Previous research da­ta show that an ideal pleurodesis should have lim­ited excretion of VEGF into the pleural space, but should not elevate its serum levels.15,17-19,28,29 VEGF is related to the increased permeability of pleura and promotes growth and metastasis of malignan­cies.15,16,18,19 We believe that accelerated release of VEGF after chemical pleurodesis with talc gives serious concerns for its use.18,29 This is especially important for patients with increased risk of respir­atory problems and maybe generally for patients with malignancies. Extremely high serum levels of VEGF were observed in a small proportion (10%) of patients following chemical pleurodesis. These values of serum VEGF significantly exceed those for which it has been shown that are dangerously high.15 TGFß1 is another multifunctional cytokine strongly connected to success of pleurodesis; in areas of inflammation, it stimulates cell prolifera­tion, increases mesothelial permeability, and the production of fibrin, collagen, and tissue fibrosis.12 Experimental studies have also shown that corti­costeroids and nonsteroidal anti-inflammatory drugs (NSAIDs) reduce the extent of talcum or doxycycline pleurodesis, but, these drugs have no influence on pleurodesis induced with TGF ß1.30 FGFß is another potent inductor of angiogenesis and fibrosis we see relevant. Female patients with breast cancer and associat­ed malignant pleural effusions are the most appro­priate population to answer our research question. Their survival is relatively good and the patients are at the presentation of pleural effusions in a good performance status.31 A large caseload ena­bles us to strictly adhere to the main inclusion cri­teria: good performance status, positive cytology and re-expanded lung after evacuation of pleural effusion. The need for many repeated thoracente­ses, regardless of positive cytology in a patient with breast cancer, is usually an indication for chemical pleurodesis. However, mechanical pleurodesis is another possible method for the prevention of ma­lignant pleural effusion recurrence, but, not widely used. In our study, the two methods of pleurode­sis show distinctly different features. Mechanical pleurodesis has on the contrary to chemical consid­erably less side effects and is proven more effective in advanced malignancy.4 Our data was collected on a small sample, but, we can plan for broader studies with targeted sample collection between 24 and 48, or up to 72 hours after the procedure. Monitoring and com­parison of specific biochemical parameters is rea­sonable on a small number of well-comparable patients as well29, which we guaranteed with the specified inclusion criteria. The timelines were set based on previous studies4 and a pilot study that we conducted at our department prior to the study that is presented. A measurable increase of growth factors levels in blood serum and pleural fluid is unlike in animal studies14 observed a lit­tle later. We recorded the largest increase of serum VEGF and pleural TGFß1 and FGF. and ß values between the 24th and 36th hour. After this period, the values were still gradually increasing up to 48 hours post pleurodesis when we stopped record­ing them. These processes might be lengthier in humans compared to small animals, which should be considered for future research. The collection of samples after 48 hours is difficult as pleurodesis is usually already quite strong and the amount of fluid drained through catheters, that may already be clogged, is minimal. There was also no relevant correlation between success of pleurodesis and the level of growth fac­tors in our study groups. We believe this is due to the high efficiency of both methods. Therefore, it is difficult to estimate the impact growth factors have in these few unsuccessful cases. We made our conclusions based on our study in the light of several other reports that describe the role of grow factors in the setting of pleural effusion and pleu­rodesis. The two observed methods, chemical and me­chanical pleurodesis, are the most commonly used for treatment of patients with primary spontaneous pneumothorax. Small amounts of drained pleural effusion after pleurodesis in this group prevent an insight into the release of cytokines and other com­ponents that contribute to fibrosis. In addition, we chose patients with malignant pleural effusion as a more appropriate observation group as we do not perform chemical pleurodesis on young patients or for benign diseases with long-term survival, such being the primary spontaneous pneumothorax. 393 The coverage of the pleura with malignant tis­sue is another factor that affects the success of pleu­rodesis, which we could verify only in the group with mechanical pleurodesis. The fact that the lungs re-expanded, normal width of the mediasti­num on the chest x-ray and positive cytology al­lows us to conclude that in the patients group with chemical pleurodesis the pleura was also covered with a comparable proportion of malignant tissue. A small number of unsuccessful pleurodesis in our study is attributed to the selection criteria, since the fibrosis process is more pronounced in healthier pleura. Judging by the data collected in the group with mechanical pleurodesis, an average coverage is only 3.4 from a total of 9 points. Our protocol included questionnaires on pain load and quality of life. The data are relevant, so we included them in this paper. Patients reported greater sensation of pain and dyspnoea after per­forming chemical compared to the mechanical pleurodesis. We see pleural inflammation as the main source of pain after pleurodesis. Additionally, we are connecting the effect, which talcum has on the lung parenchyma, with the sensation of dysp­noea. After being absorbed to some extent into tis­sue, talc causes pneumonitis, pulmonary oedema and signs of acute respiratory distress syndrome. This effect of pleurodesis is less pronounced after mechanical pleurodesis. There was a significant main effect of time on all assessed quality of life scales (global health status, physical functioning, fatigue, pain and dyspnoea) with “post” scores being significantly better than “pre” scores and “end” scores being better than “post” scores with only one exception, there was no significant difference between “pre” and “post” dyspnoea score. There was also a significant main effect of treatment group on the global health status, fatigue and pain score with better average scores across all points in time in the TMP group. We were especially interested in the interaction effect be­tween time and treatment group, broken down to comparing “post” and “end” scores to “pre” scores across both groups. These comparisons revealed significant interactions when comparing chemical and mechanical pleurodesis global health status scores (both “post” vs. “pre” and “end” vs. “pre”), “end” vs. “pre” fatigue scores and “end” vs. “pre” dyspnoea scores. Looking at the interaction graphs, this suggests that those scores improved more sub­stantially from “pre” to “end” point in the group with mechanical pleurodesis than in the group with chemical pleurodesis as well as from “pre” to “post” point in the case of global health status (Tables 6, 7). By testing our ideas and views of treatment of malignant pleural effusion, we tried to look into the effect our treatment has on tissue or on the whole body. By giving some answers as to what is important in view of better and faster healing and what should be omitted when testing new methods, we try to help the dedicated laborato­ries. These parameters can be easily recorded in laboratory animals and can help in minimising the needed numbers. We are also directing the interest of professional public towards the growth factors when trying to explain other differences between surgical treatments.31 New solutions for many clinical issues can be developed by examining the process of healing and fibrosis. We believe that the main limiting factor for the clinical use of growth factors is the cost. It is espe­cially limited for terminal patients. Nevertheless, for further experimental studies on pleurodesis and other procedures that boost or stimulate fibro­sis growth factors represent an important indicator of efficiency and safety. It is also interesting that TGF ß1 works better in an environment with a low­er pH.32 Again this experimental data could not be clarified in our study groups, mainly because the success rate was very high. It would probably be necessary to perform autopsies to reveal the extent of fibrosis and connect this data to the level of pH and the TGF ß1 level. In cases of malignant pleural effusion, a low pH value is an indicator of an ad­vanced disease, such patients accumulate effusion faster. In this case, an agent that triggers abundant secretion of TGF ß1 should enable faster pleurode­sis. Surgeons use extremely expensive devices and procedures to promote healing and fibrosis. We believe that further exploration of clinical applica­tion or promotion of topical secretion of growth factors might be useful in many cases. One option is by supplying the growth factors to the place where we want their activities. Another would be the promotion of targeted growth factors secretion that could accelerate healing. Both should be use­ful on the resection surfaces and reduce the time of tissue leakage and bleeding. Achieving this might have a significant impact on the main factors for the postoperative complications and duration of hospitalization. Conclusions We recorded an increase in serum VEGF levels after chemical pleurodesis with talcum and an increase 394 in the pleural fluid level of FGFß and TGFß1 after thoracoscopic mechanical pleurodesis with respect to compared group. The differences did not reach statistical significance; however, TGFß1, FGFß and VEGF remain the most interesting parameters for future research. Considering the mechanisms of growth factors action, we conclude that in our study group mechanical pleurodesis might be more efficient in terms of growth factors release, better-tolerated and safer method than chemical pleurodesis. Acknowledgements Sources of funding for the work contributed by University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia in full amount. We would like to thank the laboratory techni­cian, Marjetka Dugonik, statistician Miroslav Palfy and the staff from Department for thoracic surgery, UMC Maribor, for the help during our research. References 1. Astoul P. Pleurodesis for recurrent malignant pleural effusions: the quest for the Holy Grail. [Editorial]. Eur J Cardiothorac Surg 2011; 40: 277-9. 2. Tan C, Sedrakyan A, Browne J, Swift S, Treasure T. The evidence on the effec­tiveness of management for malignant pleural effusion: a systematic review. Eur J Cardiothorac Surg 2006; 29: 829-38. 3. Debeljak A, Kecelj P, Triller N, Letonja S, Kern I, Debevec L, et al. Talc pleu­rodesis: comparison of talc slurry instillation with thoracoscopic talc insuffla­tion for malignant pleural effusions. J BUON 2006; 11: 463-7. 4. Crnjac A, Sok M, Kamenik M. Impact of pleural effusion pH on the efficacy of thoracoscopic machanical pleurodesis in patients with brest carcinoma. Eur J Cardiothorac Surg 2004; 26: 432-6. 5. Mitchell RN, Cotran RS. Tissue repair: cell regeneration and fibrosis. In: Kumar V, Cotran RS, Robbins SL, editors. Robins Basic Pathology. 7th edition. Philadelphia: Saunders; 2003. p. 61-78. 6. Lorenz PH, Longaker MT. Wounds: biology, pathology, and management. In: Norton JA, Barie PS, Bollinger RR, Chang AE, Lowry SF, Mulvihill SJ, et al, editors. Surgery: basic science and clinical evidence. 2nd edition. New York: Springer; 2008. p. 191-208. 7. Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing. Wound Repair Regen 2009; 17: 153-62. 8. Midwood KS, Valenick Williams L, Schwarzbauer JE. Tissue repair and the dynamics of the extracellular matrix. Int J Biochem Cell Biol 2004; 36: 1031-37. 9. Noncollagen components of the extracellular matrix. In: Lodish H, Berk A, editors. Molecular cell biology. 4th edition. New York: W. H. Freeman; 2000. p. 985-93. 10. Antony VB, Nasreen N, Mohammed KA, Sriram PS, Frank W, Schoenfeld N, et al. Talc pleurodesis: basic fibroblast growth factor mediates pleural fibrosis. Chest 2004; 126: 1522-8. 11. Teixeira LR, Vargas FS, Acencio MMP, Sabrina C. Ribeiro SC, Sales RKB, et al. Blockage of vascular endothelial growth factor (VEGF) reduces experimental Pleurodesis. Lung Cancer 2011; 74: 392-5. 12. Gary Lee YC, Teixeira LR, Devin CJ, Vaz MAC, Vargas FS, Thompson PJ, et al. Transforming growth factor-ß2 induces pleurodesis significantly faster than talc. Am J Respir Crit Care Med 2001; 163: 640-4. 13. Duysinx BC, Corhay JL, Hubin L, Nguyen D, Henket M, Louis R. Diagnostic value of interleukine-6, transforming growth factor-beta 1 and vascular endothelial growth factor in malignant pleural effusions. Respir Med 2008; 102: 1708-14. 14. Genofre EH, Vargas FS, Acencio MMP, Antonangelo L, Teixeira LR, Marchi E. Talc pleurodesis: evidence of systemic inflammatory response to small size talc particles. Respir Med 2009; 103: 91-7. 15. Thickett DR, Armstrong L, Christie SJ, Millar AB. Vascular endothelial growth factor may contribute to increased vascular permeability in acute respira­tory distress syndrome. Am J Respir Crit Care Med 2001; 164: 1601-5. 16. Koenigsberg R, Maierhofer J, Steininger Tanja, Kienzer, G, Dittrich C. Long-term remission of a Her2/neu positive primary breast cancer under double monoclonal antibody therapy with trastuzumab and bevacizumab. Radiol Oncol 2014; 48: 184-8. 17. Hooper CE, Elversb KT, Welshc GI, Millard AB, Maskell NA. VEGF and sVEGFR-1 in malignant pleural effusions: Association with survival and pleurodesis outcomes. Lung Cancer 2012; 77: 443-9. 18. Ishii H, Yazawab T, Satob H, Suzukib T, Ikedab M, Hayashia Y, et al. Enhancement of pleural dissemination and lymph node metastasis of in­trathoracic lung cancer cells by vascular endothelial growth factors (VEGFs). Lung Cancer 2004; 45: 325-37. 19. Mercurio AM, Lipscomb EA, Bachelder RE. Non-angiogenic functions of VEGF in breast cancer. J Mammary Gland Biol Neoplasia 2005; 10: 283-90. 20. Auer J, Keller U,Schmidt M, Ott O, Fietkau R, Distel LV. Individual radiosensi­tivity in a breast cancer collective is changed with the patients’ age. Radiol Oncol 2014; 48: 80-6. 21. Kovač V, Smrdel U. Meta-analyses of clinical trials in patients with non-small cell lung cancer. Neoplasma 2004; 51: 334-40. 22. Tuzi A, Lombardi D, Crivellari D, Militello L, Perin T, La Grassa M. Epirubicin and docetaxel as neoadjuvant treatment of hormone receptor positive, HER-2 negative breast cancer: findings from two successive phase II studies. Radiol Oncol 2013; 47: 57-62. 23. Debevec L, Erzen J, Debeljak A, Crnjac A, Kovac V. Exploratory thoracotomy and its influence on the survival of patients with lung cancer. Wien Klin Wochenschr 2006; 118: 479-84. 24. Kovac V, Zwitter M, Rajer M, Marin A, Debeljak A, Smrdel U, Vrankar M. A phase II trial of low-dose gemcitabine in prolonged infusion and cisplatin for malignant pleural mesothelioma. Anticancer Drugs 2012; 23: 2308-8. 25. Klecka ME, Maldonado F. Symptom relief after large-volume thoracentesis in the absence of lung perfusion. Chest 2014; 145: 1141-3. 26. Crnjac A. The significance of thoracoscopic mechanical pleurodesis for the treatment of malignant pleural effusions. Wien Klin Wochenschr 2004; 116(Suppl 2): 28-32. 27. Aaronson NK, Ahmedzai S, Bergman B, Bullinger M, Cull A, Duez NJ, et al. The European Organisation for Research and Treatment of Cancer QLQ-C30: A quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst 1993; 85: 365-76. 28. Light RW. What I have learned in the past 40 Years. Clin Chest Med 2013; 34: xi. 29. Zebrowski BK, Yano S, Liu W, Shaheen RM, Hicklin DJ, Putnam JB, et al. Vascular endothelial growth factor levels and induction of permeability in malignant pleural effusions. Clin Cancer Res 1999; 5: 3364-8. 30. Teixeira LR, Vargas FS, Acencio MMP, Antonangelo L, Novaes NP, Costa JD, et al. Influence of parecoxib (cox-2 inhibitor) in experimental pleurodesis. Respir Med 2009; 103: 595-600. 31. Zahid I, Routledge T, Bille A, Scarci M. What is the best treatment for malig­nant pleural effusions? Interact CardioVasc Thorac Surg 2011; 12: 818-23. 32. Kubiczkova L, Sedlarikova L, Hajek R, Sevcikova S. TGF-ß – an excellent serv­ant but a bad master. J Transl Med 2012; 10: 183. 395 case report Giant solitary fibrous tumour of the pleura. Case report and review of the literature Anton Crnjac1, Bojan Veingerl1, Damjan Vidovic1, Rajko Kavalar2 Aljaz Hojski1 1 Department of Thoracic Surgery, Division of Surgery, University Medical Centre Maribor, Slovenia 2 Department of Pathology, University Medical Centre Maribor, Slovenia Radiol Oncol 2015; 49(4): 395-401. Received 16 July 2014 Accepted 4 August 2014 Correspondence to: Prof. Anton Crnjac, M.D., Ph.D., Department of Thoracic Surgery, Division of Surgery, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia. E mail: anton.crnjac@ukc-mb.si Disclosure: No potential conflicts of interest were disclosed. Background. Solitary fibrous tumours of the pleura (SFTP) are rare tumours. They are mostly benign. Only around 12% of them are malignant. In the initial stage they are mostly asymptomatic and by growing they cause chest pain, irritat­ing cough and dyspnoea on account of the pressure created on the surrounding structures. Rare giant tumours have compression symptoms on the mediastinal structures. The condition requires tiered diagnostic radiology. Preoperative biopsy is not successful in most cases. The therapy of choice is radical surgical tumour removal. Malignant or non-radically removed benign solitary fibrous tumours of the pleura additionally require neoadjuvant therapy. Case report. A 68-year old patient was hospitalized for giant solitary fibrous tumour of the pleura in the right pleural cavity. With its expansive growth the tumour caused the shift of the mediastinum by compressing the lower vena cava, right cardiac auricle as well as the intermediate and lower lobe bronchus. Due to cardiac inflow obstruction and right lung collapse, the patient’s life was endangered with signs of cardio-respiratory failure. After preoperative diagnostic radiology, the tumour was surgically removed. Postoperatively, the patient’s condition improved. No dis­ease recurrence was diagnosed after a year. Conclusions. Giant solitary fibrous tumour of the pleura may cause serious and life-threatening conditions by causing compression of the pleural cavity with its expansive growth. Early diagnosis of the condition enables less aggressive as well as video-assisted thoracic surgery in patients with significantly better state of health. Large tumour surgeries in cardio-respiratory affected patients are highly risk-associated procedures. Key words: solitary fibrous tumour of the pleura; expansive growth; mediastinum shift; surgical treatment Introduction Solitary fibrous tumours of the pleura (SFTP) are rare mesenchymal tumours representing less than 5% of all tumours of the pleura.1 Around 800 cas­es of such tumour types have been mentioned in global literature up to date.2,3 In 1870 Wagner was the first to describe a localized primary tumour of the pleura4, Klemperer and Rabin first classified them in 1931 into a diffuse and localized form and set a hypothesis that localized mesothelium, cover­ing the intact layer of mesothelial cells, stems from structures under the mesothelial layer.5 With the introduction of electronic microscopic and immu­nohistochemical examinations it was finally con­firmed that SFTP grow from deeper-lying mesen­chymal structures of the thoracic wall. Historically, SFTP are thought to derive from subpleural mesenchymal cells with fibroblasts or myofibroblast differentation.6 However, negative staining for smooth muscle markers and diffuse positivity for CD34 led van de Rijn et al. to propose an origin of this tumour from ubiquitous dispersed dendritic interstitial cels.7 Recent ultrastructural observations have highlighted that SFTP may orig­inate from peculiar perivascular multipotent mes­enchymal elements displaying features akin to per­icytes and submesothelial fibroblasts.8 To differen­tiate these tumours from other soft tissue tumours, immunohistochemical examinations are required. 396 TABLE 1. Malignancy criteria for SFTP A. Abundant cellularity with crowding and overlapping of nuclei B. High mitotic activity of more than four mitotic figures per 10 high power fields C. Pleomorphism with cytonuclear atypia d. Large necrotic or haemorrhagic areas E. Associated pleural effusion f. Atypical location and inversion of adjacent structures TABLE 2. Classification of SFTP into stages Stage 0 –peduculated SFTP without signs of malignancy Stage 1- sessile or inverted SFTP without signs of malignancy Stage 2- pedunculated SFTP with histological signs of malignancy Stage 3- sessile or inverted SFTP with histological signs of malignancy Stage 4- multiple synchronous metastatic tumours Tumours with similar histological characteristics are described by certain authors also in extratho­racic organs, such as liver, peritoneum, meninges, orbits, thyroid gland, salivary gland, breast.9-12 SFTP affect male and female patients equally; however, they may develop in all age periods (5 to 87 years) with the highest incidence between 60 and 70 years of age.13,14 Generally, there is no evidence of correlation with the genetic predisposition for the tumour, and in contrast with mesothelioma15, there is no relationship to the exposure to asbestos, tobacco or any other environmental agents.16 Most SFTP are benign, but may alter in malig­nancy with age. They usually develop in lower part of pleural cavity; from the visceral pleura in around 80%.16 Individual tumours grow over 10 cm; however, only individual cases of giant tumours measuring over 20 cm are described.1 Cytological and histological diagnosis as well as differential diagnosis by defining the benign or malignant biological potential is difficult to per­form with the bioptic material acquired by means of fine-needle aspiration biopsy or with large-core needle biopsy. The final diagnosis is usually made after the tumour has been removed. England et al. set the basis for differentiating benign from malig­nant SFTP (Table 1).13 Based on the histological and morphological characteristics of SFTP, De Perrot et al. classified tumours into five stages and thus enabled easier planning of therapy and expected outcome of treatment (Table 2).2 Most minor SFTP are asymptomatic. They are usually discovered incidentally during chest X-ray examination.17 By growing and pressuring the surrounding struc­tures they become symptomatic. The most com­mon clinical signs are coughing, dyspnoea, and chest pain, especially in tumours growing from parietal pleura. Rarely are they manifested with the signs of haemoptysis, obstructive pneumonitis or atelectasis.18 In larger tumours, digital clubbing and hypertrophic pulmonary osteoarthropathy (Pierre Marie Bamberg syndrome)19 may be present or signs of refractory hypoglycaemia on account of insulin-like growth hormone release (Doege Potter syndrome).20 Extremely large tumours cause a va­riety of clinical signs of pressure on the mediasti­num or mediastinal shift. Diagnostic radiology plays a very important role in discovering SFTP. Chest radiographs of pa­tients demonstrate a well-defined, solitary nodule or mass, which may appear at the lung periph­ery and typically abuts the pleural surface or is located within a fissure.21 Pedunculated tumours may show mobility within the pleural space.14 Computer tomography (CT) of the chest shows a homogenous, well-defined and lobulated soft tis­sue mass.20 In cases with suspected infiltrative tu­mour growth into the mediastinal structures, like in others pleural tumours, magnetic resonance im­aging (MRI) is required.22,23 Because such tumours are well-circulated, it seem sensible to perform an­giography.24 Lately, it has been recommended to perform a PET-CT scan, especially when suspect­ing malignant SFTP or to confirm the presence of potential metastases.25 Radical surgical resection is the optimal way of treating patients with SFTP. Aggressive surgery is recommended due to the high probability of their recurrence.26 The safety margin of healthy tissue after resection should be 1-2 cm wide. Wedge re­section of the lung and limited pleurectomy may suffice in peripheral tumours. For sessile tumours it is necessary to perform a lobectomy or pneumo­nectomy as well as extensive pleurectomy, some­times even partial resection of the chest wall.27 Smaller, especially pedunculated tumours, can also be radically removed with minimally invasive thoracoscopic surgical procedure (VATS), which is routinely use in different thoracic pathologies.28 In cases of larger SFTP, the continuation of surgical treatment with adjuvant chemotherapy is indicat­ed.27,29 Park et al. have found that the combination of temozolomide and bevacizumab had high rates of overall response and long term disease control.30 In their study, patients received temozolomide 150 mg/m2 orally on days 1-7 and days 15-21 and beva­cizumab 5 mg/kg intravenously on day 8 and day 22 on a 28-day cycle.30 The role of brachytherapy 397 and photodynamic therapy, a method in treating diffuse mesotheliomas, has not been sufficiently studied.31 Case report 68-year old female patient was admitted to the Department of Lung Disease with signs of severe cardio-respiratory failure. One month prior to be­ing admitted, the patient’s breathing was getting heavier, she was tired, weak, had poor appetite and increasing pain in the right hemithorax. She also had arterial hypertension and atrial fibrilla­tion. She has never smoked. Written informed consent of patient was ob­tained for the treatments and for the scientific use of clinical data, according to Declaration of Helsinki and Slovenian law requirements. At examination the patient was cyanotic, tachyp­noic with the breathing frequency 22/min. With the administration of oxygen by a nasal catheter a peripheral capillary oxygen saturation (SpO2) was 91%, the patient had signs of heart failure with atrial fibrillation. Breathing was weakened and au­dible only apically on the right side. Pulmonary function test showed a significant decrease in val­ues of the forced vital capacity (FVC, 42% of the norm), the forced expiratory volume in 1 second (FEV1, 35% of the norm), and the FEV1/FVC ra­tio or Tiffeneau index (TI, 68% of the norm). Gas analysis of arterial blood showed signs of chronic hypercapnic respiratory failure. Chest X-ray showed a large tumour mass in the right part of the thorax with mediastinal shift to the left (Figure 1). CT scan of the chest showed an extensive expansive process, larger than 20 cm. The tumour was heterogeneous, lobulated and practically extended over the entire right pleural cavity and shifted mediastinal structures to the left (Figure 2). An MRI examination did not confirm tumour infiltration of the surrounding mediastinal structures (Figure 3). Bronchoscopy showed a visibly compressed tra­chea from the right side, shift of the carina and the main right bronchus into the left and closed bron­chus for the right lower lung lobe. Transbronchial tumour biopsy was negative; the material collected by means of transthoracic needle biopsy did not suffice for histological tumour confirmation. For purposes of further diagnosis and surgi­cal treatment the patient was transferred to the Department of Thoracic Surgery. In the preopera­tive phase we performed an ultrasound (US) of the 398 heart and angiography of the tumour. Heart US showed a compressed right atrium and lower ve­na cava as well as increased pressure in the right atrium and ventricle. Angiography displayed an extensive blood circulation of the tumour from the intercostal arteries 8, 9, 11, and 12 and inferior phrenic arteries from the right side (Figure 4). Because of the clinical status of the patient with the signs of respiratory failure, cardiac inflow obstruction and the possibility of massive bleed­ing, all together representing a high-risk surgery, a detailed plan was designed. A wide approach for safe and radical tumour removal was enabled with the right thoracosternotomy (hemiclamshell). By continuous ligation of blood vessels nourish­ing the tumour, the blood loss during surgery was only 1.5 l of blood, which was recycled by a cell-saver. Surgical preparation in the mediastinal area was difficult because of compressed structures and numerous postinflammatory adhesions. A fully re­moved tumour was sent for pathohistological ex­amination (Figure 5). Macroscopic examination of the resected speci­men showed firm lobulated and bosselated white-grey tumour measuring 25 cm x 16 cm x 13 cm. The surface of the tumour was covered with thin, shiny, smooth capsule. The cut surface was rub­bery, vaguely nodular, grey-white, focally glassy and haemorrhagic. On the edges tiny calcifications were present. Macroscopically no necrotic areas were identified. Microscopically the bland appearing tumour cells were arranged in a “patternless pattern” (storiform and fascicular pattern), the hypercellular regions were mixed with hypocellular areas with hyalinised stroma. In some areas stromal myxoid change and degeneration of collagen was present, too. Stroma was highly vascularized with angiofi­bromatous and haemangiopericytic vascular pat­tern. The tumour cells were spindle and oval with scant cytoplasm and nuclei with dense chromatin. Focally spindle cells showed wavy nuclei, resem­bling schwanian cells and also there were some ar­eas with pleomorphic and giant cells population. The nuclei of the pleomorphic cells were larger, hy­perchromatic, and different in shape. Very rare mi­totic figures (<2/10 high-power fields [HPF]) were present and there was no necrosis (Figure 6). Immunochistochemically tumour cells were re­active for CD34, CD99 and bcl2 and typically no immunoreactivity was observed with S-100, WT-1, Desmin, CEA, CK AE1/AE3, CK5/6 and calretinin (Figure 7). According to the morphology and cellular im­munophenotype the diagnosis of benign giant pleural SFT was signed out. The patient was placed for seven days into a room with perioperative intensive care and ex­tubated after two days. Longer intubation was required to ventilate a long time collapsed right intermediate and lower lung lobe. She left the hos­pital on day 22 after surgery. At the follow-up after a year, no recurrence of the disease was present. discussion SFTP are a rare pathology of the pleural cavity, which most of the time develop from submesothe­ 400 Complete surgical excision Partial surgical excision Neoadjuvant treatment Pathological examination of the mass ¦ Rare ly used due to the difficulty to diagnose SFTP before surgical intervention. In addit ion1 needle biopsy is usually inconclusive. ¦ Could be used a lone or followed by surgery . Completely excised benign SFTP Completely excised malignant SFTP Part ia lly excised benign SFTP Part ia lly excised malignant SFTP No adjuvant therapy Adjuvant t herapy 6-months radiology (CT) follow-up for 2 years then yearly TABLE 3. The guidelines provided by De Perrot et al. SFTP = solitary fibrous tumours of the pleura agnosis in most studies.19 Cutting needle biopsy is probably preferable because of wider tissue sam­pling.35 Thoracoscopic procedure, an effevtive di­agnostic and therapeutic method, have also be to considered.36 In our case it was not possible to col­lect suitable material for histological analysis with transthoracic and transbronchial biopsy. Because in most cases the biological potential of SFTp is not preoperatively histologically con­firmed, neoadjuvant therapy is not appropriate. After removing the tumour surgically we opt for adjuvant therapy in malignant or non-radically removed benign SFTP in accordance with the guidelines provided by De Perrot et al. (Table 3).16 Because the benign SFTP was removed radically, our patient did not receive adjuvant therapy. It is possible that the tumour will recur, which mainly depends on the histological characteristics of SFTP and radical nature of the surgical proce­dure. The possibility of benign pedunculated tu­mours recurring is around 2%, for benign sessile tumours around 8%, malignant pedunculated tu­mour around 14%, and for malignant sessile tu­mours around 63%.31,37 It is necessary to follow the patients, usually every 6 months when control CT scan of the chest is performed. No recurrence was established in our patient after a year. Conclusions SFTP are rare pleural neoplasms, stemming from submesothelial fibroblast cells and are in more than 80% benign. Initially, they are asymptomatic and by growing they create pressure on the sur­rounding structures of the chest and cause chest pain, coughing, dyspnoea, and by pressuring on the mediastinum they can cause life-threatening signs of mediastinal shift. Tiered diagnostic ra­diology is very important and provides valuable data in the most appropriate manner of treatment. Preoperative biopsy is usually not successful and the final diagnosis is obtained in most cases only after the surgical removal of the tumour. Radical surgical resection is the method of choice when treating benign and operable SFTP and need to be upgraded in malignant or non-radically removed benign tumours with adjuvant therapy. Extensive surgical resections can be avoided with timely diagnosis of smaller tumours, which can be radi­cally removed with VATS. Regular check-up are required due to possible disease recurrence. References 1. Furukawa N, Hansky B, Niedermayer J, Gummert J, Renner A. A silent gigantic fibrous tumor of the pleura: case report. J Cardiothorac Surg 2011; 6: 122-6. 2. De Perott M, Fischer S, Brundler MA, Sekine Y, Keshavjee S. Solitary fibrous tumors of the pleura. Ann Thorac Surg 2002; 74: 285-93. 3. Sung SH, Chang JW, Kim J, Lee KS, Han J, Park S. Solitary fibrous tumors of the plevra. Surgical and cinical course. Ann Thorac Surg 2005; 79: 303-7. 4. Wagner E. Das tuberkelähnliche lymphadenom. (Der cytogene oder reticu­lirte tuberkel). Arch Heilk (Leipzig) 1870; 11: 495-525. 5. Klemperer P, Rabin CB. Primary neoplasm of the pleura: a report of five cases. Arch Pathol 1931; 11: 385-412. 6. Robinson LA. Solitary fibrous tumor of the pleura. Cancer Control 2006; 13: 264-9. 7. Van de Rijn M, Lombard CM, Rouse RV. Expression of CD34 by solitary fibrous tumors of the pleura, mediastinum and lung. Am J Surg Pathol 1994; 18: 814-20. 8. Ide F, Obara K, Mishima K. Ultrasructural spectrum of solitary fibrous to­mor: a unique perivascular, tumor with alternative lines of differentation. Virchows Arch 2005; 446: 646-52. 9. Ibrachim NB, Briggs J, Corrin B. Double primary localized fibrous tumor of the pleura and retroperitoneum. Histopathology 1993; 22: 282-4. 10. Vaswani K, Guttikonda S, Vitellas KM, Case 1. Localized fibrous tumor of the liver. AJR Am J Roentgenol 2000; 175: 875-6. 11. Sandvliet RH, Heysteeg M, Paul MA. A large thoracic mass in a 57-year-old patient: solitary fibrous tumor of the pleura. Chest 2000; 117: 897-900. 12. Safneck JR, Alguacil-Garcia A, Dort JC, Philips SM. Solitary fibrous tumor: report of two new locations in the upper respiratory tract. J Laryngol Otol 1993; 107: 252-6. 13. England DM, Hochholzer L, McCarthy MJ. Localized benign ena malignant fibrous tumor of pleura: a clinicopathologic rewiev of 223 cases. Am J Pathol 1989; 13: 640-58. 401 14. Dynes MC, White EM, Fry WA, Ghahremany GG. Imaging manifestations of pleural tumors. Radiographics 1992; 12: 1191-201. 15. Kovac V, Zwitter M, Zagar T. Improved survival after introduction of chemo­therapy for malignant pleural mesothelioma in Slovenia: Population-based survey of 444 patients. Radiol Oncol 2012; 46: 136-44. 16. Walid AA. Solitary fibrous tumors of the pleura. Eur J Cardiothorac Surg 2012; 41: 587-97. 17. Magdeleinat P, Alifano M, Petino A, Le Rochais JP, Dulmet E, Gaalateau F. Solitary fibrous tumor of pleura: clinical charateristic, surgical treatment and outcome. Eur J Cardiothorac Surg 2002; 21: 1087-93. 18. Robinson LA. Solitary fibrous tumor of the pleura. Cancer Control 2006; 13: 264-9. 19. Rena O, Filosso PL, Papalia E, Molinatti M, Di Marzio P, Maggi G. Solitary fibrous tumor of the pleura: surgical treatment. Eur J Cardiothorac Surg 2001; 191: 185-9. 20. Chamberlain MH, Taggart DP. Solitary fibrous tumor associated with hy­poglicemia: an example of the Doege-Potter syndrome. J Thorac Cardiovasc Surg 2000; 119: 185-7. 21. Dedrick CG, McLoud TC, Shepard JA, Shipley RT. Computed tomography of localized pleural mesothelioma. AJR Am J Roentgenol 1895; 144: 275-80. 22. Ferretti GR, Chiles C, Cox JE, Choplin RH, Coulomb M. Localized benign fibrous tumor of the pleura: MR appearance. J Comput Assist Tomogr 1997; 21: 115-20. 23. Podobnik J, Kocijancic I, Kovac V, Sersa I. 3T MRI in evaluation of asbestos-related thoracic diseases - preliminary results. Radiol Oncol 2010; 44: 92-6. 24. Orki A, Kosar A, Akin A, Haclibrahimoglu G, Arman SB. Solitary fibrous tumor of the pleura. Thorac Cardiovasc Surg 2008; 56: 287-90. 25. Gaint DT, Bokharl A, Shatt S, Dogra V. Imaging features of solitary fibrous tumors. AJR Am J Roentgenol 2011; 196: 487-95. 26. Sung HS, ChangJ-W, Kim J, Lee KS, Han J, Park S. Solitary fibrous tumors of the pleura: surgical outcome and clinical cozrse. Ann Thorac Surg 2005; 79: 303-7. 27. Suter M, Gebhard S, Boumghard M, Peloponisions N, Genton CY. Localized fibrous tumor of the pleura: 15 new cases and rewiev of the literature. Eur J Cardiothorac Surg 1998; 14: 453-9. 28. Crnjac A, Antonic J, Zorko A, Veingerl B. Video-assisted thoracic surgery - a new possibility for the management of traumatic hemothorax. Wien Klin Wochenschr 2001; 113(Suppl 3): 18-20. 29. Veronesi G, Spaggiari L, Mazzarol G, De Pas M, Leo F, Solli P. Huge malignant localized fibrous tumor of the pleura. J Cardiovasc Surg 2000; 41: 781-4. 30. Park MS, Patel SR, Ludwig JA, Trent JC, Conrad CA, Lazar AJ. Activity of temo­zolomide and bevacizumab in the treatment of locally advancet, recurrent and metastatic hemangiopericytoma and malignant solitary fibrous tumor. Cancer 2011; 117: 4939-47. 31. De Perrot M. Fibrous tumors of the pleura. Curr Treat Opt Oncol 2000; 1: 293-8. 32. Bicer M, Yaldiz S, Gursoy S, Ulgan M. A case of giant benign localized fibrous tumor of the pleura. Eur J Cardiothorac Surg 1998; 14: 211-3. 33. Tublin ME, Tessler FN, Rifkin MD. US case of the cay. Solitary fibrous tumor of the pleura (SFTP). Radiographic 1998; 18: 523-5. 34. Fan F, Hiu Z, Qinghai Z, Yupeng L. Computed tomography manifestations of malignant solitary fibrous tumor of the pleura with distinct blood supply from celiac trunk. Eur J Cardiothorac Surg 2014; 45: 1108-10. 35. Weynand B, Noel H, Goncette L, Noirhomme C, Colard P. Solitary fibrous tumor of the pleura: a report of five cases diagnosed by transthoracic cuttig needle biopsy. Chest 1997; 112: 1424-8. 36. Crnjac A. The significance of thoracoscopic mechanical pleurodesis for the treatment of malignant pleural effusions. Wien Klin Wochenschr 2004; 116(Suppl 2): 28-32. 37. Santos RS, Haddad R, Lima CE, Liu YL, Misztal M, Ferreira T. Patterns of rec­curence and long-term survival after curative resection of localized fibrous tunors of the pleura. Clin Lung Cancer 2005; 7: 197-201. 402 research article Impact of comorbidity on the outcome in men with advanced prostate cancer treated with docetaxel Andrej Zist1, Eitan Amir2, Alberto F. Ocana3, Bostjan Seruga1 1 Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia 2 Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Canada 3 Medical Oncology Department and Translational Research Unit, Albacete University Hospital, Spain Radiol Oncol 2015; 49(4): 402-408. Received 05 May 2015 Accepted 19 July 2015 Correspondence to: Assist. Prof. Boštjan Šeruga, M.D., Ph.D., Department of Medical Oncology, Institute of Oncology Ljubljana, Zaloška 2, SI-1000 Ljubljana, Slovenia. Phone: +386 1 5879 282; Fax: +386 1 5879 303; E-mail: bseruga@onko-i.si Disclosure: B. Seruga received honoraria for his advisory role at Sanofi. All remaining authors have declared no conflicts of interest. Background. Men with metastatic castrate-resistant prostate cancer (mCRPC) may not receive docetaxel in eve­ryday clinical practice due to comorbidities. Here we explore the impact of comorbidity on outcome in men with mCRPC treated with docetaxel in a population-based outcome study. Methods. Men with mCRPC treated with docetaxel at the Institute of Oncology Ljubljana between 2005 and 2012 were eligible. Comorbidity was assessed by the age-adjusted Charlson comorbidity index (aa-CCI) and adult comor­bidity evaluation (ACE-27) index. Hospital admissions due to the toxicity and deaths during treatment with docetaxel were used as a measure of tolerability. Association between comorbidity and overall survival (OS) was tested using the Cox proportional hazards analysis. Results. Two hundred and eight men were treated with docetaxel. No, mild, moderate and severe comorbidity was present in 2%, 32%, 53% and 13% using aa-CCI and in 27%, 35%, 29% and 8% when assessed by ACE-27. A substantial dose reduction of docetaxel occurred more often in men with moderate or severe comorbidity as compared to those with no or mild comorbidity. At all comorbidity levels about one-third of men required hospitalization or died during treatment with docetaxel. In univariate analysis a higher level of comorbidity was not associated with worse OS (aa-CCI HR 0.99; [95% CI 0.87–1.13], p = 0.93; ACE-27: HR 0.96; [95% CI 0.79–1.17], p = 0.69). Conclusions. Men with mCRPC, who have comorbidities may benefit from treatment with docetaxel. Key words: metastatic castration-resistant prostate cancer; comorbidity; chemotherapy Introduction Men with advanced prostate cancer are usually treated with hormonal therapy, which increases the risk for the development of comorbid condi­tions, such as diabetes, osteoporosis and cardiovas­cular disease.1-3 Most men with advanced prostate cancer receive hormonal therapy for several years and the average patient with metastatic castrate-re­sistant prostate cancer (mCRPC) in every-day clini­cal practice is 70 years old.4 Observational studies show that at this age more than 75% of cancer pa­tients have at least one comorbid condition, with more than 30% having moderate or severe comor­bidity.5 Based on improved overall survival (OS) in the TAX 327 and SWOG 9916 trials, docetaxel once every 3-weeks (hereafter Dq3w) in combination with prednisone is the standard treatment for men with mCRPC.6,7 A post-hoc analysis of the TAX327 showed that tolerability and efficacy of Dq3w ap­pear less favourable with advanced age.8 Men with mCRPC who are treated with docetaxel in every­day practice may be less selected, older and have 403 more comorbidities as compared to those treated with docetaxel in the pivotal randomized clinical trials.4,9 Outside of clinical trials, about 20-40% of men with mCRPC never receive treatment with docetaxel.10 Presence of comorbid conditions and/ or poor performance status may be the reasons that these patients are not treated with docetaxel.11-15 Although comorbidities are known negative prog­nostic factors for OS in men with early prostate cancer their prognostic role in men with mCRPC is less clear.11-13 We hypothesized that comorbidity has detrimental effect on the outcome of men with mCRPC who are treated with docetaxel. Due to the dose reductions, docetaxel may be less effective in comorbid men. Furthermore, treatment with doc­etaxel may increase the risk for toxic deaths in men with mCRPC. Here we explored the impact of co­morbidity on the efficacy and tolerability in men with mCRPC, who were treated with docetaxel in a population-based observational study. Patients and methods Study population and data collection In this population-based observational study we included men with mCRPC who were treated with docetaxel and subsequent systemic therapies at the Institute of Oncology Ljubljana between January 1, 2005 and June 27, 2012. Men with mCRPC, who receive docetaxel in routine clinical practice, have usually performance status (PS) 0–2 and therefore PS was not evaluated retrospectively in this study. Optimal dose intensity of docetaxel was 25 mg/m2/ week, which served as a denominator in the calcu­lation of the relative dose intensity (RDI). An optimal comorbidity index for prostate cancer patients is not established.16 We used the age-adjusted Charlson comorbidity index (aa-CCI) which is a composite index of 19 conditions weighted from 1 to 6 points and adjusted for age with each decade above 50 years of age counting for an extra point with a total score of 0-35 points. Point scores can then be classified into prognos­tic categories.17 Categories were formed using the same cut-off values as in the original publica­tion and, for easier differentiation between them, tagged as “none” (0 points), “mild” (1-2 points), “moderate” (3-4) and “severe” (> 4 points) comor­bidity. The second comorbidity index we used was the adult comorbidity evaluation-27 (ACE-27).18 The ACE-27 grades specific diseases and conditions into levels of comorbidity from grade 1 to grade 3. An overall level of comorbidity (“none”, “mild”, “moderate” or “severe”) is assigned based on the highest level of comorbidity. Only malignancies other than mCRPC were included in the final score of both comorbidity indices as its inclusion would have assigned a severe level of comorbidity to all patients. We retrieved relevant clinical information from electronic and hard copies of patients’ charts and assessed comorbidity by using the aa-CCI and ACE-27 coding protocols. In this retrospective study the number of hospi­tal admissions due to the toxicity of chemotherapy or deaths, which occurred during or 30 days after treatment discontinuation represented an estimate of tolerability of docetaxel. Dates of death were ob­tained from the national cancer registry. The protocol of our study was reviewed and approved for clinical use by the Ethics and Study Protocol Assessment Committee at the Institute of Oncology Ljubljana. Informed consent was obtained from all patients prior to the treatment; however, for including in this retrospective study it was waived. Statistical analysis Descriptive statistics were used to describe rel­evant characteristics of men at baseline. OS was calculated from the date of the first administration of docetaxel to death from any cause. Data were censored for patients who were alive at the cut-off date of March 13, 2013. OS was estimated using the Kaplan-Meier method. A Cox proportional hazard model was used to examine association between comorbidity and OS. Initial assessment was carried out in the univariable setting and subsequently for all significant (p < 0.1) variables in the multivari­able setting. Comorbidity was analysed as both cat­ egorical and dichotomous variable (score 0 vs. . 1). Discriminatory accuracy of the aa-CCI and ACE­27 in predicting death at 12 months or at any time during follow-up was tested by estimating the area under the receiver operating characteristic (ROC) curve (C-statistic). Association between comorbid­ity, RDI and tolerability was assessed by the Chi-square test. All tests were two-sided and a p-value of . 0.05 was considered statistically significant. No adjustment for multiple analyses was performed. Results Study population Our analysis included 208 men with mCRPC with median age of 69.9 years. Patients were treated 405 TABLE 2. Association of relative dose intensity of docetaxel and comorbidity <75% n (%) 0 0 10 (10%) 2 (8%) 3 (6%) 3 (4%) 5 (9%) 1 (8%) RDI 75-84% n (%) 85-94% n (%) 0 1 (33%) 2 (3%) 19 (29%) 19 (18%) 28 (28%) 3 (12%) 8 (33%) 3 (6%) 13 (24%) 10 (14%) 18 (26%) 11 (19%) 19 (33%) 0 6 (46%) .95 n (%) 2 (66%) 45 (68%) 44 (44%) 11 (46%) 35 (65%) 38 (55%) 23 (40%) 6 (46%) Median RDI - % 96% 98% 93% 93% 98% 96% 91% 94% aa-CCI = age-adjusted Charlson comorbidity index; ACE-27 = adult comorbidity evaluation 27; CI = confidence interval; RDI = relative dose intensity TABLE 3. Association of comorbidity and tolerability during treatment with docetaxel Number of patients 25 34 7 20 22 18 6 hospitalized or deceased 0 (38%) (34%) (29%) (37%) (32%) (31%) (46%) during treatment, n aa-CCI = age-adjusted Charlson comorbidity index; ACE-27 = adult comorbidity evaluation; CI = confidence interval TABLE 4. Comorbidity evaluation aa-CCI, N (%) 3 (2%) 67 (32%) 108 (53%) 27 (13%) ACE-27, N (%) 55 (27%) 73 (35%) 61 (29%) 16 (8%) aa-CCI = age-adjusted Charlson comorbidity index; ACE-27 = adult comorbidity evaluation 27 TABLE 5. Association of comorbidity and overall survival Association of comorbidity and efficacy After a median follow-up time of 14 months 133 men died. Median OS for the whole group was 19 months. For 98% of patients (N = 205) information on comorbidity was available. None, mild, mod­erate and severe comorbidity was present in 2%, 32%, 53% and 13% using aa-CCI and in 27%, 35%, 29% and 8% when assessed by ACE-27 (Table 4). In univariable analysis, a higher level of comor­bidity was not associated with worse OS (ACE-27: HR 0.96; [95% confidence interval (CI) 0.79–1.17], p = 0.69; aa-CCI HR 0.99; [CI 0.87–1.13], p = 0.93) when studied as a categorical variable (Table 5). Similarly, when analysed as dichotomous variable a higher level of comorbidity was not associated with worse OS (ACE-27: HR 0.75 [CI 0.51–1.08], p = 0.12; aa-CCI: HR 1.48 [CI 0.37–6.0], p = 0.58). Both indices were poor at discriminatory accuracy in predicting death at any time (C-statistics 0.45; p = 0.25 for aa-CCI and C-statistics 0.47; p = 0.44 Age (for every year) Log PSA (. vs. than median) Hb (per 10 units) ALP (. vs. < than median) Docetaxel (Dqw vs. Dq3w) Visceral metastases (yes vs. no) Opioids (yes vs. no) New agents (yes vs. no) aa-CCI (per category) ACE-27 (per category) 0.99 (0.97–1.01) p = 0.47 1.20 (1.07–1.35) p < 0.01 0.79 (0.72–0.86) p < 0.01 0.97 (0.87–1.10) p = 0.65 6.39 (3.19–12.81) p < 0.01 1.68 (1.08–2.61) p = 0.02 1.38 (0.65–2.92) P = 0.41 0.39 (0.19–0.80) p = 0.02 0.99 (0.87–1.13) p = 0.93 0.96 (0.79–1.17) p = 0.69 - 1.05 (0.94–1.18) p = 0.38 0.78 (0.72–0.86) p < 0.01 - 6.04 (2.77–13.20) p < 0.01 1.73 (1.10–2.73) p = 0.02 - 0.36 (0.17–0.74) p < 0.01 - - aa-CCI = age-adjusted Charlson comorbidity index; ACE-27 = adult comorbidity evaluation 27; Dqw = weekly docetaxel; Dq3w = 3-weekly docetaxel; HR = hazard ratio; CI = confidence interval 406 for ACE-27) or death within 12 months of start of treatment with docetaxel (C-statistics 0.47; p = 0.49 for aa-CCI and C-statistics 0.47; p = 0.49 for ACE­27). In multivariable analysis significant independ­ent predictors of poor OS were presence of visceral metastases (HR 1.73; p = 0.02) and weekly docetax­el schedule (HR 6.04; p < 0.01), whereas high Hb level (HR 0.78; p < 0.01) and use of abiraterone and/ or cabazitaxel (HR 0.36; p < 0.01) were independent favourable prognostic factors for OS (Table 5). Discussion One of the reasons that a substantial proportion of men with mCRPC are never treated with docetaxel is fear of poor tolerability due to comorbid condi­tions.10 Life expectancy of men with mCRPC cancer is limited and therefore the impact of comorbidity on the outcome may be less relevant in this set­ting as compared to men with early prostate can­cer. As patients enrolled into clinical trials usually do not have uncontrolled or severe comorbidities, population-based outcome studies may provide a more generalizable evaluation of the impact of co­morbidity on the outcome and tolerability of medi­cal intervention compared to post-hoc analyses of randomized trials. In our cohort of men with mCRPC treated with docetaxel, a substantial proportion had moderate or severe comorbidity. In this cohort, a substantial dose reduction of docetaxel occurred more often in men with moderate or severe comorbidity as com­pared to those with no or mild comorbidity. At all comorbidity levels about one-third of men required hospitalization or died during treatment with doc­etaxel. A higher level of comorbidity was not as­sociated with worse OS, irrespective of whether comorbidity was assessed by the aa-CCI or ACE­27 index. Both indices performed similarly poorly at discriminatory accuracy in predicting death. In concordance with our findings, investigators of the population-based outcome study in France, which enrolled elderly (. 75 years) men with mCRPC, did not find any association between comorbidity and outcome.19 To date several post-hoc analyses of rand­omized clinical trials, which evaluated the impact of comorbidity on the outcome in patients with advanced prostate cancer or other cancers, were reported. In a randomized phase II trial in which men with mCRPC were treated with docetaxel and prednisone with or without a bcl-2 antago­nist AT-101, comorbidity assessed by the CCI did not predict OS, both as a categorical or continuous variable.15 In this study patients with acute or un­controllable comorbidity were excluded and 53% had no other comorbidities. In contrast, in the post-hoc analysis of a large phase III randomized trial, which enrolled men with mCRPC receiving doc­etaxel and prednisone with or without bevacizum­ab, investigators found an association between the number of comorbidities at baseline and the risk of death.14 However, in that study comorbidity was not assessed by any comorbidity index. The lack of association between comorbidity assessed by the CCI and OS is also seen in other malignancies.20,21 Investigators in one of these studies cautioned that physicians must carefully discriminate between PS and comorbidity when assessing patients for therapy.21 According to the updated guidelines for the management of men with advanced prostate cancer published by the International Society of Geriatric Oncology (SIOG) comorbidity should be put into the context of patient’s general well-being and functional reserve.22 A higher level of comorbidity is a known risk factor for hospital admission in cancer patients.23,24 Irrespective of the level of comorbidity about one third of men required hospitalization or died dur­ing or shortly after treatment with docetaxel in our study. However, a higher proportion of men with moderate or severe comorbidity had a substantial dose reduction of docetaxel as compared to those with no or mild comorbidity (24-27% vs. 3-15%). Recent data show that patients with mCRPC treat­ed with docetaxel in routine practice experience more toxicity as compared to those treated with docetaxel within clinical trials.5 Similarly, high ad­mission rates during treatment with chemothera­py, as found in our study, were observed in older patients with other metastatic solid cancers.25,26 Accumulating evidence shows that anticancer therapies are less tolerable in patients treated in everyday clinical practise as compared to those treated within clinical trials. Recently, new treatment options with favoura­ble benefit-risk profile such as abiraterone acetate, enzalutamide and radium-223 became available for chemotherapy-naive men with mCRPC.27-29 These agents substantially changed the manage­ment of mCRPC as treatment with docetaxel may now be deferred or even omitted in some patients with very advanced prostate cancer. However, we believe that docetaxel remains an important treat­ment option for mCRPC and therefore our findings may still be clinically relevant. 407 Our study has several limitations. First, our ret­rospective analysis is based on a relatively small cohort of men with mCRPC, who were all treated at a single institution. However, all patients with mCRPC in Slovenia who are candidates for treat­ment with docetaxel are referred to the Institute of Oncology Ljubljana. Therefore, our study is less prone to selection and referral biases which often plague traditional institutional retrospective stud­ies. Second, when assessing comorbidity using aa-CCI and ACE-27 coding protocols, we retrieved relevant clinical information retrospectively from electronic and hard copies of patient’s charts, which may have limitations. An ideal set-up to study the association between comorbidity and outcome in men with mCRPC would be a prospective study design. Third, although serum PSA and ALP levels and pain are well established prognostic factors in men with mCRPC, they did not independently pre­dict the outcome in our study. Similar larger study might lead into different conclusions and more gen­eralizable results. Fourth, use of alternative comor­bidity indexes might lead to different results and conclusions. We used aa-CCI and ACE-27, as these are convenient, widely used and validated tools. Fifth, the rate of hospitalization during treatment might be underestimated as admissions to com­munity hospitals might not always be recorded in patients’ charts. Finally, use of abiraterone acetate and/or cabazitaxel after treatment with docetaxel in 22% of patients might impact the results. However, these new agents were not used more often in men with moderate or severe comorbidities as compared to men with no or minor comorbidities (Table 1). In conclusion, the presence of comorbidity may not be associated with worse outcome in men with mCRPC, who are treated with docetaxel. Irrespective of the level of comorbidity a substan­tial proportion of men (about one third) required hospitalization or died during therapy with doc­etaxel. Dose reduction occurred more often in men with moderate or severe comorbidity. It is likely that, men with mCRPC who have substantial co­morbidities may still benefit from reduced dose of docetaxel. Comorbid conditions should always be interpreted in the context of other relevant clinical characteristics when deciding about therapy with docetaxel in men with mCRPC. Author contributions Concept and design - A. Zist, B. Seruga; data col­lection - A. Zist; analysis and interpretation of data - A. Zist, E. Amir, A. Ocana, B. Seruga; manuscript writing and approval - A. Zist, E. Amir, A. Ocana, B. Seruga Acknowledgements Results of this analysis were in part presented at the European Cancer Congress 2013 in Amsterdam (abstract ID 2919)­ This work was supported by the grant P3-0321 funded by the Slovenian Research Agency. References 1. Jespersen CG, Norgaard M, Borre M. Androgen-deprivation therapy in treatment of prostate cancer and risk of myocardial infarction and stroke: a nationwide Danish population-based cohort study. Eur Urol 2014; 65: 704-9. 2. Greenspan SL, Coates P, Sereika SM, Nelson JB, Trump DL, Resnick NM. Bone loss after initiation of androgen deprivation therapy in patients with prostate cancer. J Clin Endocrinol Metab 2005; 90: 6410-17. 3. Keating NL, O’Malley AJ, Smith MR. Diabetes and cardiovascular disease during androgen deprivation therapy for prostate cancer. J Clin Oncol 2006; 24: 4448-56. 4. Templeton AJ, Vera-Badillo FE, Wang L, Attalla M, Gouveia P, Leibowitz-Amit R, et al. Translating clinical trials to clinical practice: outcomes of men with metastatic castration resistant prostate cancer treated with docetaxel and prednisone in and out of clinical trials. Ann Oncol 2013; 24: 2972-77. 5. Piccirillo JF, Vlahiotis A, Barrett LB, Flood KL, Spitznagel EL, Steyerberg EW. The changing prevalence of comorbidity across the age spectrum. Crit Rev Oncol Hematol 2008; 67: 124-32. 6. Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004; 351: 1502-12. 7. Petrylak DP, Tangen CM, Hussain MH, Lara PN Jr, Jones JA, Taplin ME, et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med 2004; 351: 1513-20. 8. Horgan AM, Seruga B, Pond GR, Alibhai SM, Amir E, De Wit R, et al. Tolerability and efficacy of docetaxel in older men with metastatic castrate-resistant prostate cancer (mCRPC) in the TAX 327 trial. J Geriatr Oncol 2014; 5: 119-26. 9. Chao HH, Mayer T, Concato J, Rose MG, Uchio E, Kelly WK. Prostate cancer, comorbidity, and participation in randomized controlled trials of therapy. J Investig Med 2010; 58: 566-8. 10. Harris V, Lloyd K, Forsey S, Rogers P, Roche M, Parker C. A population-based study of prostate cancer chemotherapy. Clin Oncol (R Coll Radiol) 2011; 23: 706-8. 11. Lund L, Borre M, Jacobsen J, Sorenson HT, Norgaard M. Impact of comorbid­ity on survival of Danish prostate cancer patients, 1995-2006: a population-based cohort study. Urology 2008; 72: 1258-62. 12. Albertsen PC, Moore DF, Shih W, Lin Y, Lu-Yao GL. Impact of comorbidity on survival among men with localized prostate cancer. J Clin Oncol 2011; 29: 1335-41. 13. Daskivich TJ, Chamie K, Kwan L, Labo J, Dash A, Greenfield S, et al. Comorbidity and competing risks for mortality in men with prostate cancer. Cancer 2011; 117: 4642-50. 14. Halabi S, Kelly WK, George DJ, Morris MJ, Kaplan EB, Small EJ. Comorbidities predict overall survival (OS) in men with metastatic castrate-resistant pros­tate cancer. [Abstract]. J Clin Oncol 2011; 29(Suppl 7): abstr 189. 15. Goyal J, Pond GR, Galsky MD, Hendricks R, Small A, Tsao CK, et al. Association of the Charlson comorbidity index and hypertension with sur­vival in men with metastatic castration-resistant prostate cancer. Urol Oncol 2014; 32: 36. e27-34. 408 16. Alibhai SM, Leach M, Tomlinson GA, Krahn MD, Fleshner NE, Naglie G. Is there an optimal comorbidity index for prostate cancer? Cancer 2008; 112: 1043-50. 17. Charlson M, Szatrowski TP, Peterson J, Gold J. Validation of a combined comorbidity index. J Clin Epidemiol 1994; 47: 1245-51. 18. Piccirillo JF, Tierney RM, Costas I, Grove L, Spitznagel EL Jr. Prognostic impor­tance of comorbidity in a hospital-based cancer registry. JAMA 2004; 291: 2441-7. 19. Italiano A, Ortholan C, Oudard S, Pouessel D, Gravis G, Beuzeboc P, et al. Docetaxel-based chemotherapy in elderly patients (age 75 and older) with castration-resistant prostate cancer. Eur Urol 2009; 55: 1368-75. 20. Vickers MM, Powell ED, Asmis TR, Jonker DJ, Hilton JF, O`Callaghan CJ, et al. Comorbidity, age and overall survival in patients with advanced pancreatic cancer - results from NCIC CTG PA.3: a phase III trial of gemcitabine plus erlotinib or placebo. Eur J Cancer 2012; 48: 1434-42. 21. Asmis TR, Powell E, Karapetis CS, Jonker DJ, Tu D, Jeffery M, et al. Comorbidity, age and overall survival in cetuximab-treated patients with advanced colorectal cancer (ACRC)--results from NCIC CTG CO.17: a phase III trial of cetuximab versus best supportive care. Ann Oncol 2011; 22: 118-26. 22. Droz JP, Aapro M, Balducci L, Boyle H, Van den Broeck T, Cathcart P, et al. Management of prostate cancer in older patients: updated recommenda­tions of a working group of the International Society of Geriatric Oncology. Lancet Oncol 2014; 15: e404-14. 23. Donze J, Lipsitz S, Bates DW, Schnipper JL. Causes and patterns of readmis­sions in patients with common comorbidities: retrospective cohort study. BMJ 2013; 347: f7171. 24. Hernandez C, Jansa M, Vidal M, Nunez M, Bertran MJ, Garcia-Avmerich J, et al. The burden of chronic disorders on hospital admissions prompts the need for new modalities of care: a cross-sectional analysis in a tertiary hospital. QJM 2009; 102: 193-202. 25. Zauderer MG, Sima CS, Korc-Grodzicki B, Kris MG, Krug LM. Toxicity of initial chemotherapy in older patients with lung cancers. J Geriatr Oncol 2013; 4: 64-70. 26. Aparicio T, Jouve JL, Teillet L, Gargot D, Subtil F, Le Bruna-Ly V, et al. Geriatric factors predict chemotherapy feasibility: ancillary results of FFCD 2001-02 phase III study in first-line chemotherapy for metastatic colorectal cancer in elderly patients. J Clin Oncol 2013; 31: 1464-70. 27. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med 2014; 371: 424-33. 28. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med 2013; 368: 138-48. 29. Parker C, Nilsson S, Heinrich D, Helle SI, O`Sullivan JM, Fossa SD, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013; 369: 213-23. 409 research article Clinical impact of post-progression survival on overall survival in patients with limited-stage disease small cell lung cancer after first-line chemoradiotherapy Norimitsu Kasahara1,6, Hisao Imai1,6, Kyoichi Kaira1,7, Keita Mori2, Kazushige Wakuda1, Akira Ono1, Tetsuhiko Taira1, Hirotsugu Kenmotsu1, Hideyuki Harada3, Tateaki Naito1, Haruyasu Murakami1, Masahiro Endo4, Takashi Nakajima5, Masanobu Yamada6, Toshiaki Takahashi1 1 Division of Thoracic Oncology, 2 Clinical Trial Coordination Office, 3 Division of Radiation Oncology, 4 Division of Diagnostic Radiology, 5 Division of Diagnostic Pathology, Shizuoka Cancer Center, Japan 6 Department of Medicine and Molecular Science, 7Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Japan Radiol Oncol 2015; 49(4): 409-415. Received 31 January 2015 Accepted 06 July 2015 Correspondence to: Hisao Imai, M.D., Ph.D., Division of Thoracic Oncology, Shizuoka Cancer Center, 1007 Shimonagakubo, Nagaizumi-chou, Suntou-gun, Shizuoka 411-8777, Japan. Phone: +81 55 989 5222; Fax: +81 55 989 5783; E-mail: m06701014@gunma-u.ac.jp Disclosure: The authors declare no conflict of interest. N. Kasahara and H. Imai contributed equally to this study. Background. The effects of first-line chemoradiotherapy on overall survival (OS) may be confounded by subsequent lines of therapy in patients with limited-stage disease small cell lung cancer (LD-SCLC). Therefore, we aimed to de­termine the relationships between progression-free survival (PFS), post-progression survival (PPS) and OS after first-line chemoradiotherapy in LD-SCLC patients. Patients and methods. We retrospectively analyzed 71 LD-SCLC patients with performance status (PS) 0-2 who received first-line chemoradiotherapy and had disease recurrence between September 2002 and March 2013 at Shizuoka Cancer Center (Shizuoka, Japan). We determined the correlation between PFS and OS and between PPS and OS at the individual level. In addition, we performed univariate and multivariate analyses to identify significant prognostic factors of PPS. Results. OS is more strongly correlated with PPS (Spearman’s r = 0.86, R2 = 0.72, p < 0.05) than PFS (Spearman’s r = 0.46, R2 = 0.38, p < 0.05). In addition, the response to second-line treatments, the presence of distant metastases at recurrence and the number of additional regimens after first-line chemoradiotherapy were significant independent prognostic factors for PPS. Conclusions. PPS has more impact on OS than PFS in recurrent LD-SCLC patients with good PS at beginning of the treatment. Moreover, treatments administered after first-line chemoradiotherapy may affect their OS. However, larger multicenter studies are needed to validate these findings. Key words: chemoradiotherapy; limited-stage disease small cell lung cancer; overall survival; post-progression sur­vival; progression-free survival Introduction Lung cancer is the leading cause of cancer-related mortality worldwide.1 In the United States, 14% of people who were diagnosed with lung cancer had small cell lung cancer (SCLC).2 Approximately 30% of SCLC patients have limited-stage disease small cell lung cancer (LD-SCLC), which is characterized by locoregional tumors in the hemithorax, medi­astinum, or supraclavicular lymph nodes, while the rest have extensive-stage disease.3 Current therapeutic options for LD-SCLC include combi­ 410 nation chemotherapy with etoposide and cisplatin, chest radiotherapy, and prophylactic cranial irradi­ation (PCI).4,5 However, due to the limited efficacy of these therapeutic strategies and the aggressive nature of SCLC tumors, the prognosis for SCLC patients is poor; the median survival time for LD­SCLC patients is less than two years.6-8 PFS and OS are two common endpoints in can­cer trials. OS is usually preferred, because it is relia­ble, precise, meaningful and easily documented by the date of death.9 However, the effect of first-line treatments on OS might be confounded by subse­quent lines of therapy. In contrast, PFS is quicker to measure, can be measured more conveniently, and therefore, may be easier to assess than OS.10 If there is a strong correlation between PFS and OS, then PFS may be a surrogate endpoint for OS. In non-small cell lung cancer (NSCLC), increases in PFS do not necessarily increase OS, but post-progression survival (PPS) is strongly associated with OS after first-line treatment.11-13 We have also demonstrated a strong correlation between PPS and OS after first-line chemotherapy in patients with extensive-stage disease SCLC.14 In LD-SCLC, though, the relation­ship between PPS and OS is unknown. Therefore, we analyzed the correlation between PFS and OS and between PPS and OS after first-line chemoradiotherapy in LD-SCLC patients to determine whether PFS or PPS has more influence on OS. We also investigated the prognostic value of baseline and tumor characteristics for PPS. Patients and methods Patients We retrospectively enrolled 71 consecutive patients with recurrent LD-SCLC after receiving first-line chemoradiotherapy at Shizuoka Cancer Center (Shizuoka, Japan) between September 2002 and March 2013. The inclusion criteria were as follows: (1) histologically or cytologically confirmed SCLC; (2) 20 years of age or older at the time of chemo­radiotherapy; (3) Eastern Cooperative Oncology Group performance status (PS) of 0–2 at the begin­ning of the first-line treatment; (4) first-line treat­ ment with . 40 Gy curative thoracic radiotherapy and platinum doublet chemotherapy, either con­currently or sequentially; and (5) disease recur­rence after first-line treatment. The study protocol was approved by the Institutional Review Board of Shizuoka Cancer Center and was conducted in accordance with the Helsinki Declaration of 1964 (revised 2008). Due to the retrospective nature of this study, the need for informed consent was waived. Treatment LD-SCLC patients were treated with a combination of chemotherapy and radiotherapy. Several dif­ferent first-line chemotherapeutic regimens were used; etoposide (80 or 100 mg/m2) was adminis­tered on days 1–3 in combination with cisplatin (80 mg/m2) on day 1, cisplatin (25 mg/m2) on days 1–3, or carboplatin (area under the curve = 5) on day 1. These drugs were injected intravenously every 3–4 weeks for maximum 4 courses. Second and third-line treatments included amrubicin, irinotecan, to­potecan, gemcitabine, and paclitaxel. The fractionation schedule for thoracic radio­therapy in LD-SCLC patients was determined by using information from chest computed tomogra­phy (CT) to calculate the pretreatment tumor vol­ume. Typically, the total planned dose was 50 Gy when it was fractionated once daily or 45 Gy when it was fractionated twice daily, based on individual physician decision. Furthermore, the maximum spinal cord dose was limited to 45 Gy when the ra­diation dose was fractionated once daily or to 36 Gy when it was fractionated twice daily. In addi­tion, no more than 35% of the normal lung volume received more than 20 Gy. Thoracic radiotherapy was started either during the first cycle of chemotherapy or after four cycles of chemotherapy. It was suspended if a patient ex­perienced grade 4 thrombocytopenia, neutropenia, radiation pneumonitis, fever caused by infection, a decrease of more than 10 mmHg in arterial oxygen pressure, or difficulty swallowing liquids. After thoracic radiotherapy, PCI (25 Gy in 10 fractions) was administered to patients with a complete or near-complete response, as shown by a scar-like shadow on a chest CT, if the treating physician rec­ommended it. Assessment of treatment efficacy Tumor responses reflect the best overall response and maximum shrinkage. Radiographic tumor re­sponses were evaluated using chest computed to­mography at every two courses of chemotherapy according to the Response Evaluation Criteria In Solid Tumors 1.0 as follows: complete response (CR), disappearance of all target lesions; partial re­ sponse (PR), . 30% decrease in the total diameter of all target lesions relative to the total baseline di­ ameter; progressive disease (PD), . 20% increase in 411 the total diameter of all target lesions relative to the smallest total diameter observed during the study; and stable disease (SD), insufficient change in the total diameter of all target lesions to qualify as PR or PD.15 PFS was defined as the time from the begin­ning of first-line treatment until documented PD or death. In addition, OS was reported as the time from the beginning of first-line treatment until death or censored at the time of the last assessment of disease status. Similarly, PPS was documented as the time from tumor progression after first-line treatment until death or censored at the time of the last assessment of disease status. Treatment-free interval In this study, we defined treatment-free interval (TFI) as the period from the date of completion of first-line treatment to first relapse. When sequen­tial radiotherapy or PCI were performed as first-line treatment, the date of completion of first-line treatment was defined as the last day of these treat­ments. Since TFI is known as a predictive factor of second-line chemotherapy, we analyzed patients according to TFI.16, 17 In many trials, the relapsed SCLC patients with TFI more than 90 days were defined as sensitive relapses. This definition was also used in this study. Statistical analyses We used Spearman’s rank correlation and linear regression analyses to determine whether PFS or PPS correlated with OS in LD-SCLC patients. We also applied the Cox proportional hazards model with a stepwise regression procedure to determine prognostic factors for PPS and estimate hazard ra­tios and 95% confidence intervals. The effects of different prognostic factors on PPS were compared using the log-rank test. P-values less than 0.05 were considered to be statistically significant for both one-tailed and two-tailed tests. All statistical analyses were performed using JMP (version 11.0; SAS Institute, Cary, NC, USA). Results Patient characteristics and treatment efficacy Between September 2002 and March 2013, 116 pa­tients with LD-SCLC were treated with chemoradio- TAble 1. Patient characteristics Gender Male 57 Female 14 Age (years) Median 69 Range 45–92 Performance status 0 32 1 37 2 2 Clinical stage II 8 III 63 Tumor histology Small cell carcinoma 68 Combined small cell carcinoma 3 Smoking history Current or former smoker 70 Never smoked 1 Number of first-line chemotherapy courses 1 2 2 1 3 4 4 63 5 1 Number of regimens after first-line treatment 0 18 1 21 2 16 3 8 4 4 5 2 6 2 Radiation dose (Gy) Median 45 Range 40–60 Chemoradiotherapy Concurrent 56 Sequential 15 First-line chemotherapy regimens Cisplatin + etoposide 49 Carboplatin + etoposide 18 Cisplatin + etoposide › Cisplatin + irinotecan 3 Cisplatin + etoposide › Cisplatin + Vincristine + Doxorubicin + etoposide Subsequent lines of chemotherapy, total (second-line/third-line or more) Platinum combination 25 (15/10) Amrubicin 36 (22/14) Irinotecan 25 (9/16) Topotecan 13 (7/6) Gemcitabine 7 (0/7) Paclitaxel 6 (0/6) Investigational drug 2 (0/2) Distant metastases at recurrence Yes 48 No 23 Prophylactic cranial irradiation Yes 27 No 44 412 therapy, and 71 patients who recurred after first-line treatment were enrolled in this study. Patient char­acteristics are summarized in Table 1. The majority of patients (80.3%) received concurrent chemothera­py and radiotherapy. Cisplatin plus etoposide com­bination chemotherapy was the most common first-line treatment. Subsequently, 21/71 (29.6%) patients received a median of one additional regimen (range: 0–6). Twenty-one patients temporarily interrupted RT, but all of them completed previously planned radiation doses. During a median follow-up peri­od of 19.1 months (range: 8.0–118.3 months), 63/71 (88.7%) patients died. Nine patients experienced a CR, 56 patients had a PR, three patients showed SD, and three patients exhibited PD. The overall response rate was 91.5% and the disease control rate was 95.7%. The median PFS and OS were 8.8 months and 21.6 months, respectively (Figures 1A, 1B). The mean OS of other 45 patients who didn’t experience recurrence after first-line treatment was 46.5 months (median not reached). Prognostic factors for post-progression survival Since OS was more strongly correlated with PPS (Spearman’s r = 0.86, R2 = 0.72, p < 0.05; Figure 2B) than PFS (Spearman’s r = 0.46, R2 = 0.38, p < 0.05; Figure 2A), we assessed the significance of poten­tial prognostic factors for PPS. Univariate analysis showed that six factors, namely, age at the be­ginning of first-line treatment, relative timing of chemotherapy and radiotherapy (sequential vs. concurrent), response to second-line treatment (non PD vs. PD), the presence of distant metastases at recurrence (yes vs. no), administration of plati­num-based chemotherapeutic agents after first-line treatment (yes vs. no), and the number of regimens after first-line treatment, were significantly associ­ated with PPS (p < 0.05; Table 2). However, multi­variate analysis revealed that only the response to second-line treatment (non PD vs. PD), the pres­ence of distant metastases at recurrence (yes vs. no) and the number of additional regimens after first-line treatment are significant independent prog­nostic factors for PPS (Table 3). We used these three prognostic factors to con­struct Kaplan-Meier plots of PPS (Figures 3A, 3B and 3C), which showed that the survival distribu­tions for response to second-line treatment (non PD vs. PD), the presence of distant metastases at recurrence (yes vs. no) and the number of addi­tional regimens after first-line treatment (< 2 vs. . 2) are significantly different (log-rank tests, p < 413 TAble 2. Univariate analysis of factors associated with post-progression survival in limited-stage small cell lung cancer patients Gender 1.42 0.78–2.81 0.25 Age (years) at the beginning of first-line treatment 1.03 1.00–1.06 0.03 Age (years) at the beginning of second-line treatment 1.02 0.99–1.06 0.10 PS at the beginning of first-line treatment 0.90 0.55–1.48 0.69 PS at the end of first-line treatment 0.77 0.47–1.25 0.29 PS at the beginning of second-line treatment 1.31 0.83–2.03 0.23 Tumor histology (small cell carcinoma/combined small cell carcinoma) 1.55 0.63–5.16 0.36 Clinical stage at the beginning of first-line treatment (II/III) 0.55 0.22–1.15 0.12 Chemoradiotherapy (sequential/concurrent) 2.21 1.14–3.99 0.01 Number of courses of first-line chemotherapy 1.08 0.75–1.77 0.69 Best response at first-line treatment PR/ nonPR 0.98 0.45–2.57 0.97 NonPD /PD 1.33 0.49–5.48 0.61 Best response at second-line treatment PR/ nonPR 0.63 0.31–1.21 0.17 NonPD/PD 0.23 0.11–0.45 < 0.01 Treatment-free interval Sensitive/refractory 0.87 0.49–1.64 0.65 Distant metastases at recurrence (yes/no) 1.77 1.05–3.10 0.03 Administration of platinum-based agents after first-line treatment (yes/no) 0.51 0.28–0.88 0.01 Administration of amrubicin after first-line treatment (yes/no) 0.71 0.39–1.28 0.25 Prophylactic cranial irradiation (yes/no) 0.75 0.44–1.25 0.28 Number of regimens after first-line treatment 0.84 0.71–0.98 0.02 CI = confidence interval; PD = progressive disease; PR = partial response; PS = performance status. Boldfaced p-values are statistically significant (p < 0.05). TAble 3. Multivariate analysis of factors associated with post-progression survival in limited-stage small cell lung cancer patients Age (years) at the beginning of first-line treatment 0.98 0.94–1.02 0.47 Chemoradiotherapy (sequential/concurrent) 2.25 0.66–7.04 0.18 Best response at second-line treatment (NonPD/PD) 0.22 0.10–0.47 < 0.01 Distant metastases at recurrence (yes/no) 2.42 1.18–5.22 0.01 Administration of platinum-based agents after first-line treatment (yes/no) 0.92 0.41–1.98 0.83 Number of regimens after first-line treatment 0.75 0.56–0.98 0.04 CI = confidence interval; PD = progressive disease. Boldfaced p-values are statistically significant (p < 0.05). 0.05). Specifically, the median PPS in patients with-for patients with distant metastases (8.7 months; out PD after second-line treatment (17.5 months) p < 0.05). In addition, the median PPS of patients was significantly greater than that for patients who received two or more regimens after first-line with PD (6.9 months; p < 0.05). Furthermore, the treatment (16.0 months) was significantly greater median PPS in patients without distant metastases than that for patients who received less than two (17.3 months) was significantly greater than that additional regimens (6.8 months; p < 0.05). 414 FIGURe 3. Three significant independent prognostic factors of post-progression survival (PPS) (Table 3) result in significantly different PPS distributions in 71 limited-stage disease small cell lung cancer (LD-SCLC) patients (log rank test, p < 0.05). (A) Response to second-line treatment (progressive disease [PD] vs. non progressive disease [non PD]). Median PPS for non PD: 17.5 months vs. PD: 6.9 months. (b) Presence of distant metastases at recurrence (Yes vs. No). Median PPS for Yes: 8.7 months vs. No: 17.3 months. (C) Number of regimens after first-line treatment. Median PPS for . 2 additional regimens: 16.0 months vs. < 2 regimens: 6.8 months. Discussion In this study, we examined the relationships be­tween OS and PFS or PPS, for recurrent LD-SCLC patients after first-line chemoradiotherapy and found that OS correlates more strongly with PPS than PFS. In addition, we determined that the re­sponse to second-line treatment, the presence of distant metastases at recurrence and the number of additional regimens after first-line treatment are significant independent prognostic factors for PPS. To our knowledge, this is the first report of indi­vidual-level factors that affect PPS for LD-SCLC patients after first-line chemoradiotherapy. Several previous meta-analyses have assessed the value of surrogate endpoints, such as time to progression for survival in cancer studies.18,19 In extensive-stage disease SCLC, tumor response and PFS have been proposed as potential surro­gate endpoints for OS, but their appropriateness is controversial in LD-SCLC.20 Computer simulations have shown that significance of OS may be diluted if PPS is long.9 Other studies have also demonstrat­ed that PPS is strongly correlated with OS for ad­vanced NSCLC after both first-line chemotherapy and subsequent lines of therapy.12,13, 21 Similarly, we have previously reported that PPS is a potential surrogate marker for advanced NSCLC and exten­sive-stage disease SCLC.14, 22 Our finding that OS is more strongly corre­lated with PPS than PFS implies that subsequent treatments have more effects on OS than the first line treatment. Therefore, LD-SCLC clinical trials should account for factors that may affect PPS to avoid confounding OS. Actually, this recommen­dation may apply to SCLC in general, because the two of three significant independent prognostic factors associated with PPS for LD-SCLC patients that we identified in this study, namely, response to second-line treatment and the number of addi­tional regimens after first-line chemotherapy, are also associated with PPS in extensive-stage SCLC patients.14 These prognostic factors for PPS also suggest that disease stabilization after disease progression following first-line chemoradiotherapy may al­low LD-SCLC patients to receive additional lines of treatment, which could prolong PPS, and con­sequently, OS. Although a number of treatment choices in SCLC are less than that of NSCLC, the large number of treatment regimens that were used after first-line chemoradiotherapy in this study is mainly due to the increasing number of chemo­therapeutic options, such as amrubicin, irinotecan, and topotecan, for subsequent-line chemotherapy for LD-SCLC. However, treatments with platinum-based chemotherapeutic agents and amrubicin after first-line treatment were not significant prog­nostic factors for PPS, which suggest that these drugs do not affect PPS or OS. Likewise, treatment with sequential or concurrent chemoradiotherapy was not a significant prognostic factor for PPS; however, relative few patients in this study were 415 treated sequentially, so there may have been insuf­ficient statistical power to detect a significant dif­ference. This study has three major limitations. First, the sample size was relatively small. This limitation is difficult to overcome, particularly in studies that analyze patients with similar backgrounds, be­cause there are relatively few LD-SCLC patients at any given institution. Nevertheless, our institution treats a fair number of these cases and uses uni­fied treatment regimens. Second, the single-center design of our study may limit the generality of our conclusions, so multicenter trials are needed to val­idate our results in larger patient populations and other clinical settings. Third, since different physi­cians documented patient responses, the timing of evaluation of PFS and tumor response rates may have been less accurate than if only a single phy­sician had documented all responses. However, this is one of the major limitations of retrospective study, and it is unavoidable. Prospective trials are needed to investigate the validity. In conclusion, PPS has more impact on OS than PFS in recurrent LD-SCLC patients after first-line chemoradiotherapy. In addition, the response to second-line treatment, the presence of distant me­tastases at recurrence and the number of additional regimens after first-line treatment are significant independent prognostic factors for PPS. These re­sults suggest that treatments administered after first-line chemoradiotherapy affect OS in LD-SCLC patients. However, larger multicenter studies are needed to validate these conclusions in other pa­tient populations and clinical settings. Acknowledgements We thank Drs. Takuya Oyakawa, Ryo Koh, Shota Ohmori, and Kazuhisa Nakashima for their assis­tance in preparing this manuscript. References 1. DeSantis CE, Lin CC, Mariotto AB, Siegel RL, Stein KD, Kramer JL, et al. Cancer treatment and survivorship statistics 2014. CA Cancer J Clin, 2014; 64: 252-71. 2. American Cancer Society. Cancer Facts & Figures 2014. Atlanta, GA: American Cancer Society; 2014. 3. National Cancer Institute. Small cell lung cancer treatment. [citated 2015 Jan 15]. Available at: http://www.cancer.gov/cancertopics/pdq/treatment/ small-cell-lung/healthprofessional/page1/AllPages#Section_267. 2014. 4. Pignon JP, Arriagada R, Ihde DC, Johnson DH, Perry MC, Souhami RL, et al.: A meta-analysis of thoracic radiotherapy for small-cell lung cancer. N Engl J Med 1992; 327: 1618-24. 5. Warde P, Payne D. Does thoracic irradiation improve survival and local control in limited-stage small-cell carcinoma of the lung? A meta-analysis. J Clin Oncol 1992; 10: 890-5. 6. von Pawel J, Schiller JH, Shepherd FA, Fields SZ, Kleisbauer JP, Chrysson NG, et al. Topotecan versus cyclophosphamide, doxorubicin, and vincristine for the treatment of recurrent small-cell lung cancer. J Clin Oncol 1999; 17: 658-67. 7. O’Brien ME, Ciuleanu TE, Tsekov H, Shparyk Y, Cucevia B, Juhasz G, et al. Phase III trial comparing supportive care alone with supportive care with oral topotecan in patients with relapsed small-cell lung cancer. J Clin Oncol 2006; 24: 5441-7. 8. Eckardt JR, von Pawel J, Pujol JL, Papai Z, Quoix E, Ardizzoni A, et al. Phase III study of oral compared with intravenous topotecan as second-line therapy in small-cell lung cancer. J Clin Oncol 2007; 25: 2086-92. 9. Broglio KR, Berry DA. Detecting an overall survival benefit that is derived from progression-free survival. J Natl Cancer Inst 2009; 101: 1642-9. 10. Soria JC, Massard C, Le Chevalier T. Should progression-free survival be the primary measure of efficacy for advanced NSCLC therapy? Ann Oncol 2010; 21: 2324-32. 11. Reck M, von Pawel J, Zatloukal P, Ramlau R, Gorbounova V, Hirsh V, et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevaci­zumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil. J Clin Oncol 2009; 27: 1227-34. 12. Hotta K, Kiura K, Fujiwara Y, Takigawa N, Hisamoto A, Ichihara E, et al. Role of survival post-progression in phase III trials of systemic chemotherapy in advanced non-small-cell lung cancer: a systematic review. PloS one 2011; 6: e26646. 13. Hayashi H, Okamoto I, Morita S, Taguri M, Nakagawa K. Postprogression survival for first-line chemotherapy of patients with advanced non-small-cell lung cancer. Ann Oncol 2012; 23: 1537-41. 14. Imai H, Mori K, Wakuda K, Ono A, Akamatsu H, Shukuya T, et al. Progression-free survival, post-progression survival, and tumor response as surrogate markers for overall survival in patients with extensive small cell lung cancer. Ann Thorac Med 2015; 10: 61-6. 15. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000; 92: 205-16. 16. Giaccone G, Donadio M, Bonardi G, Testore F, Calciati A. Teniposide in the treatment of small-cell lung cancer: the influence of prior chemotherapy. J Clin Oncol 1988; 6: 1264-70. 17. Ebi N, Kubota K, Nishiwaki Y, Hojo F, Matsumoto T, Kakinuma R, et al. Second-line chemotherapy for relapsed small cell lung cancer. Jpn J Clin Oncol 1997; 27: 166-9. 18. Johnson KR, Ringland C, Stokes BJ, Anthony DM, Freemantle N, Irs A, et al. Response rate or time to progression as predictors of survival in trials of metastatic colorectal cancer or non-small-cell lung cancer: a meta-analysis. Lancet Oncol 2006; 7: 741-6. 19. Hotta K, Fujiwara Y, Matsuo K, Kiura K, Takigawa N, Tabata M, et al. Time to progression as a surrogate marker for overall survival in patients with advanced non-small cell lung cancer. J Thorac Oncol 2009; 4: 311-7. 20. Foster NR, Qi Y, Shi Q , Krook JE, Kugler JW, Jett JR, et al. Tumor response and progression-free survival as potential surrogate endpoints for overall survival in extensive stage small-cell lung cancer: findings on the basis of North Central Cancer Treatment Group trials. Cancer 2011; 117: 1262-71. 21. Hayashi H, Okamoto I, Taguri M, Morita S, Nakagawa K. Post progression survival in patients with advanced non-small-cell lung cancer who receive second-line or third-line chemotherapy. Clin Lung Cancer 2013; 14: 261-6. 22. Imai H, Takahashi T, Mori K, Ono A, Akamatsu H, Shukuya T, et al. Individual-level data on the relationships of progression-free survival, post-progression survival, and tumor response with overall survival in patients with advanced non-squamous non-small cell lung cancer. Neoplasma 2014; 61: 233-40. Radiol Oncol 2015; 49(4): 315-319. doi:10.1515/raon-2015-0036 Imunotoksin – nova možnost za zdravljenje bolnikov s ponovljenim ali neodzivnim Hodgkinovim limfomom Jezeršek Novaković B Izhodišča. Čeprav je Hodgkinov limfom večinoma ozdravljiva bolezen, se pri nekaterih bolnikih ne odziva na zdravljenje ali pa se ponovi po uspešnem prvem zdravljenju. Pri bolnikih s ponovljenim ali neodzivnim Hodgkinovim limfomom dosežemo dolgotrajen odgovor na zdravljenje in začasno izboljšanje bolezni pri približno polovici bolnikov s t.i. “reševalno” kemoterapijo, ki ji sledi visokodozna kemoterapija in avtologna presaditev krvotvornih matičnih celic. Po drugi strani pa imajo bolniki s po­novitvijo bolezni po takšnem zdravljenju ter tisti, pri katerih je bilo zdravljenje z vsaj dvema redoma kombinirane kemoterapije neuspešno in niso bili primerni za visokodozno kemoterapijo, zelo omejene možnosti nadaljnjega zdravljenja. Zaključki. Nova možnost za zdravljenje teh bolnikov s ponovljenim ali neodzivnim Hodgkinovim limfomom je imunotoksin brentuksimab vedotin. Sestavljen je iz monoklonalnega protitelesa usmerjenega proti CD30, na katerega je vezana protitu­bulna učinkovina monometil auristatin E. To zdravilo je bilo učinkovito in je imelo sprejemljivo toksičnost. V registracijski raziskavi je bil celokupni odgovor na zdravljenje 75 % s 34 % popolnih odgovorov. Srednja vrednost trajanja odgovora je bila 20,5 mesecev pri bolnikih s popolnim odgovorom in 6,7 mesecev pri vseh bolnikih, ki so odgovorili na zdravljenje. Srednja vrednost celokupnega preživetja je bila 40,5 mesecev (3-letno celokupno preživetje 54 %) in srednja vrednost brez napredovanja bo­lezni 9,3 mesecev. Najpogostejši nehematološki neželeni učinki so bili periferna senzorična nevropatija, slabost in utrujenost, najpogostejši resni neželeni učinki pa nevtropenija, trombocitopenija, anemija in periferna senzorična nevropatija. Radiol Oncol 2015; 49(4): 320-326. doi:10.1515/raon-2015-0043 Parametri preiskave s [F-18] FDG-PET/CT kot napovedni dejavniki zdravljenja bolnikov z neoperabilnim nedrobnoceličnim rakom pljuč Nappi A, Gallicchio R, Simeon V, Nardelli A, Pelagalli A, Zupa A, Vita G, Venetucci A, Di Cosola M, Barbato F, Storto G Izhodišča. V raziskavi smo ocenili napovedni pomen standardizirane vrednosti privzema (SUVmax), presnovni volumen tumorja in skupno glikolizo v leziji pri [F-18] FDG-PET/CT preiskavi bolnikov z neoperabilnim, nedrobnoceličnim pljučnim rakom. Bolniki in metode. Pri 103 bolnikih (povprečna starost 65,6 ± 16 let) smo naredili preiskavo [F-18] FDG-PET/CT pred kemo­terapijo. Zabeležili smo vrednost SUVmax, presnovni volumen tumorja (cm3; prag 42 %) in skupno glikolizo v leziji. Bolnike smo spremljali do 18 mesecev (razpon 12–55 mesecev). Neodzivnost na zdravljenje brez napredovanja bolezni ter napredovanje bolezni in/ali smrt povezana z boleznijo so pomenili nadomestni končni cilj. Ugotavljali smo, kakšen je optimalni SUVmax, pre­snovni volumen tumorja in mejna vrednost za skupno glikolizo v leziji, ki bi lahko napovedali izid zdravljenja bolnikov. Rezultate PET/CT smo nato povezali z izidom bolezni (preživetjem brez napredovanja bolezni) Rezultati. Analiza preživetja po metodi Kaplan-Meier za SUVmax je pokazala znatno krajši čas preživetja brez napredova­nja bolezni pri bolnikih, ki so imeli nižje vrednosti v primerjavi s tistimi z višjimi (p <0,05, log-rank test). Presnovni volumen tumorja in skupna glikoliza v leziji nista bila primerna za napovedovanje preživetja brez napredovanja bolezni, razen za podskupino bolnikov s tumorsko zajetimi mediastinalnimi bezgavkami. Zaključki. Kljub razpoložljivosti novih orodij za kvantitativno oceno aktivnosti bolezni na PET/CT, ostaja SUVmax, ne pa tudi presnovni volumen tumorja in skupna glikoliza v leziji, edini napovedni dejavnik za preživetje brez napredovanja bolezni pri bolnikih z nedrobnoceličnim rakom pljuč. Presnovni volumen tumorja ima vrednost le, če se pojavi sočasna tumorska zajetost bezgavk. Radiol Oncol 2015; 49(4): 327-333. doi:10.1515/raon-2015-0016 Optimalni čas slikanja za ovrednotenje obščitničnih adenomov z [18F]-fluoroholin PET/CT Rep S, Lezaić L, Kocjan T, Pfeifer M, Jensterle Sever M, Simončič U, Tomše P, Hočevar M Izhodišča. Najpogostejši vzrok za primarni hiperparatiroidizem je adenom obščitnice. Adenomi obščitnic so benigni tumorji, ki proizvajajo in izločajo obščitnični hormon. [18F]-fluoroholin, je pozitronski emisijski tomografski (PET) označevalec celične proliferacije. Nedavno so ugotovili, da se [18F]-fluoroholin kopiči v povečanem obščitničnem tkivu, vendar optimalni čas za slikanje z [18F]-fluoroholin PET/CT po aplikaciji [18F]-fluoroholina ni znan. Cilj raziskave je bil ugotoviti optimalni čas za izvedbo preiskave [18F]-fluoroholin PET/CT pri bolnikih s primarnim hiperparatiroidizmom. Bolniki in metode. V raziskavo smo vključili 43 bolnikov s primarnim hiperparatiroidizmom. Tro-fazno slikanje PET/CT smo izvedli pet minut, eno uro in dve uri po aplikaciji [18F]-fluoroholina. Interesna območja (ROI) smo izbrali v povečanem obščitnič­nem in ščitničnem tkivu. Standardizirano vrednost privzema (SUVmean), retencijski indeks (RI) in kontrast lezij (LC) smo računali za povečano obščitnično in ščitnično tkivo. Rezultati. Kopičenje [18F]-fluoroholina je bilo statistično pomembno višje v povečanem obščitničnem tkivu v primerjavi s ščitničnim tkivom, kjer se je pokazal precej višji SUVmean v drugi in v tretji fazi (p < 0,0001). Povprečna vrednost RI se je pomemb­no zmanjšala med prvo in drugo fazo, pomembno povečala med drugo in tretjo fazo v povečanem obščitničnem tkivu ter značilno zmanjšala v vseh fazah v ščitničnem tkivu (p < 0,0001). LC med povečanim obščitničnim tkivom in ščitničnim tkivom je bil bistveno boljši v drugi in tretji fazi (p < 0,05). Zaključek. Glede na naše rezultate je optimalni čas slikanja [18F]-fluoroholin PET/CT za lokalizacijo povečanega obščitnič­nega tkiva eno uro po aplikaciji [18F]-fluoroholina. Radiol Oncol 2015; 49(4): 334-340. doi:10.1515/raon-2015-0020 Vloga razmerja deformabilnosti, izmerjenega z ultrazvočno endobronhialno elastografijo v diagnostiki mediastinalnih bezgavk Rozman A, Marc Malovrh M, Adamič K, Šubic T, Kovač V, Fležar M Izhodišča. Ultrazvočna elastografija je slikovna metoda za ocenjevanje biomehaničnih lastnosti različnih tkiv. Namen razi­skave je bil ugotoviti diagnostično vrednost razmerja deformabilnosti, izmerjenega z endobronhialno ultrazvočno elastografijo na nivoju mediastinalnih bezgavk pri bolnikih s sumom na pljučni rak. Diagnostične vrednosti razmerja deformabilnosti smo primerjali z diagnostičnimi vrednostmi pri pregledu bezgavk z endobronhialnim ultrazvokom v svetlostnem načinu in s citološko diagnozo. Bolniki in metode. V prospektivno klinično raziskavo, ki smo jo naredili samo v eni ustanovi, smo vključevali bolnike s sumom na pljučni rak in indikacijo za biopsijo. Pri bolnikih smo želeli ugotoviti zamejitev bolezni na nivoju mediastinuma, potem ko smo opravili neinvazivne slikovne preiskave. Pred punkcijo bezgavk pod nadzorom endobronhialnega ultrazvoka smo naredili oceno izbranih bezgavk v svetlostnem načinu in v elastografskem načinu z merjenjem razmerja deformabilnosti. Rezultati. Vključili smo triintrideset bolnikov z 80-imi sumljivimi mediastinalnimi bezgavkami. Maligno infiltracijo smo potrdili v 34 (42,5 %) bezgavkah. Površina pod krivuljo karakteristik sprejemnika za razmerje deformabilnosti je bila 0,87 (p < 0,0001). Pri razmerju deformabilnosti . 8 je bila ocena natančnosti za maligno infiltracijo 86,25 % (občutljivost 88,24 %, specifičnost 84,78 %, pozitivna napovedna vrednost 81,08 %, negativna napovedna vrednost 90,70 %). Razmerje deformabilnosti je bilo natančnejše, kot ocena s konvencionalnim svetlostnim načinom endobronhialnega ultrazvoka pri ločevanju maligno infiltri­ranih in benignih mediastinalnih bezgavk. Zaključki. Ocena razmerja deformabilnosti z endobronhialno ultrazvočno elastografijo lahko loči med benignimi in ma­ligno infiltriranimi mediastinalnimi bezgavkami z večjo natančnostjo, kot konvencionalni endobronhialni ultrazvočni pregled bezgavk v svetlostnem načinu. S to novo metodo bi lahko znižali število ultrazvočno vodenih punkcij, vzporedno pa tudi invazivnost in stroške ugotavljanja zamejitve nedrobnoceličnega raka pljuč na nivoju mediastinuma. Radiol Oncol 2015; 49(4): 341-346. doi:10.1515/raon-2015-0044 Analiza dejavnikov tveganja za edem možganovine po znotrajžilni embolizaciji nerupturiranih možganskih arterijskih anevrizem Lukić S, Janković S, Šurlan Popović K, Banković D, Popovič P, Mijailović M Izhodišča. Znotrajžilno zdravljenje nerupturiranih možganskih anevrizem je zdravljenje izbora, katerega zaplet je lahko edem možganovine ob anevrizmi. V raziskavi smo želeli ugotoviti dejavnike tveganja za razvoj edema možganovine ob anevrizmi, zdravljeni z znotrajžilnim zapiranjem, njegovo pogostnost in kliničen izhod. Metode. V raziskavo smo vključili 119 odraslih bolnikov, ki so imeli znotrajžilno zdravljenje nerupturirane anevrizme na Oddelku za intervencijsko nevroradiologijo v Kliničnem centru Kragujevac, Srbija. Za zapiranje anevrizem smo uporabili spirale, ki ima­jo elektrolitsko nadzorovano sprostitev: spirale iz čiste platine, hidrofilne spirale in kombinirane platinasto-hidrofilne spirale. Z magnetno resonančno preiskavo (MRI) smo ugotavljali razvoj edema možganovine ob zdravljeni anevrizmi 7, 30 in 90 dni po posegu. Rezultati. Edem smo ugotovili pri 47,6 % bolnikih zdravljenih s hidrofilnimi spiralami, 21,6 % bolnikih zdravljenih s spiralami iz platine in 53,8 % bolnikih zdravljenih s kombiniranimi spiralami. Z multivariatno logistično regresijo smo dokazali povezavo med edemom možganovine ter prostornino anevrizme, zvišanim arterijskim tlakom, sladkorno boleznijo in kajenjem. Zvišan arterijski tlak je najpomembnejši neodvisen napovedni dejavnik za razvoj edema možganovine ob zdravljeni anevrizmi, ki mu sledijo kajenje in sladkorna bolezen. Zaključki. Rezultati raziskave so pokazali, da imajo največje tveganje za razvoj edema možganovine ob anevrizmi, zdra­vljeni z znotrajžilnim zapiranjem, starejši bolniki z velikimi, nerupturiranimi anevrizmami, ki so kadilci, imajo sladkorno bolezen in zvišan arterijski tlak. Radiol Oncol 2015; 49(4): 347-356. doi:10.1515/raon-2015-0040 Ocena preživetja celic pri frakcionirani radioterapiji z uporabo različnih metod izvedenih iz linerno­kvadratnega modela Nikzad S, Hashemi B, Mahmoudi G, Baradaran-Ghahfarokhi M Izhodišča. Namen raziskave je bil uporabiti različne teoretične metode za oceno preživetja celic. Metode smo izvedli iz linearno-kvardartnega modela (LQ). Želeli smo ugotoviti vpliv števila frakcij obsevanja, časovnega intervala med njimi, doze na frakcijo in celotnega časa obsevanja. Načrtovali smo primerjavo rezultatov, temelječih na in vitro preživetju melanomskih in adenokarcinomskih celic. Glede na rezultate raziskave smo želeli predlagati najbolj primerno metodo izračuna, ki ustreza in vitro rezultatom preživetja celic. Materiali in metode. Proučili smo najpogosteje uporabljene teoretične modele iz literature avtorjev Keall in sod. in Mu in sod. za izračun preživetja celic po radioterapiji pri različnih režimih obsevanja. Skupni čas obsevanja je bil med 15 in 240 minutami. Za izračun potrebnega števila frakcij obsevanja in doze na frakcijo smo izbrali čas 30, 60 in 240 minut. Poskuse in vitro smo naredili pri obsevalni dozi 4 Gy in rezultate primerjali s teoretičnimi izračuni. Rezultati. Najpomembnejši parameter za preživetje celic je bil skupni čas obsevanja. V teoretičnih modelih je bilo po­membno tudi število frakcij obsevanja, kar nismo statistično značilno potrdili v eksperimentalnih modelih. Spreminjanje števila frakcij pa je statistično značilno pokazalo različne rezultate po izračunih Keall in sod. in Mu in sod. (p < 0.05). Zaključki. Metoda Mu in sod. najbolje napove preživetje celic po frakcionirani radioterapiji. Rezultati nakazujejo na primer­nost te metode za napoved odgovora celic na frakcionirano radioterapijo in njeno uporabo tudi v kliniče namene. Radiol Oncol 2015; 49(4): 357-364. doi:10.2478/raon-2014-0049 Urokinazni aktivator plazminogena (uPA) in inhibitor aktivatorja plazminogena 1 (PAI-1) pri primarnem raku dojk. Povezanost s klasičnimi napovednimi dejavniki Lampelj M, Arko D, Čas-Sikošek N, Kavalar R, Ravnik M, Jezeršek-Novaković B, Dobnik S, Fokter Dovnik N, Takač I Izhodišča. Urokinazni aktivator plazminogena (uPA) in inhibitor aktivatorja plazminogena 1 (PAI-1) imata ključno vlogo pri invaziji in zasevanju malignih tumorjev. Visoke vrednosti obeh proteolitičnih encimov so povezane s slabo napovedjo poteka bolezni pri bolnicah z rakom dojk. Namen raziskave je bil preučiti povezanost med klasičnimi napovednimi dejavniki ter uPA in PAI-1 v tkivu primarnega tumorja bolnic z rakom dojk. Metode. V prospektivno klinično raziskavo na Oddelku za ginekološko onkologijo in onkologijo dojk Univerzitetnega klinične­ga centra Maribor smo med leti 2004 in 2010 vključili 606 bolnic s primarnim rakom dojk. Ovrednotili smo klasične napovedne dejavnike (starost, menopavzni status, velikost tumorja, patohistološki tip tumorja, stopnjo malignosti tumorja, prizadetost paz­dušnih bezgavk, invazijo v krvne in limfne žile in stanje hormonskih receptorjev) ter vrednosti uPA in PAI-1. Dobljene podatke smo obdelali s Spearmanovo korelacijo rangov, Mann-Whitneyevim U testom in testom .2. Rezultati. Izsledki raziskave nakazujejo pozitivno povezavo med uPA in velikostjo tumorja (p < 0,001), stopnjo malignosti tumorja (p < 0,001), patohistološkim tipom tumorja (p < 0,001), invazijo v krvne in limfne žile (p = 0,01) in negativno povezavo s stanjem hormonskih receptorjev (p < 0,001). Prav tako nakazujejo pozitivno povezavo med PAI-1 in velikostjo tumorja (p = 0,004), stopnjo malignosti tumorja (p < 0,001), patohistološkim tipom tumorja (p < 0,001) in negativno povezavo s stanjem hormonskih receptorjev (p = 0,002). Zaključki. Raziskava je potrdila povezanost med uPA in PAI-1 ter klasičnimi napovednimi dejavniki. Njuno vlogo kot dejav­nikov, ki napovedujejo potek bolezni, in kot dejavnikov, ki napovedujejo odgovor na zdravljenje pri bolnicah z rakom dojke, je potrebno nadalje ovrednotiti. RadiolOncol 2015; 49(4): 365-370. doi:10.2478/raon-2014-0048 Napovedna vrednost nekaterih tumorskih označevalcev pri bolnikih z napredovalim neoperabilnim rakom ustnega žrela, zdravljenih s sočasno radiokemoterapijo Šoba E, Budihna M, Šmid L, Gale N, Lešničar H, Zakotnik B, Strojan P Izhodišča. Cilj raziskave je bil proučiti, kako tumorski označevalci p21, p27, p53, ciklin D1, receptorji epidermalnega rastnega dejavnika (EGFR), Ki-67 in CD31 napovedujejo izid bolezni pri bolnikih z napredovalim neoperabilnim rakom ustnega žrela, zdravljenih s sočasno kombinacijo obsevanja in kemoterapije. Bolniki in metode. Tkivo biopsijskih vzorcev 74 zaporedno obsevanih bolnikov z neoperabilnim rakom ustnega žrela, odvzetih pred pričetkom zdravljenja, smo v retrospektivni klinični raziskavi histokemično obarvali za prikaz tumorskih označe­valcev p21, p27, p53, ciklin D1, EGFR, Ki-67 in CD31. Ocenjevali smo, kako je izraženost tumorskih označevalcev povezana s preživetjem brez bolezni. Rezultati. Pri bolnikih z visoko izraženostjo p21 (. 10 %), p27 (> 50 %), Ki-67 (> 50 %) in CD31 (> 130 žil/mm2) in nizko izraženo­stjo p53 (< 10 %), ciklin D1 (< 10 %) in EGFR (< 10 %) (ugodni nivoji) je bilo preživetje brez bolezni boljše kot pri bolnikih z nizko izraženostjo p21 (< 10 %), p27 (. 50 %), Ki-67 (. 50 %) in CD31 (<130 žil/mm2) ter visoko izraženostjo p53 (. 10 %), ciklin D1 (. 10 %) in EGFR (. 10 %) (neugodni nivoji), a je bila statistična značilna razlika v preživetju med ugodnim in neugodnim nivojem potrjena le za p27 in ciklin D1. Preživetje brez bolezni se je statistično značilno slabšalo z naraščanjem števila označevalcev z neugodnim nivojem v tumorjih (1–4 vs. 5–7) (78 % vs. 32 %; p = 0,004). Število označevalcev z neugodnim nivojem v tumorju je ohranilo napovedno značilnost tudi v multivariatni analizi. Zaključki. Statistično značilna razlika v preživetju med bolniki z ugodnim in neugodnim nivojem tumorskih označevalcev je bila potrjena le za p27 in ciklin D1. Število označevalcev v tumorju z neugodnim nivojem izraženosti je bilo neodvisen napo­vedni dejavnik za slab izid bolezni. Radiol Oncol 2015; 49(4): 371-378. doi:10.1515/raon-2015-0034 Protonsko obsevanje dvajsetih starejših bolnikov z rakom požiralnika Ono T, Nakamura T, Azami Y, Yamaguchi H, Hayashi Y, Suzuki M, Hatayama Y, Tsukiyama I, Hareyama M, Kikuchi Y, Nemoto K Izhodišča. V starajoči se družbi je vedno več starejših bolnikov z ugotovljenim rakom požiralnika. Namen raziskave je bil oceniti klinično učinkovitost in varnost obsevanja s protoni pri starejših bolnikih z rakom požiralnika. Bolniki in metode. V raziskavo smo vključili starostnike (starost . 65 let) z rakom požiralnika, ugotovljenim med januarjem 2009 in junijem 2013. Vsi bolniki so bili napoteni na zdravljenje bodisi samo s protonskimi žarki bodisi s protonskimi žarki in iz­hodiščnim obsevanem z žarki x. Neželene učinke smo ocenjevali z lestvico neželenih učinkov zdravljenja po enotnih kriterijih (angl. Common Terminology Criteria for Adverse Events, CTCAE), verzija 4,0, ki sta jo izdelala ameriški Nacionalni inštitut za zdravljenje raka in ameriški Nacionalni inštitut za zdravje. Rezultati. Za raziskavo je bilo primernih 20 bolnikov in pri vseh smo lahko zaključili zdravljenje. Srednja starost je bila 78 let (razpon: 65–89 let) in srednji čas njihovega spremljanja 25,5 mesecev (razpon 6–62 mesecev). Sedem bolnikov je imelo zasev­ke v bezgavkah in 10 bolnikov je imelo bolezen II. ali III. stadija. Pri obsevanju samo s protoni je bila srednja doza obsevanja s protoni 72,6 Gy relativne biološke dozne učinkovitosti (angl. ralative biological dose effectiveness, RBE) (razpon 66–74,8 Gy [RBE]), pri zdravljenju s protoni in izhodiščnem obsevanju z žarki x pa je bila doza s protoni 33 Gy (RBE) (razpon 30,8–39,6 Gy [RBE] ter skupni razpon s protoni in žarki x 66,8–75,6 Gy [RBE]). Dveletno celokupno preživetje je bilo 81,8 % (95% interval zau­panja [CI]: 62,4 % – 100 %) in dveletna lokalna kontrola bolezni je bila 89,4 % (95 % CI: 75,5 % – 100 %). Stranski učinki 2. ali 3. stopnje so se pojavili pri nekaj primerih, ni pa bilo ugotovljene toksičnosti 4. ali 5. stopnje. Zaključki. Visokodozno obsevanje s protoni (66–75,5 Gy [RBE]) brez kemoterapije je bilo učinkovito in varno zdravljenje pri starejših bolnikih z rakom požiralnika. Radiol Oncol 2015; 49(4): 379-385. doi:10.1515/raon-2015-0039 Pogostnost kožnega melanoma in njegova odvisnost od letnega časa. 20-letno obdobje opazovanj v populaciji z prekomernim sončenjem Bonin S, Albano A, di Meo N, Gatti A, Stinco G, Zanconati F, Trevisan G Izhodišča. Kožni melanom je zelo agresivna oblika raka, ki je postal vse pogostejši v večini razvitih držav. Posebno številen je pri Evropejcih. Bolniki in metode. Anonimne podatke bolnikov s kožnim melanomom smo zajeli iz diagnostične baze podatkov Univerzitetne bolnice Trst za obdobje med 1. januarjem 1990 in 10. decembrom 2013. Raziskava obravnava dobro definirano populacijo s karakteristično lastnostjo prekomernega sončenja. Rezultati. Incidenca kožnega melanoma je v opazovanem obdobju stalno rastla s trendom odvisnosti od letnega časa. Ugotovili smo razlike med spoloma glede anatomske lokacije in stadija bolezni. Poleg tega je 6 % bolnikov razvilo multiple melanome. Zaključki. V dobro definirani populaciji z navado prekomernega sončenja se je incidenca kožnega melanoma podvojila v obdobju opazovanj od 1990 do 2013. Zato tej populaciji priporočamo preventivne ukrepe glede na navade, ki so odvisne od spola. Priporočamo tudi programe sledenja bolezni za ponovitev bolezni in za zgodnje odkrivanje dodatnih primarnih tumorjev, posebno glede na razlike v pojavnosti pri spolih in pojavnosti multipllih melanomov. Radiol Oncol 2015; 49(4): 386-394. doi:10.1515/raon-2015-0002 Sproščanje rastnih dejavnikov po mehanski in kemični plevrodezi pri zdravljenju malignih plevralnih izlivov. Prospektivna randomizirana raziskava primerljivih primerov Hojski A, Leitgeb M, Crnjac A Izhodišča. Rastni dejavniki so ključni spodbujevalci fibroze, obenem pa sodelujejo v vnetnih odzivih telesa, ki lahko po­večajo volumen plevralnega izliva in sprožijo sindrom akutne dihalne stiske. Primarni cilj raziskave je bil primerjava kemične plevrodeze s smukcem in torakoskopske mehanske plevrodeze glede na sproščanje rastnih dejavnikov v prvih 48 urah. Sekundarni cilji raziskave pa so bili ocena učinkovitosti obeh načinov plevrodeze, ugotavljanje lajšanja simptomov in kvalitete življenja po posegu. Bolniki in metode. V prospektivno randomizirano raziskavo smo vključili 36 zaporednih bolnic z rakom dojke in doka­zanim malignim plevralnim izlivom, ki so bile napotene na naš oddelek zaradi zdravljenja. 18 bolnic smo zdravili s kemijsko plevrodezo in 18 bolnic z mehansko plevrodezo. Pri kemijski plevrodezi smo uporabili 5g emulzije smukca, ki smo ga vbrizgali preko torakalnega drena. Po pripravljenem protokolu smo v obdobju 48 ur zbirali vzorce plevralne tekočine in seruma ter v njih ugotavljali rastne dejavnike. Z analogno vizualno skalo smo ocenjevali bolečino, s standardiziranim vprašalnikom pa kvaliteto življenja. Rezultati. Srednji izmerjeni serumski nivo rastnega dejavnika žilnega endotela (VEGF) po kemični plevrodezi je znašal 930.68 pg/ml (95% interval zaupanja CI: 388.22–4656.65), po mehanski pa 808.54 pg/ml. (95% CI: 463.20-1235.13) (p = 0.103). Nasprotno so bile srednje vrednosti transformirajočega rastnega dejavnika (TGF) ß1 v plevralni tekočini višje po mehanski plevrodezi (4814.00 pg/ml [95% CI: 2726.51–7292.94]) v primerjavi s tistimi po kemijski plevrodezi (1976.50 pg/ml [95% CI: 1659.82–5136.26]) (p = 0.078). Podobne rezultate smo zabeležili za fibroblastni rastni dejavnik (FGF) ß; serumski nivo je bil višji po mehanski plevrodezi (30.45 pg/ml [95% CI: 20.40–59.42]) kot po kemijski (13,39 pg/ml (13.39 pg/ml [95% CI: 5.04 – 74.60]) (p = 0.076). Bolnice po kemijski in mehanski plevrodezi so imele primerljivo trajanje hospitalizacije, pogostost ponovnega ple­vralnega izliva in ponovnih torakalnih drenaž ter celokupno preživetje; nasprotno pa so bolnice po mehanski plevrodezi imele krajši čas drenaže po posegu (p = 0.030), manj bolečin in boljšo kakovost življenja (p = 0,047). Zaključki. Primerjava zdravljenja z različno plevrodezo je pokazala povečanje serumskih vrednosti VEGF pri bolnicah po kemijski plevrodezi. Nasprotno so imele bolnice po mehanski plevrodezi povišane vrednosti TGF ß1 in FGF ß v plevralni tekočini. Razlike niso dosegle statistične značilnosti; vendar ostajajo VEGF, TGFß1 in FGF ß najbolj zanimivi parametri za nadaljnje razi­skave. Ob upoštevanju mehanizmov delovanja rastnih dejavnikov se je v naši raziskavi mehanska plevrodeza nakazovala kot potencialno učinkovitejša, s krajšim časom plevralne drenaže, izrazitejšem zmanjšanjem bolečine in boljšo kvaliteto življenja. Radiol Oncol 2015; 49(4): 395-401. doi:10.2478/raon-2014-0036 Gigantski solitarni fibrozni tumor plevre. Prikaz primera in pregled literature Crnjac A, Veingerl B, Vidovič D, Kavalar R, Hojski A Izhodišča. Mezenhimski solitarni tumorji plevre so redki. Večinoma so benigni, okoli 12 % je malignih. V začetni fazi so naj­večkrat asimptomatski, z rastjo tumorja pa zaradi pritiska na okoliške strukture povzročajo bolečine v prsih, dražeč kašelj in oteženo dihanje. Redko povzročajo simptome pritiska na mediastinalne strukture. Potrebna je stopenjska radiološka diagnosti­ka. Predoperativna biosija je v večini primerov neuspešna. Terapija izbora tovrstne patologije je radikalna kirurška odstranitev tumorja, ki jo je - pri malignih tumorjih ali neradikalno odstranjenih benignih tumorjih - potrebno dopolniti z adjuvantno terapijo. Prikaz primera. Opisujemo primer 68-letne bolnice, ki smo je sprejeli na Klinični oddelek za torakalno kirurgijo UKC Maribor zaradi gigantskega fibroznega tumorja v desnem plevralnem prostoru. Tumor je s svojo ekspanzivno rastjo povzročal premik mediastinuma in utesnjeval spodnjo votlo veno, desni predvor ter srednji in spodnji pljučni reženj. Zaradi vtočnih motenj in kolapsa desnih pljuč je bila bolnica življenjsko ogrožena z znaki kardiorespiratorne odpovedi. Po predoperativni radiološki diagnostiki smo bolnici kirurško radikalno odstranili tumor. Pooperativno se je bolnici zdravstveno stanje izboljšalo. Po letu dni nismo zasledili ponovitve bolezni. Zaključki. Veliki fibrozni tumorji plevre lahko zaradi omejenosti plevralnega prostora povzročajo s svojo ekspanzivno rastjo resna, življenje ugrožujoča stanja. Pravočasna prepoznava bolezni omogoča izvedbo manj agresivnega, lahko tudi videoto­rakoskopskega posega pri bolnikih v bistveno boljšem zdravstvenem stanju. Operacije velikih tumorjev pri kardiorespiratorno prizadetih bolnikih so sicer edini možni, a nadvse tvegan kirurški poseg. Radiol Oncol 2015; 49(4): 402-408. doi:10.1515/raon-2015-0038 Vpliv spremljajočih bolezni na izhod bolezni pri bolnikih z napredovalim rakom prostate, zdravljenimi z docetakselom Žist A, Amir E, Ocana AF, Šeruga B Izhodišča. Moškim z metastatskim, proti kastraciji odpornim rakom prostate lahko zdravljenje z docetakselom v vsakodnevni klinični praksi neupravičeno odtegnemo zaradi pridruženih bolezni. Zato smo v raziskavi preučevali vpliv pridruženih bolezni na izid zdravljenja z docetakselom pri bolnikih s takšnim rakom prostate. Metode. V raziskavo smo vključili vse bolnike z metastatskim, proti kastraciji odpornim rakom prostate, ki smo jih med letoma 2005 in 2012 zdravili z docetakselom na Onkološkem inštitutu Ljubljana. Stopnjo pridruženih bolezni smo ovrednotili s starostno prilagojenim Charlsonovim indeksom pridruženih bolezni (angl. age-adjusted Charlson comorbidity index, aa-CCI) in z inde­ksom ocenjevanja 27 pridruženih bolezni odraslih (angl. adult comorbidity evaluation-27, ACE-27). Kako bolniki prenašajo zdravljenje, smo ocenili s številom hospitalizacij in smrti, ki so nastopile zaradi toksičnih sopojavov med zdravljenjem. Povezavo med pridruženimi boleznimi in skupnim preživetjem smo testirali s Cox-ovo analizo. Rezultati. Z docetakselom smo zdravili 208 moških. Brez pridruženih bolezni ali z blago, zmerno in hudo stopnjo je bilo 2 %, 32 %, 53 % in 13 % bolnikov, če smo jih ocenjevači z aa-CCI ter 27 %, 35 %, 29 % in 8 %, če smo jih ocenjevali z ACE-27. Pomembno zmanjšanje odmerka docetaksela je bilo pogostejše pri bolnikih z zmerno ali hudo stopnjo pridruženih bolezni v primerjavi s tistimi brez ali z blago stopnjo pridruženih bolezni. Ne glede na pridružene bolezni je približno ena tretjina moških potrebovala hospitalizacijo ali je umrla med zdravljenjem z docetakselom. V univariantni analizi višja stopnja pridruženih bolezni ni bila po­vezana s slabšim skupnim preživetjem (aa-CCI: razmerje obetov [HR] 0,99; 95 % interval zaupanja [CI] 0,87–1,13; p = 0,93 ter ACE-27: HR 0,96; 95 % CI 0,79–1,17; p = 0,69). Zaključki. Moškim z metastatskim, proti kastraciji odpornim rakom prostate ter pridruženimi boleznimi lahko koristimo, če jih zdravimo z docetakselom. Radiol Oncol 2015; 49(4): 409-415. doi:10.1515/raon-2015-0037 Klinični pomen preživetja po napredovanju bolezni po primarni kemoradioterapiji na celokupno preživetje pri bolnikih z omejeno obliko drobnoceličnega pljučnega raka Kasahara N, Imai H, Kaira K, Mori K, Wakuda K, Ono A, Taira T, Kenmotsu H, Harada H, Naito T, Murakami H, Endo M, Nakajima T, Yamada M, Takahashi T Izhodišča. Pri bolnikih z omejeno obliko drobnoceličnega pljučnega raka lahko učinke primarnega zdravljenja s kemora­dioterapije prekrijemo s sekundarnim zdravljenjem. To lahko namreč znatno vpliva na celokupno preživetje. Zato smo želeli ugotoviti povezavo med preživetjem brez napredovanja bolezni oz. preživetjem po napredovanju bolezni in celokupnim preživetjem po primarnem zdravljenju s kemoradioterapijo. Bolniki in metode. Retrospektivno smo analizirali 71 bolnikov z omejeno obliko drobnoceličnega pljučnega raka in s splo­šnim stanjem zmogljivosti 0–2. Prejeli so primarno kemoradioterapijo v Shizuoka Cancer Center (Shizuoka, Japonska) in imeli ponovitev bolezni med septembrom 2002 in marcem 2013. Ugotavljali smo korelacijo med preživetjem brez napredovanja bolezni in celokupnim preživetjem ter med preživetjem po napredovanju bolezni in celokupnim preživetjem na individualni ravni. Poleg tega smo izvedli univariatne in multivariatne analize za prepoznavanje pomembnih napovednih dejavnikov preživetja po napredovanju bolezni. Rezultati. Celokupno preživetje je bolj povezano s preživetjem po napredovanju bolezni (Spearman r = 0,86, R2 = 0,72, p < 0,05) kot s preživetjem brez napredovanja bolezni (Spearman r = 0,46, R2 = 0,38, p < 0,05). Poleg tega smo odkrili, da so pomembni neodvisni napovedni dejavniki za preživetje po napredovanju bolezni odgovor na zdravljenje po sekundarnem zdravljenju, prisotnost oddaljenih metastaz ob ponovitvi bolezni in število dodatnih zdravljenj po primarni kemoradioterapiji. Zaključki. Preživetje po napredovanju bolezni ima večji vpliv na celokupno preživetje kot pa preživetje brez napredovanja bolezni pri bolnikih z omejeno obliko drobnoceličnega pljučnega raka, z dobrim splošnim stanjem zmogljivosti na začetku zdravljenja in s ponovitvijo bolezni. Poleg tega lahko zdravljenje po primarni kemoradioterapije vpliva na njihovo celokupno preživetje. Vendar pa so potrebne večje multicentrične raziskave za potrditev te ugotovitve. Fundacija "Docent dr. J. Cholewa" je neprofitno, neinstitucionalno in nestrankarsko združenje posameznikov, ustanov in organizacij, ki želijo materialno spodbujati in poglabljati raziskovalno dejavnost v onkologiji. Dunajska 106 1000 Ljubljana IBAN: SI56 0203 3001 7879 431 Activity of "Dr. J. Cholewa" Foundation for Cancer Research and Education – a report for the final quarter of 2015 The “Docent Dr. J. Cholewa Foundation for Cancer Research and Education” is named after Dr. Josip Cholewa, one of the first researchers in cancer in Slovenia and the founder of the “Banovinski Inštitut za raziskovanje in zdravljenje novotvorb” in 1937, that later became the Institute of Oncology in Ljubljana, Slovenia. His laboratory and clinical research work was based on an innovative and far-reaching multidis­ciplinary approach that included studies on prevention, detection and treatment of cancer. This pioneering approach facilitated the understanding of the complexities of all the problems and troubles experienced by cancer patients, their doctors and other medical staff when facing this disease. It could also be regarded as a harbinger of the progress observed in a large part of the world in the last half of the previous century. Therefore, the Foundation is a non-profit, non-political and non-government organisation that helps pro­fessionals, institutions and individuals obtaining financial help for cancer research and education in the Republic of Slovenia with the goal of continuing and expanding the great work and efforts of Dr. Josip Cholewa. The “Docent Dr. J. Cholewa Foundation for Cancer Research and Education” hopes and strives to provide at least part of the financial support needed by qualified individuals and organisations interested in cancer research in the Republic of Slovenia. One of the objectives of the Foundation is to facilitate the transmission of the latest diagnostic and therapy procedures to the clinical environment in Slovenia, thus benefiting the ever increasing number of patients with various types of cancer in Slovenia. With this in mind, it is impor­tant to note that the incidence rates of many cancer, like colon, prostate and breast cancer have kept rising in recent decades in Slovenia. The Foundation continues to provide financial support to "Radiology and Oncology”, an international sci­entific journal that is edited and published in Ljubljana, Slovenia. It publishes scientific research articles, reviews, and letters to the editor about research and studies in experimental and clinical oncology, sup­portive therapy, radiology, radiophyics, prevention and early diagnostics of different types of cancer. It is an open access journal freely available in pdf format and with a respectable Science Citation Index Impact factor. All the abstracts in “Radiology and Oncology” are available in Slovenian and the journal can thus provide sufficient scientific information from various fields of high quality cancer research to interested lay public in Slovenia. The “Docent Dr. J. Cholewa Foundation for Cancer Research and Education” has thus an important role in support of cancer research, cancer education and many of the related fields in the Republic of Slovenia. Borut Štabuc, M.D., Ph.D. Tomaž Benulič, M.D. Viljem Kovač, M.D., Ph.D. Andrej Plesničar, M.D., M.Sc. Kakovostna in količinska sestava 1 ml raztopine vsebuje 1,5 mg benzidaminijevega klorida, kar ustreza 1,34 mg benzidamina. V enem razpršku je 0,17 ml raztopine. En razpršek vsebuje 0,255 mg benzidaminijevega klorida, kar ustreza 0,2278 mg benzidamina. En razpršek vsebuje 13,6 mg 96 odstotnega etanola, kar ustreza 12,728 mg 100 odstotnega etanola, in 0,17 mg metilparahidroksibenzoata (E218). Terapevtske indikacije Samozdravljenje: lajšanje bolečine in oteklin pri vnetju v ustni votlini in žrelu, ki so lahko posledica okužb in stanj po operaciji. Po nasvetu in navodilu zdravnika: lajšanje bolečine in oteklin v ustni votlini in žrelu, ki so posledica radiomukozitisa. Odmerjanje in način uporabe Uporaba 2- do 6-krat na dan (vsake 1,5 do 3 ure). Odrasli: 4 do 8 razprškov 2- do 6-krat na dan. Otroci od 6 do 12 let: 4 razprški 2- do 6-krat na dan. Otroci, mlajši od 6 let: 1 razpršek na 4 kg telesne mase; do največ 4 razprške 2 do 6-krat na dan. Kontraindikacije Znana preobčutljivost za zdravilno učinkovino ali katerokoli pomožno snov. Posebna opozorila in previdnostni ukrepi Pri manjšini bolnikov lahko resne bolezni povzročijo ustne/žrelne ulceracije. Če se simptomi v treh dneh ne izboljšajo, se mora bolnik posvetovati z zdravnikom ali zobozdravnikom, kot je primerno. Zdravilo vsebuje aspartam (E951) (vir fenilalanina), ki je lahko škodljiv za bolnike s fenilketonurijo. Zdravilo vsebuje izomalt (E953) (sinonim: izomaltitol (E953)). Bolniki z redko dedno intoleranco za fruktozo ne smejo jemati tega zdravila. Uporaba benzidamina ni priporočljiva za bolnike s preobčutljivostjo za salicilno kislino ali druga nesteroidna protivnetna zdravila. Pri bolnikih, ki imajo ali so imeli bronhialno astmo, lahko pride do bronhospazma. Pri takih bolnikih je potrebna previdnost. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij Pri ljudeh raziskav o interakcijah niso opravljali. Nosečnost in dojenje Tantum Verde z okusom mentola 3 mg pastile se med nosečnostjo in dojenjem ne smejo uporabljati. Vpliv na sposobnost vožnje in upravljanja s stroji Uporaba benzidamina lokalno v priporočenem odmerku ne vpliva na sposobnost vožnje in upravljanja s stroji. Neželeni učinki Bolezni prebavil Redki: pekoč občutek v ustih, suha usta. Bolezni imunskega sistema Redki: preobčutljivostna reakcija. Bolezni dihal, prsnega koša in mediastinalnega prostora Zelo redki: laringospazem. Bolezni kože in podkožja Občasni: fotosenzitivnost. Zelo redki: angioedem. Rok uporabnosti 4 leta. Zdravila ne smete uporabljati po datumu izteka roka uporabnosti, ki je naveden na ovojnini. Posebna navodila za shranjevanje Za shranjevanje pastil niso potrebna posebna navodila. Plastenko z raztopino shranjujte v zunanji ovojnini za zagotovitev zaščite pred svetlobo. Shranjujte pri temperaturi do 25°C. Shranjujte v originalni ovojnini in nedosegljivo otrokom. Literatura: 1. Lipton A, et al. Eur J Cancer. 2012; 48: 3082–3092. Podatki integrirane analize preskušanj III. faze, ki so vključevala bolnike z rakom dojke, rakom prostate, ostalimi solidnimi tumorji ali diseminiranim plazmocitomom, ki so imeli zasevke v kosteh.1 * XGEVA® značilno podaljša čas do pojava prvega zapleta kostnih zasevkov (ZKZ) za 8,21 mesecev in zmanjša tveganje za pojav prvega ZKZ za 17% (HR; 0,83 [95% CI; 0,76–0,90]; p < 0,001) v primerjavi z zoledronsko kislino. °Opredelitev kostnih zapletov: Preprečevanje zapletov kostnih zasevkov (ZKZ) in z njimi povezane bolečine pri odraslih s kostnimi zasevki solidnih tumorjev. XGEVA® 120 mg raztopina za injiciranje (denosumab) – SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA Samo za strokovno javnost. Pred predpisovanjem si preberite celoten Povzetek glavnih značilnosti zdravila. .Za to zdravilo se izvaja dodatno spremljanje varnosti. Poročati je potrebno o vseh domnevnih neželenih učinkih zdravila. SESTAVA ZDRAVILA: Ena viala vsebuje 120 mg denosumaba v 1,7 ml raztopine (70 mg/ml). Pomožne snovi s prepoznavnim delovanjem: 1,7 ml raztopine vsebuje 78 mg sorbitola (E420). TERAPEVTSKE INDIKACIJE: Preprečevanje skeletnih dogodkov (patoloških zlomov obsevanja kosti, kompresije hrbtenjače ali operacije kosti) pri odraslih s kostnimi metastazami solidnih tumorjev. Zdravljenje odraslih bolnikov in skeletno dozorelih mladostnikov z gigantocelularnim kostnim tumorjem, ki ni operabilen, ali pri katerem bi kirurška odstranitev verjetno povzročila hudo obolevnost. ODMERJANJE IN NAČIN UPORABE: Vsi bolniki morajo prejemati dodatek vsaj 500 mg kalcija in 400 i.e. vitamina D dnevno, razen če ima bolnik hiperkalciemijo. Bolniki morajo dobiti navodilo za uporabo in opozorilno kartico za bolnika. Kostne metastaze solidnih tumorjev: Priporočeni odmerek zdravila XGEVA® za preprečevanje skeletnih dogodkov je 120 mg enkrat na 4 tedne v enkratni subkutani injekciji v stegno, trebuh ali nadlaket. Gigantocelularni kostni tumor: Priporočeni odmerek zdravila XGEVA® za zdravljenje gigantocelularnega kostnega tumorja je 120 mg enkrat na 4 tedne v enkratni subkutani injekciji v stegno, trebuh ali nadlaket ter dodaten odmerek 120 mg 8. in 15. dan zdravljenja v prvem mesecu terapije. Bolnike z gigantocelularnim kostnim tumorjem je treba v rednih presledkih ocenjevati, da bi ugotovili, ali jim zdravljenje še koristi. Pri bolnikih, ki imajo bolezen z zdravilom XGEVA® obvladano, niso ocenili učinka prekinitve ali prenehanja zdravljenja, toda omejeni podatki pri teh bolnikih ne kažejo povratnega učinka po prenehanju zdravljenja. Bolniki z okvaro edvic: Prilagoditev odmerka ni potrebna. Bolniki z okvaro jeter: Varnost in učinkovitost denosumaba nista raziskani. Starejši bolniki (stari . 65 et): Prilagoditev odmerka ni potrebna. Pediatrični bolniki: Odmerjanje za zdravljenje skeletno dozorelih mladostnikov z gigantocelularnim kostnim tumorjem, ki ni operabilen, ali pri katerem bi kirurška odstranitev verjetno povzročila hudo obolevnost, je enako kot za odrasle. Zdravilo XGEVA® ni priporočljivo za pediatrične bolnike (stare < 18 let), ker varnost in učinkovitost zdravila XGEVA® pri teh bolnikih nista ocenjeni, z izjemo skeletno dozorelih mladostnikov z gigantocelularnim kostnim tumorjem. Za subkutano uporabo. Zdravilo XGEVA® mora aplicirati zdravstveni delavec. KONTRAINDIKACIJE: Preobčutljivost na zdravilno učinkovino ali katero koli pomožno snov Huda, nezdravljena hipokalciemija. Nezaceljene lezije po zobnih ali ustnih kirurških posegih. POSEBNA OPOZORILA IN PREVIDNOSTNI UKREPI: Vsi bolniki morajo prejemati dodatek kalcija in vitamina D, razen če ima bolnik hiperkalciemijo. Obstoječo hipokalciemijo je treba odpraviti še pred začetkom zdravljenja z zdravilom XGEVA®. Hipokalciemija se lahko pojavi kadarkoli med zdravljenjem z zdravilom XGEVA® Kontrolo koncentracije kalcija je treba izvesti (i) pred prvim odmerkom zdravila XGEVA®, (ii) v dveh tednih po prvem odmerku, (iii) če se pojavijo simptomi, sumljivi za hipokalciemijo. O dodatnih kontrolah koncentracije kalcija med zdravljenjem je treba razmisliti pri bolnikih, ki imajo dejavnike tveganja za hipokalciemijo, ali če so takšne kontrole sicer umestne glede na bolnikovo klinično stanje. Bolnikom je treba naročiti, naj zdravnika obvestijo o simptomih, ki kažejo na hipokalciemijo. Če se med prejemanjem zdravila XGEVA® pojavi hipokalciemija, je lahko potrebno dodatno dodajanje kalcija in dodatne kontrole. Bolniki s hudo okvaro ledvic (očistek kreatinina < 30 ml/min) ali bolniki na dializi imajo večje tveganje za pojav hipokalciemije. Tveganje za pojav hipokalciemije in spremljajočega zvišanja paratiroidnega hormona se povečuje s povečano stopnjo okvare ledvic. Pri takšnih bolnikih so redne kontrole koncentracije kalcija posebej pomembne. Pri bolnikih, ki imajo nezaceljene lezije mehkih tkiv v ustih, je treba začetek zdravljenja/nov ciklus zdravljenja odložiti. Pred zdravljenjem z zdravilom XGEVA® je priporočljivo opraviti zobozdravstveni pregled in preventivno zobozdravstveno oskrbo ter individualno oceno koristi in tveganja. Pri ocenjevanju bolnikovega tveganja za pojav osteonekroze čeljustnice je treba upoštevati naslednje dejavnike tveganja: moč zdravila, ki zavira resorpcijo kosti (tveganje je večje z zelo močnimi spojinami), pot uporabe (tveganje je večje v primeru parenteralne uporabe) in kumulativni odmerek zdravila, uporabljenega za zdravljenje resorpcije kosti, rak, sočasne bolezni (npr. anemijo, koagulopatije, okužbo), kajenje, sočasna zdravljenja: kortikosteroide, kemoterapijo, zaviralce angiogeneze, radioterapijo glave in vratu, slabo ustno higieno, periodontalno bolezen, slabo prilegajoče se zobne proteze, že obstoječo zobno bolezen, invazivne zobozdravstvene posege, npr ekstrakcije zob. Vsem bolnikom je treba naročiti, da morajo med zdravljenjem z zdravilom XGEVA® vzdrževati dobro ustno higieno, redno opravljati zobozdravniške preglede in nemudoma obvestiti zdravnika, če se pojavi kakršen koli simptom v ustih, na primer majanje zob, bolečina, oteklina, rana, ki se ne celi, ali izcedek. Med zdravljenjem je izvajanje invazivnih zobozdravniških posegov dovoljeno le po skrbnem razmisleku in se jim je treba izogniti v bližini termina za odmerjanje zdravila XGEVA®. Načrt vodenja bolnikov, ki se jim pojavi osteonekroza čeljustnice, je treba oblikovati na podlagi tesnega sodelovanja med lečečim zdravnikom in zobozdravnikom ali ustnim kirurgom, ki ima izkušnje z osteonekrozo čeljustnice. Razmisliti je treba o začasnem prenehanju zdravljenja z zdravilom XGEVA®, dokler se to stanje ne razreši in se sovpleteni dejavniki tveganja ublažijo, če je mogoče. Atipični zlomi stegnenice se lahko pojavijo že ob majhni poškodbi ali celo brez poškodbe, in sicer v subtrohanternem in diafiznem predelu stegnenice. Za te dogodke so značilni specifični radiografski izvidi. O njih so poročali tudi pri bolnikih z določenimi sočasnimi bolezenskimi stanji (npr s pomanjkanjem vitamina D, revmatoidnim artritisom, hipofosfatazijo) in med uporabo določenih zdravil (npr. difosfonatov glukokortikoidov, zaviralcev protonske črpalke). Ti dogodki so se pojavili tudi brez antiresorpcijskega zdravljenja. Podobni zlomi, opisani v zvezi z difosfonati, so pogosto obojestranski, zato je treba pri bolnikih, ki se zdravijo z denosumabom in so imeli zlom srednjega dela stegnenice, opraviti tudi pregled druge stegnenice. Pri bolnikih, pri katerih obstaja sum na atipičen zlom stegnenice, je treba razmisliti o prenehanju uporabe zdravila XGEVA® ob vrednotenju bolnika glede na individualno oceno koristi in tveganja. Bolnikom je treba naročiti, da morajo med zdravljenjem z zdravilom XGEVA® zdravniku poročati o novi ali nenavadni bolečini v stegnu, kolku ali dimljah. Bolnike s takšnimi simptomi je treba preiskati glede nepopolnega zloma stegnenice. Bolniki, zdravljeni z zdravilom XGEVA®, sočasno ne smejo prejemati drugih zdravil, ki vsebujejo denosumab (za indikacije pri osteoporozi), in difosfonatov Malignost pri gigantocelularnem kostnem tumorju ali napredovanje do metastatske bolezni je redek dogodek in je znano tveganje pri bolnikih z gigantocelularnim kostnim tumorjem. Bolnike je treba kontrolirati glede radioloških znakov malignosti, nove radiolucentnosti ali osteolize. Razpoložljivi klinični podatki ne kažejo povečanega tveganja za malignost pri bolnikih z gigantocelularnim kostnim tumorjem, zdravljenih z zdravilom XGEVA®. Bolniki z redko prirojeno motnjo intolerance za fruktozo ne smejo uporabljati zdravila XGEVA® . INTERAKCIJE: Študij medsebojnega delovanja niso izvedli. V kliničnih preskušanjih sočasna kemoterapija in/ali hormonsko zdravljenje ali predhodna intravenska izpostavljenost difosfonatom niso klinično pomembno spremenili najmanjše koncentracije denosumaba v serumu in farmakodinamike denosumaba (N-telopeptid v urinu, prilagojen na kreatinin, uNTx/Cr). POVZETEK NEŽELENIH UČINKOV: Zelo pogosti (. 1/10): dispneja, driska, mišično-skeletna bolečina. Pogosti (. 1/100 do < 1/10): hipokalciemija, hipofosfatemija, ekstrakcija zoba, hiperhidroza, osteonekroza čeljustnice. Redki (. 1/10.000 do < 1/1.000): preobčutljivost na zdravilo, anafilaktična reakcija, atipični zlom stegnenice. FARMACEVTSKI PODATKI: Shranjujte v hladilniku (2 °C 8 °C). Ne zamrzujte. NAČIN IN REŽIM PREDPISOVANJA TER IZDAJE ZDRAVILA: Predpisovanje in izdaja zdravila je le na recept s posebnim režimom ZZ. IMETNIK DOVOLJENJA ZA PROMET: Amgen Europe B.V., Minervum 7061, NL 4817 ZK Breda, Nizozemska. Dodatna pojasnila lahko dobite v lokalni pisarni: Amgen zdravila d.o.o., Šmartinska 140, SI 1000 Ljubljana. DATUM ZADNJE REVIZIJE BESEDILA: Julij 2015. DATUM PRIPRAVE INFORMACIJE: Oktober 2015. Podrobne informacije o zdravilu so objavljene na spletni strani Evropske agencije za zdravila http://www.ema.europa.eu/. SLHR SL P 162x 0615 107510(1) SLHR-SL-P-003-0615-107089(1) NEULASTA® 6 mg raztopina za injiciranje (pegfilgrastim) – SKRAJŠAN POVZETEK GLAVNIH ZNAČILNOSTI ZDRAVILA Samo za strokovno javnost. Pred predpisovanjem si preberite celoten Povzetek glavnih značilnosti zdravila. SESTAVA ZDRAVILA: Ena napolnjena injekcijska brizga vsebuje 6 mg pegfilgrastima v 0,6 ml (10 mg/ml) raztopine za injiciranje. TERAPEVTSKE INDIKACIJE: Skrajšanje trajanja nevtropenije in zmanjšanje incidence febrilne nevtropenije pri odraslih bolnikih, zdravljenih s citotoksično kemoterapijo za maligne bolezni (z izjemo kronične mieloidne levkemije in mielodisplastičnih sindromov). ODMERJANJE IN NAČIN UPORABE: Zdravljenje z zdravilom Neulasta® morajo uvesti in nadzorovati zdravniki, izkušeni v onkologiji in/ali hematologiji. Za vsak cikel kemoterapije priporočajo en 6 mg odmerek (eno napolnjeno injekcijsko brizgo) zdravila Neulasta®, ki je dana vsaj 24 ur po citotoksični kemoterapiji. Zdravilo Neulasta® se injicira subkutano. Injekcije se morajo dati v stegno, trebuh ali zgornji del roke. Varnost in učinkovitost zdravila Neulasta® pri otrocih še nista bili dokazani in priporočil o odmerjanju ni mogoče dati. Pri bolnikih z okvaro ledvic in s končno odpovedjo ledvic odmerka ni treba spreminjati. KONTRAINDIKACIJE: Preobčutljivost za zdravilno učinkovino ali katerokoli pomožno snov. POSEBNA OPOZORILA IN PREVIDNOSTNI UKREPI: Pri bolnikih z de novo akutno mieloično levkemijo omejeni klinični podatki kažejo primerljiv učinek pegfilgrastima in filgrastima na čas do okrevanja po hudi nevtropeniji. Dolgoročni učinki zdravila Neulasta® pri akutni mieloični levkemiji niso ugotovljeni, zato ga je treba pri tej populaciji bolnikov uporabljati previdno. Varnost in učinkovitost zdravila Neulasta® nista raziskani pri bolnikih z mielodisplastičnim sindromom, s kronično mielogeno levkemijo in s sekundarno akutno mieloično levkemijo (AML), zato ga pri takšnih bolnikih ne smete uporabljati. Posebno pozornost je treba nameniti razlikovanju diagnoze blastne transformacije kronične mieloične levkemije od akutne mieloične levkemije. Varnost in učinkovitost uporabe zdravila Neulasta® pri bolnikih z de novo AML, mlajših od 55 let in s citogenetiko t(15;17), nista ugotovljeni. Varnosti in učinkovitosti zdravila Neulasta® niso raziskovali pri bolnikih, ki prejemajo kemoterapijo v velikih odmerkih. Tega zdravila ne smete uporabljati za zvečevanje odmerka citotoksične kemoterapije preko uveljavljenih shem odmerjanja. Neželene reakcije na pljučih: Bolj ogroženi so lahko bolniki z nedavno anamnezo pljučnih infiltratov ali pljučnice. Pojav pljučnih znakov, kot so kašelj, zvišana telesna temperatura in dispneja v povezavi z radiološkimi znaki pljučnih infiltratov, in poslabšanje pljučne funkcije skupaj z zvečanim številom nevtrofilcev utegnejo biti preliminarni znaki sindroma akutne dihalne stiske (ARDS -'Acute Respiratory Distress Syndrome'). V takih primerih je treba zdravilo Neulasta® po presoji zdravnika prenehati dajati in poskrbeti za ustrezno zdravljenje. Glomerulonefritis: Na splošno so primeri glomerulonefritisa minili po zmanjšanju odmerka ali prenehanju uporabe filgrastima ali pegfilgrastima. Priporočljivo je spremljanje laboratorijskih izvidov urina. Sindrom kapilarne prepustnosti: Bolnike, ki se jim pojavijo simptomi sindroma kapilarne prepustnosti, je treba natančno kontrolirati in deležni morajo biti standardnega simptomatskega zdravljenja, ki lahko vključuje potrebo po intenzivni negi. Splenomegalija in ruptura vranice: Skrbno je treba spremljati velikost vranice (s kliničnim pregledom, ultrazvokom). Na diagnozo rupture vranice moramo misliti pri bolnikih, ki poročajo o bolečini v zgornjem levem delu trebuha ali v predelu lopatice. Trombocitopenija in anemija: Zdravljenje s samim zdravilom Neulasta® ne prepreči trombocitopenije in anemije, ker se hkrati vzdržuje mielosupresivna kemoterapija s polnimi odmerki po predpisani shemi. Priporočajo redno spremljanje števila trombocitov in hematokrita. Posebna previdnost je potrebna med uporabo posameznih kemoterapevtikov ali njihovih kombinacij, za katere je znano, da povzročajo hudo trombocitopenijo. Srpastocelična anemija: Pri bolnikih s srpastocelično dispozicijo ali s srpastocelično anemijo je bila uporaba pegfilgrastima povezana s srpastocelično krizo, zato se mora pri teh bolnikih zdravilo Neulasta® predpisovati previdno in spremljati ustrezne klinične parametre in laboratorijski status in biti pozoren na morebitno povezavo tega zdravila z zvečanjem vranice in vazookluzivno krizo. Levkocitoza: Zaradi kliničnih učinkov zdravila Neulasta® in zaradi možnosti levkocitoze je treba med zdravljenjem redno kontrolirati število belih krvničk. Če število levkocitov po pričakovanem najmanjšem številu preseže 50 x 109/l, je treba nemudoma prenehati z zdravljenjem s tem zdravilom. Preobčutljivost: Dokončno prenehajte z zdravljenjem z zdravilom Neulasta® pri bolnikih s klinično signifikantno preobčutljivostjo. Zdravila Neulasta® ne dajajte bolnikom z anamnezo preobčutljivosti na pegfilgrastim ali filgrastim. V primeru resne alergijske reakcije je treba poskrbeti za ustrezno zdravljenje in pazljivo spremljanje bolnika še nekaj dni. Imunogenost: Kot pri vseh terapevtskih beljakovinah obstaja možnost imunogenosti. Stopnja nastajanja protiteles proti pegfilgrastimu je na splošno nizka. Vezavna protitelesa se pojavijo po pričakovanjih pri vseh bioloških zdravilih, vendar jih doslej niso povezali z nevtralizacijskim delovanjem. Varnosti in učinkovitosti zdravila Neulasta® za mobilizacijo matičnih krvotvornih celic pri bolnikih ali zdravih dajalcih niso primerno ovrednotili. Pokrovček igle pri napolnjeni injekcijski brizgi vsebuje suho naravno gumo (derivat lateksa), ki lahko povzroča alergične reakcije. Povečana hemopoetična aktivnost kostnega mozga zaradi zdravljenja z rastnimi dejavniki je bila povezana s prehodnimi pozitivnimi izvidi pri slikanju kosti, kar je treba upoštevati pri interpretaciji izvidov na podlagi slikanja kosti. Zdravilo Neulasta® vsebuje sorbitol. Bolniki z redko prirojeno motnjo intolerance za fruktozo ne smejo dobiti tega zdravila. Zdravilo Neulasta® vsebuje manj kot 1 mmol (23 mg) natrija na 6 mg odmerek, kar v bistvu pomeni ˝brez natrija˝. Za izboljšanje sledljivosti granulocitne kolonije spodbujajočih faktorjev (G-CSF) je treba v bolnikovi dokumentaciji jasno zabeležiti zaščiteno ime uporabljenega zdravila. MEDSEBOJNO DELOVANJE ZDRAVIL IN DRUGE OBLIKE INTERAKCIJ: Zaradi možne občutljivosti hitro se delečih mieloidnih celic za citotoksično kemoterapijo je treba zdravilo Neulasta® dati vsaj 24 ur po aplikaciji citotoksične kemoterapije. Sočasne uporabe zdravila Neulasta® s katerimkoli kemoterapevtskim zdravilom pri bolnikih niso ovrednotili. NEŽELENI UČINKI: Podatki opisujejo neželene učinke, zabeležene v kliničnih preskušanjih in med spontanim poročanjem. Zelo pogosti (. 1/10): glavobol, navzea, bolečina v kosteh. Pogosti (. 1/100 do < 1/10): trombocitopenija, levkocitoza, mišično-skeletna bolečina (mialgija, artralgija, bolečina v okončini, bolečina v hrbtu, mišično-skeletna bolečina, bolečina v vratu), bolečina na mestu injiciranja, bolečina v prsih, ki ne izvira od srca. Občasni (. 1/1.000 do < 1/100): srpastocelična kriza, splenomegalija, ruptura vranice, preobčutljivostne reakcije, anafilaksija, zvišanje sečne kisline, sindrom kapilarne prepustnosti, sindrom akutne dihalne stiske, pljučne neželene reakcije (intersticijska pljučnica, pljučni edem, pljučni infiltrati in pljučna fibroza), Sweetov sindrom (akutna febrilna dermatoza), kožni vaskulitis, reakcije na mestu injiciranja, zvišanje laktat-dehidrogenaze in alkalne fosfataze, prehodno zvišanje jetrnih funkcijskih testov za ALT ali AST, glomerulonefritis. FARMACEVTSKI PODATKI: Shranjujte v hladilniku (2°C – 8°C ). Ne zamrzujte. Zdravilo Neulasta® sme biti izpostavljeno sobni temperaturi (ne nad 30°C) za enkratno obdobje, ki ne sme preseči 72 ur. Zdravilo Neulasta® ni kompatibilno z raztopinami natrijevega klorida. NAČIN IN REŽIM PREDPISOVANJA TER IZDAJE ZDRAVILA: Predpisovanje in izdaja zdravila je le na recept s posebnim režimom – H/Rp. IMETNIK DOVOLJENJA ZA PROMET: Amgen Europe B.V., 4817 ZK Breda, Nizozemska. Dodatna pojasnila lahko dobite v lokalni pisarni: Amgen zdravila d.o.o., Šmartinska 140, SI-1000 Ljubljana. DATUM ZADNJE REVIZIJE BESEDILA: Maj 2015. Podrobni podatki o tem zdravilu so na voljo na spletni strani Evropske agencije za zdravila http://www.ema.europa.eu/. Literatura: 1.) Povzetek glavnih značilnosti zdravila Neulasta®, Amgen, 2015. Erbitux 5 mg/ml raztopina za infundiranje Skrajšan povzetek glavnih znaËilnosti zdravila Sestava: En ml raztopine za infundiranje vsebuje 5 mg cetuksimaba in pomožne snovi. Cetuksimab je himerno monoklonsko IgG1 protitelo. Terapevtske indikacije: Zdravilo Erbitux je indicirano za zdravljenje bolnikov z metastatskim kolorektalnim rakom z ekspresijo receptorjev EGFR in nemutiranim tipom RAS v kombinaciji s kemoterapijo na osnovi irinotekana, kot primarno zdravljenje v kombinaciji s FOLFOX in kot samostojno zdravilo pri bolnikih, pri katerih zdravljenje z oksaliplatinom in zdravljenje na osnovi irinotekana ni bilo uspešno in pri bolnikih, ki ne prenašajo irinotekana. Zdravilo Erbitux je indicirano za zdravljenje bolnikov z rakom skvamoznih celic glave in vratu v kombinaciji z radioterapijo za lokalno napredovalo bolezen in v kombinaciji s kemoterapijo na osnovi platine za ponavljajoËo se in/ali metastatsko bolezen. Odmerjanje in naËin uporabe: Zdravilo Erbitux pri vseh indikacijah infundirajte enkrat na teden. Pred prvo infuzijo mora bolnik prejeti premedikacijo z antihistaminikom in kortikosteroidom najmanj 1 uro pred uporabo cetuksimaba. ZaËetni odmerek je 400 mg cetuksimaba na m2 telesne površine. Vsi naslednji tedenski odmerki so vsak po 250 mg/m2 . Kontraindikacije: Zdravilo Erbitux je kontraindicirano pri bolnikih z znano hudo preobËutljivostno reakcijo (3. ali 4. stopnje) na cetuksimab. Kombinacija zdravila Erbitux s kemoterapijo, ki vsebuje oksaliplatin, je kontraindicirana pri bolnikih z metastatskim kolorektalnim rakom z mutiranim tipom RAS ali kadar status RAS ni znan. Posebna opozorila in previdnostni ukrepi: Pojav hude reakcije, povezane z infundiranjem, zahteva takojšnjo in stalno ukinitev terapije s cetuksimabom. »e pri bolniku nastopi blaga ali zmerna reakcija, povezana z infundiranjem, lahko zmanjšate hitrost infundiranja. PriporoËljivo je, da ostane hitrost infundiranja na nižji vrednosti tudi pri vseh naslednjih infuzijah. »e se pri bolniku pojavi kožna reakcija, ki je ne more prenašati, ali huda kožna reakcija (. 3. stopnje po kriterijih CTCAE), morate prekiniti terapijo s cetuksimabom. Z zdravljenjem smete nadaljevati le, Ëe se je reakcija izboljšala do 2. stopnje. »e ugotovite intersticijsko bolezen pljuË, morate zdravljenje s cetuksimabom prekiniti, in bolnika ustrezno zdraviti. Zaradi možnosti pojava znižanja nivoja elektrolitov v serumu se pred in periodiËno med zdravljenjem s cetuksimabom priporoËa doloËanje koncentracije elektrolitov v serumu. Pri bolnikih, ki prejemajo cetuksimab v kombinaciji s kemoterapijo na osnovi platine, obstaja veËje tveganje za pojav hude nevtropenije. Takšne bolnike je potrebno skrbno nadzorovati. Pri predpisovanju cetuksimaba je treba upoštevati kardiovaskularno stanje in indeks zmogljivosti bolnika in soËasno dajanje kardiotoksiËnih uËinkovin kot so fluoropirimidini. »e je diagnoza ulcerativnega keratitisa potrjena, je treba zdravljenje s cetuksimabom prekiniti ali ukiniti. Cetuksimab je treba uporabljati previdno pri bolnikih z anamnezo keratitisa, ulcerativnega keratitisa ali zelo suhih oËi. Cetuksimaba ne uporabljajte za zdravljenje bolnikov s kolorektalnim rakom, Ëe imajo tumorje z mutacijo RAS ali pri katerih je tumorski status RAS neznan. Interakcije: Pri kombinaciji s fluoropirimidini se je v primerjavi z uporabo fluoropirimidinov, kot monoterapije, poveËala pogostnost srËne ishemije, vkljuËno z miokardnim infarktom in kongestivno srËno odpovedjo ter pogostnost sindroma dlani in stopal. V kombinaciji s kemoterapijo na osnovi platine se lahko poveËa pogostnost hude levkopenije ali hude nevtropenije. V kombinaciji s kapecitabinom in oksaliplatinom (XELOX) se lahko poveËa pogostnost hude driske. Neželeni uËinki: Zelo pogosti (. 1/10): hipomagneziemija, poveËanje ravni jetrnih encimov, kožne reakcije, blage ali zmerne reakcije povezane z infundiranjem, mukozitis, v nekaterih primerih resen. Pogosti (. 1/100 do < 1/10): dehidracija, hipokalciemija, anoreksija, glavobol, konjunktivitis, driska, navzeja, bruhanje, hude reakcije povezane z infundiranjem, utrujenost. Posebna navodila za shranjevanje: Shranjujte v hladilniku (2 °C -8 °C). Pakiranje: 1 viala z 20 ml ali 100 ml raztopine. NaËin in režim izdaje: Izdaja zdravila je le na recept-H. Imetnik dovoljenja za promet: Merck KGaA, 64271 Darmstadt, NemËija. Datum zadnje revizije besedila: november 2014. Pred predpisovanjem zdravila natanËno preberite celoten Povzetek glavnih znaËilnosti zdravila. Samo za strokovno javnost. Podrobnejše informacije so na voljo pri predstavniku imetnika dovoljenja za promet z zdravilom: Merck d.o.o., Ameriška ulica 8, 1000 Ljubljana, tel.: 01 560 3810, faks: 01 560 3830, el. pošta: info@merck.si www.merckserono.net www.Erbitux-international.com PM-ONC-05/2015 /13.10.2015 jetrne bolezni; treba jih je nadzorovati in voditi v skladu z lokalnimi zdravstvenimi standardi, da bi preprečili reaktivacijo hepatitisa. Progresivna multifokalna levkoencefalopatija (PML): Pri bolnikih, zdravljenih z zdravilom Gazyvaro, je bila opisana PML. Na diagnozo PML je treba pomisliti pri vsakem bolniku z novonastalimi nevrološkimi spremembami ali spremembi že obstoječih nevroloških stanj. Simptomi PML so nespecifični in se lahko razlikujejo glede na prizadet možganski predel. Pogosti so motorični simptomi s prizadetostjo kortikospinalnega trakta, senzorične nepravilnosti, cerebelarni simptomi in izpadi vidnega polja. Pojavijo se lahko nekateri znaki ali simptomi, ki veljajo za “kortikalne”. Ovrednotenje PML vključuje posvet z nevrologom, magnetnoresonančno slikanje možganov in lumbalno punkcijo. Med preiskovanjem suma na PML je treba zdravljenje z zdravilom Gazyvaro prekiniti, če je PML potrjena, pa ga je treba trajno končati. V poštev pride tudi prenehanje ali zmanjšanje morebitne sočasne kemoterapije ali imunosupresivnega zdravljenja. Bolnika je treba napotiti k nevrologu za ovrednotenje in zdravljenje PML. Imunizacija: Varnost imunizacije z živimi ali oslabljenimi virusnimi cepivi po zdravljenju z zdravilom Gazyvaro ni raziskana in cepljenje z živimi virusnimi cepivi med zdravljenjem in do okrevanja celic B ni priporočljivo. Izpostavljenost obinutuzumabu in utero in cepljenje novorojenčkov z živimi virusnimi cepivi: Zaradi možnega zmanjšanega števila celic B pri novorojenčkih po izpostavljenosti obinutuzumabu med nosečnostjo je treba pri njih kontrolirati njihovo število. Cepljenja z živimi virusnimi cepivi je treba odložiti, dokler se število celic B pri dojenčku ne popravi. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij: Študij medsebojnega delovanja niso izvedli. Farmakokinetično medsebojno delovanje: Obinutuzumab ni substrat, zaviralec ali induktor CYP450, encimov uridindifosfat-glukuroniltransferaze (UGT) ali prenašalcev, kakršen je P-glikoprotein. Zato ni pričakovati farmakokinetičnih medsebojnih delovanj z zdravili, ki se presnavljajo s temi encimskimi sistemi. Farmakodinamično medsebojno delovanje: Zaradi imunosupresivnega učinka obinutuzumaba cepljenje z živimi virusnimi cepivi ni priporočljivo med zdravljenjem in do okrevanja celic B. Obinutuzumab v kombinaciji s klorambucilom lahko poveča delež nevtropenije. Neželeni učinki: Najpogostejši neželeni učinek pri prejemnikih zdravila Gazyvaro so bile reakcije, povezane z infuzijo, ki so se med prvim ciklusom zdravljenja pojavile pri večini bolnikov. Incidenca z infundiranjem povezanih simptomov se je nato bistveno zmanjšala: s 65 % med infuzijo prvih 1000 mg zdravila Gazyvaro na manj kot 3 % med poznejšimi infuzijami. V ključni študiji se je nevtropenija pojavila pri 41 % bolnikov in trombocitopenija pri 15 % bolnikov. Med drugimi resnimi neželenimi učinki, zabeleženimi med kliničnim razvojem, so bili sindrom razpada tumorja, srčni dogodki in, zelo redko, PML. Ostali neželeni učinki, zabeleženih z večjo incidenco (razlika . 2 %) med prejemniki kombinacije zdravila Gazyvaro in klorambucila ter med prejemniki klorambucila samega ali kombinacije rituksimaba in klorambucila vseh stopenj: Zelo pogosti: anemija, diareja in zvišana telesna temperatura. Pogosti: okužba sečil, nazofaringitis, herpes labialis, rinitis, faringitis, ploščatocelični karcinom kože, levkopenija, hiperurikemija, atrijska fibrilacija, hipertenzija, kašelj, zaprtost, alopecija, artralgija, bolečina v hrbtu, mišično-skeletna bolečina v prsnem košu, zmanjšanje števila belih krvnih celic, zmanjšanje števila nevtrofilcev in povečanje telesne mase. Poročanje o domnevnih neželenih učinkih: Poročanje o domnevnih neželenih učinkih zdravila po izdaji dovoljenja za promet je pomembno. Omogoča namreč stalno spremljanje razmerja med koristmi in tveganji zdravila. Od zdravstvenih delavcev se zahteva, da poročajo o katerem koli domnevnem neželenem učinku zdravila na: Univerzitetni klinični center Ljubljana, Interna klinika, Center za zastrupitve, Zaloška cesta 2, SI-1000 Ljubljana, Faks: + 386 (0)1 434 76 46, e-pošta: farmakovigilanca@kclj.si. Režim izdaje zdravila: H Imetnik dovoljenja za promet: Roche Registration Limited, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW, Velika Britanija Verzija: 2.0/15 Informacija pripravljena: avgust 2015. Samo za strokovno javnost DODATNE INFORMACIJE SO NA VOLJO PRI: Roche farmacevtska družba d.o.o., Vodovodna cesta 109, 1000 Ljubljana PG-56-15-GAZ-TG Powered by Siemens’ exclusive FlowMotion™ technology, Biograph mCT Flow™ is the world’s first PET•CT system to eliminate stop and go imaging. Now with Biograph mCT Flow, planning and scanning is based on a continuous flow motion of the patient table. The new Biograph mCT Flow provides the finest** image resolution for every patient in every scan. FlowMotion enables accurate and reproducible quantification in every dimension. Simple and precise range planning eliminates over-scanning and its associated radiation exposure, while at the same time streamlining workflow. And FlowMotion’s sense of continuous progress provides a more comfortable exam experience for every patient. Biograph mCT Flow enables you to make unprecedented progress in diagnosing the most challenging diseases. Overcoming the limitations of conventional PET/CT, the new Biograph mCT Flow with FlowMotion technology is the end of stop and go. * Biograph mCT Flow is not commercially available in all countries. Due to regulatory reasons its future availability cannot be guaranteed. Please contact your local Siemens organization for further details. ** Based on competitive literature available at time of publication. Data on file. Answers for life. Skrajšan povzetek glavnih značilnosti zdravila Adcetris ˇ (brentuksimab vedotin) Pred predpisovanjem preberite celoten povzetek glavnih značilnosti zdravila (SmPC). Farmacevtska oblika: 50 mg prašek za koncentrat za raztopino za infundiranje Terapevtske indikacije: Zdravljenje odraslih bolnikov s ponovljenim ali z refraktarnim CD30+ Hodgkinovim limfomom (HL) po presaditvi avtolognih matičnih celic (ASCT) ali po vsaj dveh predhodnih zdravljenjih, ko ASCT ali večtirna kemoterapija kot obliki zdravljenja ne prideta v poštev. Zdravljenje odraslih bolnikov s ponovljenim ali z refraktarnim sistemskim anaplastičnim velikoceličnim limfomom (sALCL). Odmerjanje in način uporabe: Zdravilo Adcetris se mora uporabljati pod nadzorom zdravnika, ki ima izkušnje z uporabo zdravil proti raku. Priporočeni odmerek je 1,8 mg/ kg v obliki 30-minutne intravenske infuzije vsake 3 tedne (če je telesna masa bolnika večja od 100 kg, je pri izračunu odmerka treba uporabiti vrednost 100 kg). Zdravljenje se mora nadaljevati do napredovanja bolezni ali pojava nesprejemljivih toksičnih učinkov. Bolniki, pri katerih se bolezen stabilizira ali izboljša, morajo prejeti najmanj 8 ciklov in do največ 16 ciklov. Pred aplikacijo vsakega odmerka tega zdravila je treba preveriti celotno krvno sliko bolnika. Bolnike je treba nadzirati med infundiranjem zdravila in po njem. Za navodila glede rekonstitucije in načina uporabe zdravila glejte poglavje 6.6 v SmPC-ju. Prilagoditve odmerka: Če se med zdravljenjem razvije nevtropenija, jo je treba uravnati z odložitvijo odmerka (glejte SmPC). Če se med zdravljenjem pojavi ali poslabša periferna senzorična ali motorična nevropatija, bo odmerek morda treba odložiti ali zmanjšati ali zdravljenje z zdravilom Adcetris prekiniti (glejte SmPC). Okvara ledvic ali jeter: Podatkov o uporabi pri bolnikih z okvaro ledvic ali jeter ni na voljo. Starejši bolniki (. 65 let): Podatkov ni na voljo. Pediatrična populacija (< 18 let): Podatkov ni na voljo. V predkliničnih študijah so opazili upadanje priželjca. Kontraindikacije: Preobčutljivost na zdravilno učinkovino ali katero koli navedeno pomožno snov. Kombinirana uporaba bleomicina in zdravila Adcetris povzroča pljučno toksičnost. Posebna opozorila in previdnostni ukrepi: Pri bolnikih, ki so zdravilo Adcetris prejeli po predhodnem zdravljenju z več režimi kemoterapije, so poročali o progresivni multifokalni levkoencefalopatiji (PML). Bolnike je treba skrbno nadzirati glede nastanka novih ali poslabšanja obstoječih nevroloških, kognitivnih ali vedenjskih znakov ali simptomov, ki lahko nakazujejo na PML. V primeru kakršnega koli suma na PML je treba uporabo zdravila Adcetris začasno prekiniti. Če se diagnoza PML potrdi, je treba zdravljenje z zdravilom Adcetris trajno prekiniti. Pri bolnikih, ki so se zdravili z brentuksimabom vedotinom, so poročali o pojavih akutnega pankreatitisa, vključno s smrtnimi izidi. Bolnike je treba skrbno nadzirati glede pojava nove ali poslabšanja že obstoječe bolečine v trebuhu. V primeru potrjene diagnoze akutnega pankreatitisa je treba zdravljenje z brentuksimabom vedotinom trajno ukiniti. Pri bolnikih, ki so se zdravili z brentuksimabom vedotinom, so poročali o primerih toksičnih učinkov na pljuča. Vzročna povezava z brentuksimabom vedotinom sicer ni bila ugotovljena, vendar pa tveganja toksičnosti za pljuča ni mogoče izključiti. V primeru pojava novih ali poslabšanja že obstoječih pljučnih simptomov (npr. kašelj, dispneja) je nemudoma treba izvesti ustrezne diagnostične postopke in bolnika ustrezno zdraviti. septični šok (vključno s smrtnimi izidi) in herpes zoster ter o oportunističnih okužbah, kot so pljučnica in oralna kandidoza. Bolnike je treba med zdravljenjem skrbno nadzirati glede pojava možnih resnih in oportunističnih okužb. Poročali so tako o takojšnjih in zakasnelih reakcijah, povezanih z infundiranjem, kot o ana.laktičnih reakcijah. Bolnike je treba med infundiranjem in po njem skrbno nadzirati. Če se pojavi ana.laktična reakcija, je treba dajanje zdravila Adcetris takoj in trajno ukiniti. Če se pojavijo reakcije, povezane z infundiranjem, je treba z infundiranjem prenehati (glejte SmPC). Pri bolnikih s hitro proliferacijskimi tumorji in z veliko tumorsko obremenitvijo obstaja tveganje za sindrom tumorske lize. Te bolnike je treba skrbno nadzirati in zdraviti po najboljši medicinski praksi. Zdravilo Adcetris lahko povzroči periferno nevropatijo, ki je v večini primerov reverzibilna. Bolnike je treba nadzirati glede simptomov nevropatije. Pri bolnikih, pri katerih se pojavi nova ali poslabša obstoječa periferna nevropatija, bo morda treba odmerek zdravila Adcetris odložiti ali zmanjšati ali zdravljenje prekiniti (glejte SmPC). Pri zdravljenju z zdravilom Adcetris se lahko pojavijo anemija, trombocitopenija in nevtropenija 3. ali 4. stopnje. Za prilagoditev odmerka v primeru nevtropenije glejte SmPC. Bolnike je treba skrbno nadzirati glede pojava zvišane telesne temperature. Če se pojavi febrilna nevtropenija, jo je treba zdraviti po najboljši medicinski praksi. Če se pojavi Stevens-Johnsonov sindrom ali toksična epidermalna nekroliza, je treba zdravljenje z zdravilom Adcetris prekiniti in uvesti ustrezno zdravljenje. Poročali so o zvišanih vrednostih ALT in AST. Pri bolnikih, ki prejemajo brentuksimab vedotin, je treba jetrno funkcijo rutinsko spremljati. Pri bolnikih, pri katerih se pojavi hiperglikemija, je treba skrbno spremljati vrednost glukoze v serumu in po potrebi uvesti ustrezno zdravljenje. Zmerna ali huda okvara ledvic in nizke koncentracije albumina v serumu lahko vplivajo na očistek MMAE (glejte SmPC). Zdravilo Adcetris vsebuje največ 2,1 mmol (ali 47 mg) natrija na odmerek. Plodnost, nosečnost in dojenje: Podatkov o uporabi zdravila med nosečnostjo ni. Študije na živalih so pokazale vpliv na sposobnost razmnoževanja. Ženske v rodni dobi morajo med zdravljenjem in še 6 mesecev po njem uporabiti dve obliki učinkovite kontracepcije. Ni podatkov o tem, ali se brentuksimab vedotin ali njegovi presnovki izločajo v materino mleko. Tveganja za dojenega novorojenca/otroka ne moremo izključiti. Plodnost: V predkliničnih študijah je zdravljenje z zdravilom Adcetris povzročilo testikularno toksičnost, lahko pa bi vplivalo tudi na plodnost moških. Medsebojno delovanje z drugimi zdravili in druge oblike interakcij: Pri sočasni uporabi zdravila Adcetris in močnih zaviralcev CYP3A4 in P-gp, kot je ketokonazol, se lahko poveča pojavnost nevtropenije. Pri sočasni uporabi rifampicina, močnega induktorja CYP3A4, se plazemska izpostavljenost zdravilu Adcetris ni spremenila, vendar pa se je izpostavljenost MMAE zmanjšala. Pri sočasni uporabi midazolama, substrata za CYP3A4, se presnova midazolama ni spremenila, zato se ne pričakuje, da bi se pri sočasni uporabi zdravila Adcetris spremenila izpostavljenost zdravilom, ki se presnavljajo z encimi CYP3A4. Neželeni učinki: Zelo pogosti (> 10 %): Okužba, srbenje, mialgija, utrujenost, zvišana telesna temperatura in reakcije, povezane z infundiranjem. Pogosti (. 1/100 do < 1/10): Sepsa/septični šok, okužba zgornjih dihal, herpes zoster, pljučnica, anemija, trombocitopenija, hiperglikemija, periferna motorična nevropatija, omotica, demielinizacijska polinevropatija, kašelj, dispneja, zaprtost, zvišanje vrednosti ALT/AST, izpuščaj, artralgija, bolečina v hrbtu in mrzlica. Občasni (. 1/1.000 do < 1/100): Oralna kandidoza, pljučnica, ki jo povzroča Pneumocystis jiroveci, sta.lokokna bakteriemija, sindrom tumorske lize in akutni pankreatitis. Redki (. 1/10.000 do  1/1.000): StevensJohnsonov sindrom in toksična epidermalna nekroliza. Neznana (pogostnosti iz razpoložljivih podatkov ni mogoče oceniti): Progresivna multifokalna levkoencefalopatija, febrilna nevtropenija in ana. laktična reakcija. Resni neželeni učinki zdravila pri bolnikih v študijah 2. faze so bili: nevtropenija, trombocitopenija, zaprtost, driska, bruhanje, zvišana telesna temperatura, periferna motorična in periferna senzorična nevropatija, hiperglikemija, demielinizacijska polinevropatija, sindrom tumorske lize in Stevens-Johnsonov sindrom. Posebna navodila za shranjevanje: Vialo shranjujte v hladilniku (2 °C-8 °C), zaščiteno pred svetlobo. Po rekonstituciji/redčenju je raztopina dokazano kemično in .zikalno stabilna 24 ur pri temperaturi od 2 °C do 8 °C. Datum revizije besedila: Januar 2015. Način izdajanja zdravila: Predpisovanje in izdaja zdravila je le na recept. Številka dovoljenja za promet z zdravilom: EU/1/12/794/001. Ime in naslov imetnika dovoljenja za promet z zdravilom: Takeda Pharma A/S, Dybendal Alle 10, 2630 Taastrup, Danska. Dodatne informacije so na voljo pri: Takeda GmbH, Podružnica Slovenija, Dalmatinova ul. 2, Ljubljana, tel: 059 082 480. Adcetris® je registrirana blagovna znamka Millennium Pharmaceuticals Inc. Zdravilo Adcetris je v Evropi prejelo pogojno dovoljenje za promet. Pogojno dovoljenje za promet se izda za zdravilo, ki izpolnjuje še neizpolnjene zdravstvene potrebe bolnikov, če korist takojšnje dostopnosti zdravila za interese javnega zdravja odtehta tveganje, ki izhaja iz dejstva, da so potrebni še dodatni podatki. Evropska agencija za zdravila bo vsaj enkrat letno ponovno pregledala nove podatke o zdravilu Adcetris. Če bo potrebno, bo posodobljen tudi povzetek glavnih značilnosti zdravila. Poročali so o resnih okužbah, kot so pljučnica, sta.lokokna bakteriemija, sepsa/ nevtropenija, periferna senzorična nevropatija, driska, navzea, bruhanje, alopecija, Samo za strokovno javnost. instructions Instructions for authors The editorial policy Radiology and Oncology is a multidisciplinary journal devoted to the publishing original and high quality scientific papers and review articles, pertinent to diagnostic and interventional radiology, computerized tomography, magnetic resonance, ultrasound, nuclear medicine, radiotherapy, clinical and experimental oncology, radiobiology, radiophysics and radiation protection. Therefore, the scope of the journal is to cover beside radiology the diagnostic and therapeutic aspects in oncology, which distinguishes it from other journals in the field. The Editorial Board requires that the paper has not been published or submitted for publication elsewhere; the authors are responsible for all statements in their papers. Accepted articles become the property of the journal and, therefore cannot be published elsewhere without the written permission of the editors. Submission of the manuscript The manuscript written in English should be submitted to the journal via online submission system Editorial Manager avail­able for this journal at: www.radioloncol.com. In case of problems, please contact Sašo Trupej at saso.trupej@computing.si or the Editor of this journal at gsersa@onko-i.si All articles are subjected to the editorial review and when the articles are appropriated they are reviewed by independent ref­erees. In the cover letter, which must accompany the article, the authors are requested to suggest 3-4 researchers, competent to review their manuscript. However, please note that this will be treated only as a suggestion; the final selection of reviewers is exclusively the Editor’s decision. The authors’ names are revealed to the referees, but not vice versa. Manuscripts which do not comply with the technical requirements stated herein will be returned to the authors for the cor­rection before peer-review. The editorial board reserves the right to ask authors to make appropriate changes of the contents as well as grammatical and stylistic corrections when necessary. Page charges will be charged for manuscripts exceeding the recommended length, as well as additional editorial work and requests for printed reprints. Articles are published printed and on-line as the open access (www.degruyter.com/view/j/raon). All articles are subject to 700 EUR + VAT publication fee. Exceptionally, waiver of payment may be negotiated with editorial office, upon lack of funds. Manuscripts submitted under multiple authorship are reviewed on the assumption that all listed authors concur in the submission and are responsible for its content; they must have agreed to its publication and have given the corresponding author the authority to act on their behalf in all matters pertaining to publication. The corresponding author is responsible for informing the coauthors of the manuscript status throughout the submission, review, and production process. Preparation of manuscripts Radiology and Oncology will consider manuscripts prepared according to the Uniform Requirements for Manuscripts Submitted to Biomedical Journals by International Committee of Medical Journal Editors (www.icmje.org). The manuscript should be written in grammatically and stylistically correct language. Abbreviations should be avoided. If their use is neces­sary, they should be explained at the first time mentioned. The technical data should conform to the SI system. The manu­script, excluding the references, tables, figures and figure legends, must not exceed 5000 words, and the number of figures and tables is limited to 8. Organize the text so that it includes: Introduction, Materials and methods, Results and Discussion. Exceptionally, the results and discussion can be combined in a single section. Start each section on a new page, and number each page consecutively with Arabic numerals. The Title page should include a concise and informative title, followed by the full name(s) of the author(s); the institutional affiliation of each author; the name and address of the corresponding author (including telephone, fax and E-mail), and an abbreviated title (not exceeding 60 characters). This should be followed by the abstract page, summarizing in less than 250 words the reasons for the study, experimental approach, the major findings (with specific data if possible), and the principal conclusions, and providing 3-6 key words for indexing purposes. Structured abstracts are required. Slovene authors are requested to provide title and the abstract in Slovene language in a separate file. The text of the research article should then proceed as follows: Introduction should summarize the rationale for the study or observation, citing only the essential references and stating the aim of the study. Materials and methods should provide enough information to enable experiments to be repeated. New methods should be described in details. Results should be presented clearly and concisely without repeating the data in the figures and tables. Emphasis should be on clear and precise presentation of results and their significance in relation to the aim of the investigation. Discussion should explain the results rather than simply repeating them and interpret their significance and draw conclu­sions. It should discuss the results of the study in the light of previously published work. instructions Charts, Illustrations, Images and Tables Charts, Illustrations, Images and Tables must be numbered and referred to in the text, with the appropriate location indi­cated. Charts, Illustrations and Images, provided electronically, should be of appropriate quality for good reproduction. Illustrations and charts must be vector image, created in CMYK color space, preferred font “Century Gothic”, and saved as .AI, .EPS or .PDF format. Color charts, illustrations and Images are encouraged, and are published without additional charge. Image size must be 2.000 pixels on the longer side and saved as .JPG (maximum quality) format. In Images, mask the identities of the patients. Tables should be typed double-spaced, with a descriptive title and, if appropriate, units of numeri­cal measurements included in the column heading. The files with the figures and tables can be uploaded as separate files. References References must be numbered in the order in which they appear in the text and their corresponding numbers quoted in the text. Authors are responsible for the accuracy of their references. References to the Abstracts and Letters to the Editor must be identified as such. Citation of papers in preparation or submitted for publication, unpublished observations, and personal communications should not be included in the reference list. If essential, such material may be incorporated in the appropri­ate place in the text. References follow the style of Index Medicus. All authors should be listed when their number does not exceed six; when there are seven or more authors, the first six listed are followed by “et al.”. The following are some examples of references from articles, books and book chapters: Dent RAG, Cole P. In vitro maturation of monocytes in squamous carcinoma of the lung. Br J Cancer 1981; 43: 486-95. Chapman S, Nakielny R. A guide to radiological procedures. London: Bailliere Tindall; 1986. Evans R, Alexander P. Mechanisms of extracellular killing of nucleated mammalian cells by macrophages. In: Nelson DS, editor. Immunobiology of macrophage. New York: Academic Press; 1976. p. 45-74. Authorization for the use of human subjects or experimental animals When reporting experiments on human subjects, authors should state whether the procedures followed the Helsinki Declaration. Patients have the right to privacy; therefore the identifying information (patient’s names, hospital unit num­bers) should not be published unless it is essential. In such cases the patient’s informed consent for publication is needed, and should appear as an appropriate statement in the article. Institutional approval and Clinical Trial registration number is required. The research using animal subjects should be conducted according to the EU Directive 2010/63/EU and following the Guidelines for the welfare and use of animals in cancer research (Br J Cancer 2010; 102: 1555 – 77). Authors must state the committee approving the experiments, and must confirm that all experiments were performed in accordance with relevant regulations. These statements should appear in the Materials and methods section (or for contributions without this section, within the main text or in the captions of relevant figures or tables). Transfer of copyright agreement For the publication of accepted articles, authors are required to send the License to Publish to the publisher on the address of the editorial office. A properly completed License to Publish, signed by the Corresponding Author on behalf of all the authors, must be provided for each submitted manuscript. The non-commercial use of each article will be governed by the Creative Commons Attribution-NonCommercial-NoDerivs license. Conflict of interest When the manuscript is submitted for publication, the authors are expected to disclose any relationship that might pose real, apparent or potential conflict of interest with respect to the results reported in that manuscript. Potential conflicts of interest include not only financial relationships but also other, non-financial relationships. In the Acknowledgement section the source of funding support should be mentioned. The Editors will make effort to ensure that conflicts of interest will not compromise the evaluation process of the submitted manuscripts; potential editors and reviewers will exempt themselves from review process when such conflict of interest exists. The statement of disclosure must be in the Cover letter accompany­ing the manuscript or submitted on the form available on www.icmje.org/coi_disclosure.pdf Page proofs Page proofs will be sent by E-mail to the corresponding author. It is their responsibility to check the proofs carefully and return a list of essential corrections to the editorial office within three days of receipt. Only grammatical corrections are ac­ceptable at that time. Open access Papers are published electronically as open access on www.degruyter.com/view/j/raon, also papers accepted for publication as E-ahead of print. BISTVENI PODATKI IZ POVZETKA GLAVNIH ZNAČILNOSTI ZDRAVILA SUTENT 12,5 mg, 25 mg, 37,5 mg, 50 mg trde kapsule Sestava in oblika zdravila: Ena kapsula vsebuje 12,5 mg, 25 mg, 37,5 mg ali 50 mg sunitiniba (v obliki sunitinibijevega malata). Indikacije: Zdravljenje neizrezljivega in/ali metastatskega malignega gastrointestinalnega stromalnega tumorja (GIST) pri odraslih, če zdravljenje z imatinibom zaradi odpornosti ali neprenašanja ni bilo uspešno. Zdravljenje napredovalega/metastatskega karcinoma ledvičnih celic (MRCC) pri odraslih. Zdravljenje neizrezljivih ali metastatskih, dobro diferenciranih nevroendokrinih tumorjev trebušne slinavke (pNET), kadar gre za napredovanje bolezni pri odraslih (izkušnje z zdravilom Sutent kot zdravilom prve izbire so omejene). Odmerjanje in način uporabe: Terapijo mora uvesti zdravnik, ki ima izkušnje z uporabo zdravil za zdravljenje rakavih bolezni. GIST in MRCC: Priporočeni odmerek je 50 mg peroralno enkrat na dan, 4 tedne zapored; temu sledi 2-tedenski premor (Shema 4/2), tako da celotni ciklus traja 6 tednov. pNET: Priporočeni odmerek je 37,5 mg peroralno enkrat na dan, brez načrtovanega premora. Prilagajanje odmerka: Odmerek je mogoče prilagajati v povečanjih po 12,5 mg, upoštevaje individualno varnost in prenašanje. Pri GIST in MRCC dnevni odmerek ne sme preseči 75 mg in ne sme biti manjši od 25 mg; pri pNET je največji odmerek 50 mg na dan, z možnimi prekinitvami zdravljenja. Pri sočasni uporabi z močnimi zaviralci ali induktorji CYP3A4 je treba odmerek ustrezno prilagoditi. Pediatrična populacija: Uporaba sunitiniba ni priporočljiva. Starejši bolniki (. 65 let): Med starejšimi in mlajšimi bolniki niso opazili pomembnih razlik v varnosti in učinkovitosti. Okvara jeter: Pri bolnikih z jetrno okvaro razreda A in B po Child-Pughu prilagoditev odmerka ni potrebna; pri bolnikih z okvaro razreda C sunitinib ni bil preizkušen, zato njegova uporaba ni priporočljiva. Okvara ledvic: Prilagajanje začetnega odmerka ni potrebno, nadaljnje prilagajanje odmerka naj temelji na varnosti in prenašanju pri posameznem bolniku. Način uporabe: Zdravilo Sutent se uporablja peroralno, bolnik ga lahko vzame s hrano ali brez nje. Če pozabi vzeti odmerek, ne sme dobiti dodatnega, temveč naj vzame običajni predpisani odmerek naslednji dan. Kontraindikacije: Preobčutljivost na zdravilno učinkovino ali katerokoli pomožno snov. Posebna opozorila in previdnostni ukrepi: Bolezni kože in tkiv: obarvanje kože, gangrenozna pioderma (običajno izgine po prekinitvi zdravljenja), hude kožne reakcije (multiformni eritem (EM), Stevens-Johnsonov sindrom (SJS) in toksična epidermalna nekroliza (TEN)). Če so prisotni znaki EM, SJS ali TEN, je treba zdravljenje prekiniti. Krvavitve v prebavilih, dihalih, sečilih, možganih; najpogosteje epistaksa; krvavitve tumorja, včasih s smrtnim izidom. Pri bolnikih, ki se sočasno zdravijo z antikoagulanti, se lahko redno spremlja celotna krvna slika (trombociti), koagulacijski faktorji (PT / INR) in opravi telesni pregled. Bolezni prebavil: polegdiareje, navzee/bruhanja, bolečine v trebuhu, dispepsije, stomatitisa/bolečine v ustih in ezofagitisa tudi hudi zapleti (včasih s smrtnim izidom), vključno z gastrointestinalno perforacijo. Hipertenzija: pri bolnikih s hudo hipertenzijo, ki je ni mogoče urediti z zdravili, je priporočljivo začasno prenehanje zdravljenja. Hematološke bolezni: zmanjšanje števila nevtro.ilcev, trombocitov, anemija. Bolezni srca in ožilja: srčno-žilni dogodki, vključno s srčnim popuščanjem, kardiomiopatijo, miokardno ishemijo in miokardnim infarktom, v nekaterih primerih s smrtnim izidom; sunitinib povečuje tveganje za pojav kardiomiopatije; previdna uporaba pri bolnikih s tveganjem za te dogodke, ali ki so te dogodke imeli v preteklosti. Podaljšanje intervala QT: previdna uporaba pri bolnikih z znano anamnezo podaljšanja intervala QT, tistih, ki jemljejo antiaritmike ali zdravila, ki lahko podaljšajo interval QT, in tistih z relevantno, že obstoječo srčno boleznijo, bradikardijo ali elektrolitskimi motnjami. Venski in arterijski trombembolični dogodki; arterijski včasih s smrtnim izidom. Trombotična mikroangiopatija (TMA): TMA, vključno s trombotično trombocitopenično purpuro in hemolitično-uremičnim sindromom, v nekaterih primerih z odpovedjo ledvic ali smrtnim izidom. Dogodki na dihalih: dispneja, plevralni izliv, pljučna embolija ali pljučni edem; redki primeri s smrtnim izidom. Moteno delovanje ščitnice: bolnike je treba med zdravljenjem rutinsko spremljati glede delovanja ščitnice vsake 3 mesece. Pankreatitis, tudi resni primeri s smrtnim izidom. Hepatotoksičnost, nekateri primeri s smrtnim izidom. Holecistitis, vključno z akalkuloznim in em.izemskim holecistitisom. Delovanje ledvic: primeri zmanjšanega delovanja ledvic, odpovedi ledvic in/ali akutne odpovedi ledvic, v nekaterih primerih s smrtnim izidom. Fistula: če nastane .istula, je treba zdravljenje s sunitinibom prekiniti. Oteženo celjenje ran: pribolnikih, pri katerih naj bi bil opravljen večji kirurški poseg, je priporočljiva začasna prekinitev zdravljenja s sunitinibom. Osteonekroza čeljustnic: pri sočasnem ali zaporednem dajanju zdravila Sutent in intravenskih bisfosfonatov je potrebna previdnost; invazivni zobozdravstveni posegi predstavljajo dodatni dejavnik tveganja. Preobčutljivost/angioedem. Motnje okušanja. Konvulzije: obstajajo poročila, nekatera s smrtnim izidom, o preiskovancih s konvulzijami in radiološkimi znaki sindroma reverzibilne posteriorne levkoencefalopatije. Sindrom lize tumorja, v nekaterih primerih s smrtnim izidom. Okužbe: hude okužbe z ali brez nevtropenije (okužbe dihal, sečil, kože in sepsa), vključno z nekaterimi s smrtnim izidom; redki primeri nekrotizitajočega fasciitisa, vključno s prizadetostjo presredka, ki so bili včasih smrtni. Hipoglikemija: če se pojavi simptomatska hipoglikemija, je treba zdravljenje s sunitinibom začasno prekiniti. Pri sladkornih bolnikih je treba redno preverjati raven glukoze v krvi in, če je treba, prilagoditi odmerek antidiabetika. Medsebojno delovanje z drugimi zdravili: (Študije so izvedli le pri odraslih.) Zdravila, ki lahko zvečajo koncentracijo sunitiniba v plazmi (ketokonazol, ritonavir, itrakonazol, eritromicin, klaritromicin ali sok grenivke). Zdravila, ki lahko zmanjšajo koncentracijo sunitiniba v plazmi (deksametazon, fenitoin, karbamazepin, rifampin, fenobarbital, Hypericum perforatum oz. šentjanževka). Plodnost, nosečnost in dojenje: Zdravila Sutent ne smemo uporabljati med nosečnostjo in tudi ne pri ženskah, ki ne uporabljajo ustrezne kontracepcije, razen če možna korist odtehta možno tveganje za plod. Ženske v rodni dobi naj med zdravljenjem z zdravilom Sutent ne zanosijo. Ženske, ki jemljejo zdravilo Sutent, ne smejo dojiti. Neklinični izsledki kažejo, da lahko zdravljenje s sunitinibom poslabša plodnost samcev in samic. Vpliv na sposobnost vožnje in upravljanja s stroji: Sutent lahko povzroči omotico. Neželeni učinki: Najbolj resni neželeni učinki (nekateri s smrtnim izidom) so: odpoved ledvic, srčno popuščanje, pljučna embolija, gastrointestinalna perforacija in krvavitve (npr. v dihalih, prebavilih, tumorju, sečilih in možganih). Najpogostejši neželeni učinki (ki so se pojavili pri vsaj 20 % bolnikov v registracijskih preskušanjih) so: zmanjšan tek, motnje okušanja, hipertenzija, utrujenost, prebavne motnje (npr. driska, navzea, stomatitis, dispepsija in bruhanje), sprememba barve kože in sindrom palmarno-plantarne eritrodisestezije. Med najbolj pogostimi neželenimi učinki so tudi hematološke motnje (nevtropenija, trombocitopenija, anemija in levkopenija). Ostali zelo pogosti (. 1/10) neželeni učinki so: hipotiroidizem, nespečnost, omotica, glavobol, dispneja, epistaksa, kašelj, bolečina v trebuhu, zaprtje, obarvanje kože, izpuščaj, spremembe barve las, suha koža, bolečine v udih, artralgija, bolečine v hrbtu, vnetje sluznice, edem, pireksija. Način in režim izdaje: Predpisovanje in izdaja zdravila je le na recept, zdravilo pa se uporablja samo v bolnišnicah. Izjemoma se lahko uporablja pri nadaljevanju zdravljenja na domu ob odpustu iz bolnišnice in nadaljnjem zdravljenju. Imetnik dovoljenja za promet:P.izer Limited, Ramsgate Road, Sandwich, Kent, CT13 9NJ, Velika Britanija. Datum zadnje revizije besedila: 25.06.2015 Pred predpisovanjem se seznanite s celotnim povzetkom glavnih značilnosti zdravila. SUT-03-15 Samo za strokovno javnost