Medical Care |

Medical Care

##SEVER##

/r/radium.no1.html

 

Euroboss

Scandinavian Sarcoma Group and
Oncologic Center, Lund, Sweden
Euroboss I
A European treatment protocol for bone sarcoma
in patients older than 40 years
February 1, 2003
Euroboss I Scandinavian Sarcoma Group& Oncologic centerLund, Sweden EUROBOSS I
A European treatment protocol for bone sarcoma in
patients older than 40 years

This document describes a collaborative study in patients aged 41 to 65 years with high-grade bonesarcoma and provides information for entering patients into the protocol. The trial committee does notintend it to be used as an aide-memoire or guide for treatment of non-registered patients. Protocolamendments may be necessary; these will be circulated to known participants in the trial, but institutionsentering patients are advised to contact the appropriate study centers to confirm the correctness of theprotocol in their possession. Before entering patients clinicians must ensure that the study protocol isapproved by their respective ethical committee.
Due to their rarity, the treatment of the tumors object of the present study is recommended only incenters experienced in bone sarcomas.
Euroboss I The trial EUROBOSS I is a multicenter prospective study for patients older than 40 years with high-grade bone sarcoma. Because of the low incidence of bone sarcomas, multiinstitutional collaboration isessential to address questions concerning diagnosis and treatment. The current participating intergroups(ISG, COSS and SSG) cover a population of approximately 200 million people, and together theseorganisations claim to provide an adequate patient volume to explore the aims of the current study. Thetrial is open for participation by other internationally recognized osteosarcoma groups or institutionsprovided agreement from the other groups.
The present EUROBOSS I protocol is based on the experience of the participating intergroups in thetreatment of bone sarcomas and their past and present osteosarcoma protocols.
A working committee consisting of the three intergroup coordinators (Stefano Ferrari, Stefan Bielackand Sigbjørn Smeland) have initiated this protocol: Protocol chairman and
ISG coordinator:

Sigbjørn Smeland Dept. of Chemotherapy Dept. of Oncology Dept. of Pediatric Oncology Istituto Ortopedico Rizzoli Norwegian Radium Hospital University Hospital of Münster Sezione di Chemioterapia. Istituto Ortopedico Rizzoli, via Pupilli 1, 40136 Bologna, Italy.
Phone: +39-051-636 61 99, Fax: +39-051-636 62 77E-mail: [email protected].
Data managing: Annalisa Nobile Scandinavian Sarcoma Group (SSG)
SSG coordinator: Sigbjørn Smeland
SSG Secretariat: Regional Tumor Registry, Lund University Hospital , SE-221 85 Lund, Sweden.
Phone: +46-46-17 75 55, Fax: +46-46-18 81 43
E-mail: [email protected]
Data managing: Evy Nilsson
Cooperative Osteosarcoma Study Group (COSS)
COSS coordinator: Stefan Bielack
COSS secretariat: Cooperative Osteosarkomstudiengruppe (COSS), Universitätsklinikum Münster,
Klinik und Poliklinik für Kinderheilkunde-Pädiatrische Hämatologie/Onkologie, Albert Schweitzer-
Str. 33, D-48149 Münster, Germany.
Phone: +49-251-835 24 24, Fax: +49-251-835 64 89
E-mail: [email protected]
Italian Sarcoma Group (ISG)
Protocol chairman and ISG coordinator: Stefano Ferrari
ISG secretariat: Sezione di Chemioterapia. Istituto Ortopedico Rizzoli, via Pupilli 1, 40136 Bologna,
Italy. Phone: +39-051-636 61 99, Fax: +39-051-636 62 77
E-mail: [email protected].
Euroboss I Scandinavian Sarcoma Group
The Scandinavian countries (Denmark, Finland, Iceland, Norway and Sweden) have a total popula-tion of about 25 million. They possess similar social structures, a modern medical service covering allinhabitants, as well as an effective registration system for all cancer patients. This serves as a good basisfor cooperation. Accordingly, the Scandinavian Sarcoma Group was founded in 1979 with the aim toimprove the prognosis for sarcoma patients within Scandinavia. Its work has led to a systematic orga-nisation of sarcoma treatment, with guidelines for diagnosis, pathology, and treatment now generallyaccepted by all tumor centers in Scandinavia.
The first Scandinavian non-randomised neo-adjuvant chemotherapy trial for high-grade osteosarcomalocalised to the extremities, SSG II, was based on the Rosen T-10 protocol and was carried out during1982–1989. The protocol represented a breakthrough in the treatment of osteosarcoma in Scandinaviawith a > 40% improvement in outcome compared to historical controls. The second osteosarcoma trial(SSG VIII), using a more aggressive preoperative combination chemotherapy, high-dose methotrexate,doxorubicin and cisplatinum, opened in 1990 and was closed in 1997. The outcome analysis of theSSG VIII study shows a further improvement of 10% in long-time survival. The first joint Italian/Scandinavian study, ISG/SSG I, activated March 1997, was undertaken to explore the effect of maxi-mum dose-intensive chemotherapy including high-dose ifosfamide scheduled for all patients. The protocolwas closed by September 2000 and replaced by the currently ongoing SSG XIV protocol.
The SSG version of the EUROBOSS I protocol has been completed by the following five SSG
members (here defined as intergroup members):

Dr. Sigbjørn Smeland (chairman) Dr. Øyvind Bruland Dr. Carl Blomqvist Dept. of Oncology Dept. of Oncology Norwegian Radium Hospital Norwegian Radium Hospital University Hospital Dr. Mikael Eriksson Dr. Thor A. Alvegård Dept. of Oncology Dept. of Oncologic Center University Hospital University Hospital Printing and distribution of the final protocol will be arranged by the Oncologic Center in Lund.
The EUROBOSS I protocol will be activated by February 2003.
Lund, January 2003 Euroboss I Organization of the Scandinavian Sarcoma Group (SSG)
Scandinavian Sarcoma Group Secretariat Southern Swedish Regional Tumor Registry Lund University Hospital Phone: +46-46-17 75 55, Fax: +46-46-18 81 43 E-mail SSG: [email protected] Secretary: Evy Nilsson T.A. Alvegård, Lund Publication ombudsman C. Blomqvist, Helsinki
H. Bauer, Stockholm P. Lindholm, Turku T.A. Alvegård, Lund T. Böhling, Helsinki H. Bauer, Stockholm K. Sundby Hall, Oslo C. Blomqvist, Helsinki A. Rydholm , Lund L.G. Kindblom, Gothenburg T. A. Alvegård, Lund U. Nilsonne, Stockholm K. Sundby Hall, Oslo H. Anderson, LundR. Perfekt, Lund U. Nilsonne, Stockholm L. Angervall, Gothenburg Ø. Solheim, Oslo Diagnostic radiology and
H. Bauer, Stockholm L.G. Kindblom, Gothenburg Ö. Berlin, Gothenburg L. Hardell, Örebro P. Gustafson, Lund V. Söderlund, Stockholm J. Wejde, Stockholm C. Trovik, Bergen M. Winderen, Oslo B. Bjerkehagen, Oslo R. Huuhtanen, Helsinki H. Willén, GothenburgT. Böhling, HelsinkiH. Domanski, Lund G. Follerås, Oslo R. Wedin, Stockholm S. Knuutila, Helsinki O. Brosjö, Stockholm F. Mertens, Lund
Clinical Pharmacology
C. Blomqvist, HelsinkiCoordinatorI. Turesson, Uppsala Ø. Bruland, Oslo Past and present
O. Brosjö, Stockholm T. Böhling, Ø. Bruland, Oslo T.A. Alvegård, Lund T. Holmström, Helsinki SSG II, SSG VIII, L.G. Kindblom, Gothenburg O. Björk, Stockholm SSG XIV, EUROBOSS I
Ewing's sarcoma I. Elomaa, Helsinki O. Brosjö, Stockholm M. Åkerman, Lund C. Blomqvist, Helsinki T. Wiklund, Helsinki T. Wiebe, LundM. Nilbert, LundL. Hjorth, Lund Localized soft tissue sarcoma T.A. Alvegård, Lund L. Angervall, Gothenburg C. Blomqvist, Helsinki SSG I, SSG XIII
I. Turesson, Uppsala L.G. Kindblom, Gothenburg I. Turesson, Uppsala Advanced soft tissue sarcoma T.A. Alvegård, Lund L.G. Kindblom, Gothenburg C. Blomqvist, Helsinki SSG X, SSG XII, SSG XV K. Sundby Hall, Oslo G. Follerås, Oslo B. Bjerkehagen, Oslo I. Turesson, Uppsala SSG Register H. Bauer, Stockholm L.G. Kindblom, Gothenburg P. Gustafson, Lund J. Wejde, Stockholm Euroboss I I. TREATMENT PROTOCOL
Preface . 3Organization of the Scandinavian Sarcoma Group . 5 Treatment schedule . 8 Introduction . 11 Aims and general protocol design . 12 Evaluation criteria and estimated patient number . 13 Data collection . 14 "Resource Group" . 14 Ethical considerations . 15 Criteriae for eligibility . 16 Criteriae for exclusion . 16 Pretreatment investigations and follow-up . 16 Chemotherapy administration . 18 13.1 General consideration . 18 13.3 Cisplatinum . 19 13.4 Doxorubicin . 20 13.5 Ifosfamide . 20 13.6 Methotrexate . 21 Dose adjustments . 22 Serious adverse events, stopping rules . 22 Participating centers within SSG . 28 Patient information . 29 Reporting of serious adverse events . 30 Management of methotrexate toxicity and delayed methotrexate excretion . 30 Calculation of renal tubular reabsorption of phosphate – TmP/GFR . 32 WHO performance status . 33 Associated research projects . 33 Guidelines for pathology . 36 Guidelines for surgery . 39 Guidelines for radiotherapy . 41 Euroboss I Pathology report I Pathology report II Radiation therapy Form 7A: Preoperative chemotherapy or postoperative chemotherapy with good histopathology response: Chemotherapy flow-sheet Form 7B: Chemotherapy toxicity flow-sheet Form 8A: Postoperative chemotherapy with poor histological response: Chemotherapy flow-sheet Form 8B: Chemotherapy toxicity flow-sheet Form 10: Serious adverse event report Euroboss I
A European treatment protocol for bone sarcoma in patients older than 40 years.
100 mg/m2, 48 hours iv 60 mg/m2 , 24 hours iv Ifosfamide 3 000 mg/m2/day, 2 days iv Euroboss I
A European treatment protocol for bone sarcoma in patients older than 40 years.
100 mg/m2, 48 hours iv 60 mg/m2 , 24 hours iv 3 000 mg/m2/day, 2 days iv 8 000 mg/m2, 4 hours iv Euroboss I Euroboss I Wide surgical removal of the neoplastic lesion combined with adjuvant or neoadjuvant chemotherapy iscurrently considered the "standard" in the management of patients with osteosarcoma (1–3).
Methotrexate (MTX), doxorubicin (DOX), cisplatin (CDP), and ifosfamide (IFO) are the four drugswith proven efficacy against osteosarcoma that have been used according to different schedules inchemotherapy protocols adopted in large monocentric and multicentric studies (4–9). Since the incidenceof osteosarcoma peaks in the second decade of life (2), the studies reported in the literature usuallyinvolve patients younger than 40 years of age treated with dose intensive chemotherapy protocols(4–9).
Only a few studies contain data on the use of chemotherapy in patients older than 40 years with highgrade osteosarcoma (10–16). In the retrospective EMSOS study (15), forms of 486 patients from 13different centers were evaluated. Here it was shown that secondary osteosarcoma (post radiationosteosarcoma and Paget's osteosarcoma), as well as patients older than 60 years have a very poorprognosis. Moreover, it was shown an advantage in terms of prognosis for those patients who receivedsome kind of chemotherapy. Unfortunately, the study did not provide details on the differentchemotherapy protocols used. In the COSS experience (16), 54 osteosarcoma patients 40–68 yearsold were more likely to present with axial tumor, secondary osteosarcomas, a prolonged history ofsymptoms compared to their younger counterparts. Also they were more likely to experience a delayedstart ot treatment. An age of 40 years or older was associated with inferior overall and event-freesurvival probabilities (55% and 42% at 5 years and 42% and 37% at 10 years). In multivariate analy-sis, however, age was not a prognostic factor, rather, the poor outcome seems to be due to the predilectionfor unfavourable sites. In fact, the corresponding survival figures for the >40 year old patients withextremity tumors (including both localized and primary metastatic tumors) were 59% and 50% at 5years and 56% and 32% at 10 years. An Italian study (14) reported the results obtained in a selectedpopulation of 29 patients aged 40–60 years with non-metastatic osteosarcoma of the extremity. Theywere treated with adequate surgery and adjuvant chemotherapy with DOX, CDP and IFO obtainingan 8-years overall survival of 62%. The remaining studies (10–13) gave few data on this topic.
The proven efficacy of the chemotherapy regimens in children and adolescents with osteosarcomawould suggest the use of the same antineoplastic drugs also in older patients. Among the latter the useof the dose intensive chemotherapy protocols may however be complicated by concomitant diseases(co-morbidity). Furthermore, physiological changes occur with aging, including decreased cardiovascularperformance, decreased haematopoietic tissue activity as well as decreased renal function withimplications for drug toxicity (17). On the other hand, age itself can not be considered a sufficientparameter to exclude patients from chemotherapy but "exclusions should be based on physiologicfunctional parameters, such as measures of renal, liver, and marrow function, or performance status"(18–19).
The vast majority of the osteosarcoma chemotherapy protocols are specifically planned for adolescentsand children. Dose and schedule adjustments are to be made in planning chemotherapy protocols forolder patients, especially for renally excreted agents, and for cardiotoxic drugs, and a strict hemopoieticsupport with hemopoietic growth factors is recommended (19–20).
Due to their rarity, a "standard" treatment option is not established for the non-osteogenic, non Ewing'stumors of bone: Fibrosarcoma, Malignant Fibrous Histiocytoma (MFH), Leiomyosarcoma,Dedifferentiated Chondrosarcoma, Angiosarcoma. The wide surgical removal of the primary lesion isthe cornerstone of the treatment also for these tumors (1), but the role played by the chemotherapy, andthe antineoplastic drugs to be used is still under discussion.
Euroboss I A similar chemosensitivity, and similar ultrastructural characteristics have been reported for MFH,fibrosarcoma and leiomyosarcoma suggesting that these tumors can be effectively treated with multidrugchemotherapy protocols as for osteosarcoma based on cisplatin (CDP), doxorubicin (DOX), ifosfamide(IFO), and methotrexate (MTX) (21–27).
Chondrosarcoma is not a chemosensitive tumor (1), but a high-grade non-cartilaginous sarcoma candevelop within a pre-existing chondrosarcoma (2). The dedifferentiated component usually shows thecharacteristics of MFH or osteosarcoma with a more aggressive and malignant behaviour than that ofthe cartilaginous component (2). Despite a wide surgical resection, distant metastases develop. Scantdata come from the literature on the type of chemotherapy used, but usually the protocols were basedon drugs active against the dedifferentiated component (28).
The rarity of high-grade bone sarcomas requires collaborative trials in order to establish the mosteffective drugs, the dose, and the duration of chemotherapy treatment. For osteosarcoma and MFHwe have substantial data on the key role of the chemotherapy in the management of these tumors(especially in patients younger than 40). Little is known about the activity of chemotherapy in patientswith rarer histologic subtypes (fibrosarcoma, leiomyosarcoma, angiosarcoma of bone, dedifferentiatedchondrosarcoma), also characterized by an aggressive metastatic behaviour. In the current trial, allpatients are scheduled to receive similar treatment with the same chemotherapy regimen. With a com-mon treatment for all histological subtypes, in addition to obtain general information of efficacy andfeasibility, the protocol opens for separate sub-analyses according to the different histologic categoriesincluded.
3. Aims and general protocol design
The present study is a first step of a process to establish the standard chemotherapy treatment with theaim to improve outcome for patients with these rare tumors. For this reason the study will be a non-controlled clinical trial.
In this regard, the study aims to determine the feasibility of intensive chemotherapy in this age group,and/or separate efficacy analyses according to the different histologic categories and whether the numberof patients recruited by the cooperating groups permits future randomized studies.
Primary aim
To evaluate clinical outcome and chemotherapy-related toxicity in patients 41–65 years old with high-
grade bone sarcoma treated with a three drug chemotherapy regimen containing adriamycin (DOX),
cisplatin (CDP) and ifosfamide (IFO), and the addition of methotrexate (MTX) to poor histologic
responders.
Secondary aims
To evaluate the histologic response to preoperative chemotherapy based on DOX, CDP and IFO in
high-grade bone sarcomas in patients 41–65 years, and to evaluate the prognostic significance of this
histologic response.
Treatment strategy
Wide surgical removal of the tumor with the addition of a systemic treatment based on the antineoplastic
drugs active against osteosarcoma (DOX, CDP, IFO, MTX). The indication for radiotherapy will be
for patients with unresectable tumors. It is recommended in patients who underwent inadequate surgical
removal of the tumor. The addition of radiation therapy can not compensate for an inadequate surgical
treatment.
Euroboss I All patients eligible for the study will receive the planned systemic treatment. Depending on clinical
features, and feasibility of adequate surgical removal of the tumor, patients may receive primary
chemotherapy followed by a postoperative chemotherapy treatment or only an adjuvant chemotherapy.
In case of immediate surgery, patients will receive an adjuvant treatment with the 3-drugs regimen
(CDP-DOX-IFO). In patients who will receive primary chemotherapy, the histologic response will be
evaluated. The evaluation of the histologic response will be performed in referral centers.
Each group will indicate the different referral centers for the pathology.
Following grading
systems are allowed: Huvos system, SSG (30), Salzer-Kuntschik system, COSS (31), and percentage
of necrosis system, ISG (32).
For the patients with a histologic response graded Huvos I, Salzer-Kuntschik 5–6 or less than 50%necrosis, MTX will be added in the postoperative phase in case of adequate glomerular function (definedas creatinine clearance > 70 ml/min).
Start of the protocol
The protocol will be active after approval by the appropriate ethic's committe of at least two of the
participating groups.
Withdrawal
In case of withdrawal of two groups the protocol will be closed early.
4. Evaluation criteria and estimated patient number
Clinical outcome
Date of study entry: date of the diagnostic biopsy
Event-free survival (EFS): calculated from the date of study entry to the date of first adverse event(distant or local recurrence, secondary malignancy or treatment related death) or last follow-up. Patientswho never achieve a complete surgical remission have to be considered as an event on day 1 frombiopsy.
Progression-free survival (PFS): calculated from the date of study entry to the date of tumor pro-gression or last follow-up. (Clinical and/or radiological suspicion of local progression before surgery isnot considered as progression) Disease-free survival (DFS): calculated from the date of surgery of primary lesion and metastases, ifpresent, to the date of distant or local recurrence or last follow-up.
Metastasis-free survival (MFS): calculated from the date of surgery of primary lesion and metasta-ses, if present, to the date of distant recurrence or last follow-up.
Overall survival (OS): calculated from the date of study entry to the date of death or last follow-up.
Chemotherapy toxicity: chemotherapy toxicity will be graded and recorded according to NCIexpanded common toxicity criteria (version 2.0-March 1998) (33).
Estimated number of patients: Patients will be enrolled over a three-year period for an estimated
number of patient/year of 45.
Pathologic review: A panel of pathologists from the participating groups will review the histopathologic
sections of biopsies and of the resected tumors.
Euroboss I 5. Data collection
Common database
A common database will be created at the intergroup secretariat (Sezione di Chemioterapia-Rizzoli;
Bologna, Italy), and reports will be sent to the various group secretariats to collect all the information
(see enclosed list of data required) required from all the participating centres.
Each group secretariat will enter the data into the Excel database and will send files with new patientsand updated cases to the intergroup secretariat every six months.
The Intergroup secretariat will paste data in the common Excel database and will send this updated fileto all the group secretariats.
Case Report Form
Each group will prepare CRFs reporting data on the patients enrolled according to their standards.
The only requirement of forms is to contain the informations requested for the common database.
Data relating to EUROBOSS I must not be reported or published without prior consultation with thestudy chairmen. Any publication arising from the trials will have as its authors those who have producedthe paper and acknowledgement to the intergroup members.
A final report of EUROBOSS I will be provided within 5 years after the completion of the projectedpatient accrual.
7. "Resource groups"
In a multicenter study employing aggressive poly-drug chemotherapy as an integrated part of a multi-disciplinary treatment, unforeseen situations and complications that may not be sufficiently covered inthe protocol are anticipated. In an attempt to minimize protocol violations and to ensure uniform hand-ling of such situations, the participating groups have formed their "Resource Group". Its task is to aideach treating physician to solve these problems. In the event of a problem, the clinician should contacta member of the resource group from his own country who, in turn, will assist either directly or arrangea telephone conference with some or all members of the group. Chemotherapy problems should besolved within 24–48 hours, whereas surgical problems may require consultation with X-rays, etc.
Written documentation regarding the problem's nature and solution should be sent to the clinician inquestion, to all members of the resource group, and should be included in the patient's file at the studysecretariat.
In the case of a serious adverse event the study secretariat will forward the incoming report from theresponsible physician to all the appropriate members of the "Resource group".
Euroboss I Members of the SSG Resource Group:
Dr. Mikael Eriksson Dr. Carl Blomqvist Dept. of Oncology Dept. of Oncology Dept. of Oncology Norwegian Radium Hospital Lund University Hospital University Central Hospital Tel +46-46-17 75 20 Tel +47-22-93 40 00 Fax +46-46-18 81 43 Fax +47-22-93 45 53 Fax +358-9-471 731 81 Surgery:
Dr. Otte Brosjö
Dr. Gunnar Follerås Dr. Aarne Kivioja Dept. of Orthopedics Dept. of Orthopedics Karolinska Hospital Norwegian Radium Hospital University Central Hospital SE-171 76 Stockholm Tel +46-8-517 700 00 Fax +46-8-517 746 99 Tel +47-22-93 40 00 Fax +47-22-93 45 53 Fax +358-9-471 874 81E-mail: [email protected] 8. Ethical considerations
1. EUROBOSS I is a non-randomized phase II study based on the experience from previous osteosarcoma protocols of the participating intergroups (ISG, COSS, SSG) and the experiencefrom recent medical literature.
2. Before the start of treatment, the patients will receive written and oral information about the nature of the disease, the treatment plan, its benefits and the side effects, according to the standardprocedures in each country. Written informed concent is mandatory before inclusion.
3. The outcome and side effects of the treatment will be recorded and reported in the international 4. The physician responsible for the individual patient may deviate from the protocol or may terminate treatment for various medical reasons on medical indications. The EUROBOSS "Resource Group"of specialists is established to assist in such situations.
5. Before entering patients clinicians must ensure that the study protocol has received clearance from their ethical committee.
Euroboss I 9. Criteriae for eligibility
1. Histologically proven diagnosis of high-grade sarcoma of bone of any site and stage 2. Any of the following histological types: osteosarcoma (high-grade surface, central primary and secondary), fibrosarcoma, malignant fibrous histiocytoma, leiomyosarcoma, dedifferentiatedchondrosarcoma, angiosarcoma 3. Age: 41–65 years 4. Normal bone marrow, hepatic, cardiac and renal function 5. Absence of contraindications to the use of cisplatin, doxorubicin and ifosfamide 6. Written informed consent 10. Criteriae for exclusion
1. Planned chemotherapy and/or follow-up not feasible 2. Previous chemotherapy treatment, which contraindicates the use of one or more drugs, included in the present procol 3. Previous chemotherapy treatment for the current tumor 4. White blood count <3.0 × 109/L, and platelets <100 × 109/L 5. Creatinine clearance <70 ml/min 6. Left ventricular ejection fraction <55% or fractional shortening rate of the left ventricle <28% 7. Serum transaminases and bilirubin >2 times the normal values 8. ECOG performance status >2 9. Chondrosarcoma or small/round cell bone sarcoma including mesenchymal chondrosarcoma and Ewing's family tumors 11. Pretreatment investigations and follow-up
1. Medical history and physical examination 2. Complete blood count 3. Serum creatinine, Na, K, urine analysis, GFR, TmP/GFR, bilirubin, transaminases, alkaline phosphatase (optional bone specific alkaline phosphatase), LDH Euroboss I 6. Estimation of left ventricular ejection fraction (LVEF) with cardiac ultrasound or radionuclide ventriculography (MUGA) before first course of doxorubicin treatment 7. Conventional X-rays of the primary lesion 8. CT and MRI scan of the primary lesion 9. Total bone scan 10. CT scan of the chest 11. Additional investigations when clinically required 1. Physical examination 2. Complete blood count 3. Serum creatinine, Na, K, Mg., GFR, TmP/GFR, bilirubin, transaminases, alkaline phosphatase (optional bone specific alkaline phosphatase), LDH 4. Conventional X-rays of the primary lesion 5. CT and MRI scan of the primary lesion 6. CT scan of the chest 7. Additional investigations when clinically required *For those patients treated with primary chemotherapy End of treatment (about 4 weeks after chemotherapy completion)
1. Physical examination 2. Complete blood count 3. Serum creatinine, Na, K, Mg, GFR, TmP/GFR, bilirubin, transaminases, alkaline phosphatase (optional bone specific alkaline phosphatase), LDH 5. Estimation of left ventricular ejection fraction (LVEF) with cardiac ultrasound or radionuclide ventriculography (MUGA) before first course containing doxorubicin 6. Conventional X-rays of the primary lesion 7. CT scan of the chest 8. Additional investigations when clinically required Euroboss I Patients should be followed at 3 months intervals for 3 years, at 4 months intervals during the 4th and5th years, and then at yearly intervals until 10 years after treatment was completed.
Mandatory investigations at follow-up:
1. Complete physical examination 2. A-P and lateral chest X-rays at each visit. The CT scan of the chest is optional as routine, but it must always be done if chest X-ray shows metastasis or is inconclusive 3. Blood count (Hb, white blood counts, Trc), transaminases, ALP, LDH, serum creatinine, Na, K, Mg, P at each visit 4. GFR and TmP/GFR at 3 months, 6 months, at 6 months intervals during the second and third years, 5. Estimation of left ventricular ejection fraction (LVEF) with cardiac ultrasound or radionuclide ventriculography (MUGA) at 6 months, 12 months and then at 3 years intervals 6. Audiogram one year after the completion of therapy 7. Bone scan and plain X-rays on clinical suspicion of bone metastases; if inconclusive supplement 13. Chemotherapy administration
13.1 General consideration
Before each cycle of chemotherapy
1. Physical examination 2. Complete blood count, serum creatinine, Na, K, Mg, transaminases, alkaline phosphatase, LDH, urine analysis. GFR before each cycle with MTX. Tmp/GFR before each cycle with ifosfamide 3. After MTX administration: complete blood count, serum creatinine, Na, K, Mg, transaminases, serum MTX levels, urine analysis 4. After each cycle of chemotherapy with DOX, CDP, IFO: complete blood count, from day 9 to 16, on alternate days are suggested, but longer intervals are allowed if clinically feasible 5. Additional investigations when clinically required Bone marrow function
Cycles with CDP, DOX, IFO require a minimum number of 3 × 109/L leukocytes (or 1 × 109/L of
neutrophils), and of 100 ×109/L platelets. Cycles with MTX require a minimum number of 2 × 109/L
leukocytes (or 0.5 × 109/L neutrophils), and of 80 × 109/L platelets.
Euroboss I Renal function
Cycles with CDP, IFO require serum creatinine levels in a normal range. Cycles with MTX require a
GFR > 70 ml/min. Episodes of reversible renal toxicity do not contraindicate further administration of
CDP and IFO. Persistent reduction of TmP/GFR and electrolyte disturbances contraindicates the ad-
ministration of IFO and CDP. Delayed MTX excretion with renal toxicity, even if reversible, is a
contraindication to further MTX administrations.
Cardiac function
An EF > 55% (or fractional shortening rate of the left ventricle >28%) is mandatory for the use of
DOX. Before each cycle with DOX physical examination and EKG (optional) and before the last cycle
an echocardiogram or MUGA-scan is required. In case of clinical signs or changes in EKG suggesting
a possible heart dysfuncion DOX is not administered, and the patient undergoes additional tests to
assess the cardiac function. In case of no evidence of cardiac dysfunction DOX is administered.
A >10% reduction of the ejection fraction (compared to the baseline) contraindicates further DOX
administration. Despite a reduction of the ejection fraction, DOX can be delivered (short infusion) with
the use of dexrazone (cardioxane) if considered of clinical relevance.
Antiemetic treatment
The antiemetic treatment will be decided by each institution, but the use of 5HT3 antagonists and
dexametazone is recommended for the cycles with CDP, DOX, IFO.
13.2 G-CSF
G–CSF support is mandatory (within ISG and SSG) after each course with IFO/DOX, IFO/CDP orCDP/DOX chemotherapy. G–CSF support is not necessary after MTX.
G–CSF is administered as a subcutaneous injection or i.v. infusion once daily at a dose of 300 µg topatients < 80 kg and otherwise 480 µg. Administration of G–CSF should be started 48–72 hours aftertermination of chemotherapy and 7–8 daily doses are recommended. G–CSF must be discontinued atleast 24 hours before starting the next course of chemotherapy and it should be stopped when the totalwhite blood count exceeds 5.0 × 109/l.
13.3 Cisplatinum (CDP/DOX, IFO/CDP)
General considerations
CDP (cisplatinum) is administered in combination with doxorubicin (CDP/DOX) or (IFO/CDP).
IFO/CDP: Ifo (Ifosfamide) is administrated on day 1 and 2 and Cis (Cisplatinum) on day 3 and 4.
Drug interactions: Aminoglucosides may augment the nephrotoxicity of cisplatinum.
Blood check-ups on second day of cisplatinum infusion: GOT, GPT (=ASAT, ALAT), creatinine,
Mg, Ca, Na, K and P.
Basal solution: 0.9% NaCl with 20 mmol KCl/L and 3 mmol Mg/L.
a. Prehydration: 1000 ml of basal solution for 2 hours.
b. Cisplatinum dose: 50 mg/m2/day is administered in 2 000 ml/m2/day of basal solution as a continuous infusion for 2 days (48 hours).
NOTE: CaCl must not be infused together with cisplatinum in the same infusion line because it causes
the formation of stable CaSO complexes, which blocks the catheter.
c. Posthydration: 2000 ml of basal solution should be given over a 12 hours period.
d. Diuresis: If < 800 ml in 6 hours, give furosemide 10–20 mg.
Euroboss I 13.4 Doxorubicin (CDP/DOX, IFO/DOX)
Doxorubicin (DOX) is administered in combination with cisplatinum (IFO/DOX or CDP/DOX).
DOX is started day 3 in both courses.
Doxorubicin (DOX) 60 mg/m2 (CDP/DOX) is given as a 24 hours continuous infusion in 1 000 ml5% glucose.
13.5 Ifosfamide (IFO/CDP, IFO/DOX)
Blood check-ups daily: Hematocrit, white blood counts, platelets, venous acid/base (or serumbicarbonate), uristix, creatinine, Na, K, Ca, Mg, GOT and GPT (=ASAT, ALAT).
Basal solution: 0.9% NaCl with 40 mmol NaHCO /L + 20 mmol KCl/L.
a. Prehydration and alkalinization: Infuse 500 ml basal solution with Mesna 400 mg/m2 over a
1 hour period.
b. Dose: The doses of ifosfamide is 3000 mg/m2/day, each for two consecutive days, giving a total
dose of ifosfamide of 6 000 mg/m2. Ifosfamide is resolved in 500 ml basal solution and infused in2 hours.
c. Postifosfamide alkalinization and mesna administration: Immediately following the ifosfamide
infusion: mesna 3 000 mg/m2 in 2000 ml/m2 basal solution in 22 hours.
e. Diuresis: If <400 ml/m2 in 6 hours, give furosemide 10–20 mg. Check for hematuria every 24
hours. With macroscopic hematuria or microscopic hematuria (+++ or more) confirmed bymicroscopic examination (>10 red blood cells/field), ifosfamide should be withheld and basal solu-tion, 2000 ml/8hours with 1000mg/m2 mesna should be infused until the urine clears. The ifosfamideinfusion should then be (re)started.
NOTE: Uristix may be falsely negative or positive during treatment with ifosfamide.
Additional alkalinization: If urine pH <7 or venous acid/base indicates metabolic acidosis
(serum bicarbonate <21 mmol/L), give 2 mmol NaHCO /kg intravenously during 30 minutes.
g. Treatment and prophylaxis for ifosfamide-induced CNS toxicity: The cause of ifosfamide-
induced acute encephalopathy is unknown. It may be dose-dependent and may be aggravated by
metabolic acidosis. The condition is reversible. The commonest symptom of mild CNS toxicity is
undue somnolence, which usually does not require specific measures other than to keep the serum
bicarbonate levels >21 mmol/L. The ifosfamide infusion should not be interrupted. Severe
encephalopathy
is recognized by disorientation, visual and cognitive disturbances, undue fear,
nightmares, hallucinations or even convulsions. The symptoms usually start insidiously and slowly
increase. The ifosfamide infusion should be stopped and treatment instituted with methylene blue
50 mg i. v. every 8 hours together with infusion of basal solution, 2000ml/8 hours with 1 000mg/m2
mesna. Metabolic acidosis must be corrected according to recommendation above. The symptoms
generally disappear quickly and 2–3 methylene blue infusions usually suffice. This Ifo course should
not be re-started.
In subsequent Ifo courses, prophylactic treatment with oral methylene blue 50 mg × 3 daily shouldbe given when starting ifosfamide. This will usually prevent further CNS toxicity (23).
Methylene blue is a non-toxic agent. Its exact mechanism of action in this context is not preciselyknown.
NOTE: Methylene blue is not routinely available in hospital pharmacies and must be purchased in
advance in institutions giving Ifo treatment!
Euroboss I High-dose Methotrexate is given postoperatively to poor responders only.
Drug interactions: Avoid simultaneous use of the following drugs because of the risk of interactions:
penicillin, NSAID probenicid, sulfamethoxazole trimethoprim and salicylic acid.
a. Daily blood checks after starting the MTX infusion and until the serum MTX is <0.2 µmol/L: GOT, GPT (= ASAT, ALAT), Na, K, S-creatinine.
b. Prealkalinization and prehydration: Use the following solution i.v.: 500 ml/m2 5% glucose with 100 mmol NaHCO /L and 20 mmol KCL/L over a period of 60 minutes.
c. Dose of methotrexate: 8000 mg/m2 d. Methotrexate should be dissolved in 500 ml of 5 % glucose with 40 mmol NaHCO /L and 20 mmol KCl/L. This methotrexate solution is infused over four hours.
e. Total fluid input/day until serum MTX concentration <0.2 µmol/L 2 500 ml/m2 (including prealkalinization, methotrexate infusion and oral fluids) (T –T ): 2 000ml/m2 (T –T ): 2 000 ml/ m2 (T –T ): 2 000 ml/ m2 For all i.v. fluid in the posthydration, use 5% glucose with 40 mmol NaHCO /L + 20 mmol KCl/L.
Leucovorin (folinic acid) rescue: 8 mg/m2 intravenously or orally every 6th hour, beginning 24hours after starting the methotrexate infusion. Normally, leucovorin is given by eleven doses untilT . It is sufficient to give leucovorin until six hours after the methotrexate concentration has fallen below 0.2 µmol/L.
g. Determinations of serum methotrexate levels: Capillary or venous blood (not taken from the vein used for the methotrexate infusion). Blood samples for methotrexate concentrations should betaken just before the end of the methotrexate infusion (T sample), and then at least at T and every 24th hour until serum MTX is <0.2 µmol/L.
h. Diuresis: Give furosemide 10–20 mg if diuresis <400 ml/m2. If the total fluid volume is increased to 3 000 ml/m2/24 hours because of delayed MTX excretion, the minimum level of diuresis should beincreased to 600 ml/m2 in 6 hours.
Additional alkalinization: If the urine pH is <7 give 2 mmol NaHCO /kg during Monitoring MTX and fluid volume: All serum MTX values, i.v. and oral fluids, diuresis, urinarypH, supplemental NaHCO and furosemide should be listed on a detailed chart to ensure accurate monitoring of MTX clearance and fluid balance.
Euroboss I 14. Dose adjustments
Methotrexate
No dose reduction. In case of delayed MTX excretion and concomitant nephrotoxicity the following
cycles of MTX are omitted.
Ifosfamide/cisplatin course
If neutropenic (neutrophils <0,5 × 109/L) fever reduce following IFO/CDP cycle: IFO 75%, if repeated:
50%.
If negligible myelosuppression with reduced dose: back to previous dose level.
If creatinine >120 µmol/L following CDP: reduce the next dose to 75%. Omit CDP if repeated.
Peripheral neuropathy >Grade 3: omit CDP (sensory loss or paresthesia interfering with activities ofdaily living).
Ifosfamide/doxorubicin course
If neutropenic (neutrophils <0,5 x 109/L) fever: reduce following IFO/DOX cycle: IFO 75%, if repeated:
50%.
If negligible myelosuppression with reduced dose: back to previous dose level.
Cisplatin/doxorubicin cycle
If neutropenic (neutrophils <0,5 × 109/L) fever reduce following CDP/DOX cycle: CDP 75%,
if repeated 50%.
If negligible myelosuppression with reduced dose: back to previous dose level.
If creatinine >120 µmol/L following CDP: reduce the next dose to 75%. Omit CDP if repeated.
Peripheral neuropathy >Grade 3: omit CDP (sensory loss or paresthesia interfering with activities ofdaily living).
15. Serious adverse events, stopping rules
Adverse events
Death (other than death of disease) under treatment or within 12 months from the end of treatment will
be regarded as adverse event, unless it is proven that there is no relation with therapy, e.g. traffic
accidents.
Life-threatening treatment-related complications, i.e. CTC grade 4 toxicity of the following categoriesare regarded to be adverse events: cardiac, renal, hepatic*, central nervous, peripheral nervous, skin.
CTC grade 4 neutropenia (and neutropenic infection), thrombocytopenia which resolve and do nothave life-threatening consequences are to be expected with the procol presented here and are notregarded as serious adverse events.
Any life-threatening event; however, must be reported (see SAE form) immediately, i.e. within the nextworking day, and followed up, regardless whether it falls within the categories listed above or not. Allparticipating groups (institutions) will be notified about important toxicities according to GCP guidelines.
* except following methotrexate chemotherapy Euroboss I Stopping rules
Toxic deaths: The expected toxicity is mainly based on previous experiences of the participating groups
in chemotherapy protocols applied to a younger population (4, 6, 8). In the current protocol a dose
adaptation of the drugs has been planned due to the age of patients included.
Interim analyses on severe acute toxicity (grade 4 other than haematologic toxicity and mucositis) andtoxic deaths will take place twice a year by the "Resource group".
Log rank and crude percentage comparison tests will compare deaths not related to the underlyingmalignant disease to historical control in patients aged <41 years. If any of these tests is significant atp <0.001, the conclusion will be that there is a relative excess of toxic deaths; then a full analysis will beconsidered.
Crude percentage will also be compared to the theoretically acceptable toxic death rate. If the lowerboundary of the 99.9% confidence interval of the observed percentage is above this limit, the conclusionwill be that there is an absolute excess of toxic deaths; then a full analysis will be considered. Based onprevious experience (4, 6, 8) in younger patients the limit percentage has been set at 3%.
Number of treated patients K: number of toxic deaths leading to the conclusions of an absolute excess of toxic deaths.
If the analysis concludes that there is an absolute excess of toxic deaths, the study will be stoppedimmediately by the study-coordinators.
Euroboss I 1. Malawer MM, Link P, Donaldson SS. Sarcomas of bone. In: DeVita VT Jr, Hellman S, Rosenberg SA, editors. Cancer principles and practice of oncology. Philadelphia-New York: Lippincott-Raven,1997: 1789-1852.
2. Campanacci M. High grade osteosarcomas. In: Campanacci M ed. Bone and soft tissue tumors.
Wien-New York: Springer-Verlag 1999: 463-557.
3. National Cancer Institute, PDQ. Osteosarcoma. Available from (URL): http://www.cancernet.nci 4. Sæter G, Alvegard TA, Elomaa I, Stenwig AE, Holmström T, Solheim OP: Treatment of osteosarcoma of the extremities with the T-10 protocol, with emphasis on the effects of preoperative chemotherapywith single-agent high-dose methotrexate: a Scandinavian Sarcoma Group study. J. Clin. Oncol., 9:1766-1775, 1991.
5. Bramwell VHC, Burgers M, Sneath ., Souhami R, van Oosterom AT, Voute PA, Rouesse J, Spooner D, Craft AW, Somers R, Pringle J, Malcom AJ, van der Eijken J, Thomas D, Uscinska B, MachinD, van Glabbeke M: A comparison of two short intensive adjuvant chemotherapy regimens inoperable osteosarcoma of limbs in children and young adults: the first study of the EuropeanOsteosarcoma Intergroup. J. Clin. Oncol., 10: 1579-91, 1992.
6. Bacci G., Picci P, Ferrari S, Ruggieri P, Casadei R, Tienghi A, Brach Del Prever A, Gherlinzoni F, Mercuri M, Monti C: Primary chemotherapy and delayed surgery for non metastatic osteosarcomaof the extremity. Results in 164 patients preoperatively treated with high doses of methotrexate,followed by cisplatin and doxorubicin. Cancer, 72: 1216-1226, 1993.
7. Provisor AJ, Ettiger LJ, Nachman JB, Krailo MD, Makley JT, Yunis EJ, Huvos AG, Betcher DL, Baum ES, Kisker T, Miser JS: Treatment of nonmetastatic osteosarcoma of the extremity withpreoperative and postoperative chemotherapy: a report from the Children's Cancer Group. J. Clin.
Oncol., 15: 76-84, 1997.
8. Fuchs N, Bielack SS, Epler D, Bieling P, Delling G, Koholz D, Graf N, Heise U, Jurgens H, Kotz R, Salzer-Kuntschik M, Weinel P, Werner M, Winkler K: Long-term results of the co-operativeGerman-Austrian-Swiss osteosarcoma study group's protocol COSS-86 of intensive multidrugchemotherapy and surgery for osteosarcoma of the limbs. Ann. Oncol., 9: 893-899, 1998.
9. Meyers PA, Gorlick R, Heller G, Casper E, Lane J, Huvos AG, Healey JH: Intensification of preoperative chemotherapy for osteogenic sarcoma: results of the Memorial Sloan-Kettering (T-12) protocol. J. Clin. Oncol., 16: 2452-2458, 1998.
10. Huvos AG: Osteogenic sarcoma of bones and soft tissues in older persons. A clinicopathologic analysis of 117 patients older than 60 years. Cancer, 57: 1442-1449, 1986.
11. Frassica FJ, Sim FH, Frassica DA, Wold LE: Survival and management considerations in postirradiation osteosarcoma and Paget's osteosarcoma. Clin Orthop, 270: 120-127, 1991.
12. Healey JH, Buss D: Radiation and Pagetic Osteogenic sarcomas. Clin Orthop, 270: 128-134, 13. Naka T, Fukuda T, Shinohara N et al: Osteosarcoma versus Malignant Fibrous Histiocytoma of bone in patients older than 40 years. Cancer, 76: 972-984, 1995.
14. Bacci G, Ferrari S, Donati D et al: Neoadjuvant chemotherapy for osteosarcoma of the extremity in patients in the fourth and fifth decade of life. Oncol Rep, 5: 1259-1263, 1998.
Euroboss I 15. Grimer RJ: Osteosarcoma over the age of 40. Proceedings of EMSOS meeting, London, May 4-5, 2000.
16. Bielack S, Kempf-Bielack B, Delling G et al: Prognostic factors in high-grade osteosarcoma of the extremities or trunk. An analysis ot 1702 patients treated on neoadjuvant Cooperative OsteosarcomaStudy Group protocols. J Clin Oncol., 20: 776–90, 2002.
17. Lichtman SM: Physiological aspects of aging. Implications for the treatment of cancer. Drugs Aging, 7: 212-225, 1995.
18. Begg CB, Carbone PP: Clinical trials and drug toxicity in the elderly. The experience of the Eastern Cooperative Oncology Group. Cancer, 52: 1986-1992, 1983.
19. Walsh JW, Begg CB, Carbone PP: Cancer chemotherapy in the elderly. Sem Oncol, 16:66-75, 20. Balducci L, Beghe C: Pharmacology and chemotherapy in the older cancer patient. Cancer Control, 6: 466-470, 1999.
21. Picci P, Bacci G, Ferrari S, Mercuri M: Neoadjuvant chemotherapy in malignant fibrous histiocytoma of bone and in osteosarcoma located in the extremities: Analogies and differences between the twotumors. Ann Oncol, 8: 1107-1115, 1997.
22. Earl HM, Pringle J, Kemp H, Morittu L, Miles D, Souhami R: Chemotherapy of malignant fibrous histiocytoma of bone. Ann Oncol, 4: 409-415, 1993.
23. Bramwell VHC, Steward WP, Nooij M et al: Neoadjuvant chemotherapy with doxorubicin and cisplatin in Malignant Fibrous Histiocytoma of bone: A European Osteosarcoma Intergroup Study.
J Clin Oncol, 17: 3260-3269, 1999.
24. Waddell AE, Davis AM, Ahn H, Wunder JS, Blackstein ME, Bell RS: Doxorubicin-cisplatin chemotherapy for high-grade nonosteogenic sarcoma of bone. Comparison of treatment and controlgroups. Can J Surg, 42: 190-199, 1999.
25. Antonescu CR, Erlandson RA, Huvos AG.: Primary fibrosarcoma and malignant fibrous histiocytoma of bone, a comparative ultrastructural study: evidence of a spectrum of fibroblastic differentiation.
Ultrastruct Pathol, 24: 83-91, 2000.
26. Antonescu CR, Erlandson RA, Huvos AG: Primary leiomyosarcoma of bone: a clinicopathologic, immunohistochemical and ultrastructural study of 33 patients and a literature review. Am J SurgPathol, 21: 1281-1294, 1997.
27. Derre J, Lagace R, Nicolas A et al: Leiomyosarcoma and most malignant fibrous histiocytomas share very similar comparative genomic hybridization imbalances: an analysis of a series of 27leiomyosarcomas. Lab Invest, 81: 211-215, 2000.
28. Mercuri M, Picci P, Campanacci L, Rulli E: Dedifferentiated Chondrosarcoma. Skeletal Radiol, 24: 409-416, 1995.
29. Enneking WF, Spanier SS, and Goodman MA: Current concepts review. The surgical staging of musculoskeletal sarcoma. J. Bone and Joint Sur., 62-A: 1027-1030, 1980.
30. Rosen G, Marcove RC, Caparros B, Nirenberg A, Kosloff C, Huvos AG: Primary osteogenic sarcoma: the rationale for preoperative chemotherapy and delayed surgery. Cancer, 43: 2163-2177, 1979.
31.Salzer-Kuntschik M, Delling G, Beron G, Sigmund R: Morphological grades of regression in osteosarcoma after polychemotherapy-study COSS 80. J Cancer Clin Oncol, 106 (s): 21-24,1983.
Euroboss I 32. Picci P, Bacci G, Campanacci M et al: Histologic evaluation of necrosis in osteosarcoma induced by chemotherapy. Regional mapping of viable and nonviable tumor. Cancer, 56: 1515-1521, 1985.
33. National Cancer Institute common toxicity criteria, version 2.0. Available from (URL): 34. Aeschlimann C, Wermuth B, Cerny T: Prophylaxis and reversal of ifosfamide encephalopathy with methylene blue, Lancet, 343, 763–764, 1994.
35. American Society of Clinical Oncology. Recommendations for the use of hematopoietic colony- stimulating factors: evidence-based clinical practice guidelines. J Clin Oncol, 12, 2471-508 1994.
Euroboss I II. APPENDIX
Participating centers within SSG Patient information Reporting of serious adverse events Management of methotrexate toxicity and delayed methotrexate excreation Calculation of renal tubular reabsorption of phosphate – TmP/GFR WHO performance status Associated research projects Guidelines for pathology Guidelines for surgery Guidelines for radiotherapy Euroboss I 1. Participating centers and referral centers within SSG
(Referral centers marked in bold) Orthopedic Surgeon Karolinska Hospital, Stockholm
O. Brosjö
Sahlgrenska University Hospital, Gothenburg
University Hospital, Uppsala University Hospital, Umeå University Hospital, Linköping University Hospital, Lund
H. Domanski
A. Rydholm
University Hospital, Helsinki
T. Böhling
A. Kivioja
University Hospital, Turku University Hospital, Tampere University Hospital, Kuopio University Hospital, Oulo The Norwegian Radium Hospital, Oslo
G. Follerås
Haukeland University Hospital, Bergen University Hospital (St. Olav), Trondheim University Hospital, Tromsø The evaluation of histologic response will be performed at the referral centers.
A conclusion that a tumor is inoperable must be done at a referral center.

Euroboss I 2. Patient information
EUROBOSS I
A European treatment protocol for high-grade bone sarcoma in patients
older than 40 years.

You have recently been diagnosed with bone cancer. Primary bone tumors are very rare diseases, andin particular in your age group. The knowledge and the current standard therapy are mainly based onstudies in children and adolescent, in which the use of modern chemotherapy has significantly improvedsurvival. Evidence exists that this therapy is also effective in adult patients, but due to their rarity as upto now no firm conclusion can be made. This study aims to answer some of these questions. Acollaboration between several European groups covering a population of more than 200 mill. shouldallow enough information to be obtained to make progress in the knowledge and treatment also foradult patients.
The planned chemotherapy according to this protocol is very intensive. We have made some adjustmentsto treatment given to younger patients since it is well known that normal changes occur with ageingincluding decreased bone marrow and kidney activity. In addition, we have included guidelines fordose adjustments during therapy to safely modify your treatment if necessary.
Acute side-effects almost always follow intensive chemotherapy. For you, these will include nausea,hair loss and bone marrow suppression. Those are all reversible and we will give you supportivemedicine to reduce your discomfort. Due to the expected bone marrow toxicity you will be carefullymonitored with blood tests between the cycles. Patients with low white blood cell counts harbours ahigh risk of developing serious and life-threatening infections. Infections start with fever and you willbefore starting therapy receive oral and written information with strict guidelines on how to react uponfever in periods with low blood values.
Intensive chemotherapy also has a significant risk of development of late organ complications. Themost common include impaired function of heart, kidney and your sense of hearing. Men have a highrisk of developing infertility but the sexual hormone production is generally not affected. Routinely allpatients will go through specified investigation to ensure proper organ functions before, during and aftercompletion of therapy. If necessary, treatment will be adjusted according to changes in organ functionduring therapy.
Your participation in this project will not change your therapy or require additional investigations sincewe consider the protocol as the current standard management of patients with bone tumors at your agegroup. With your participating you allow us to collect clinical data, blood samples and tumor tissue forthe present and possible future studies. Selected clinical data will be recorded anonymously and storedin a common study-database which is placed at the Rizzoli Institute in Bologna, Italy. If you do not wantto participate, this will of course have no consequences for the treatment you will receive. You are alsofree to withdraw from the study, at any time, without any further justification. By signing the formulabelow, you accept your willingness to participate and confirm that adequate information has been given.
Dr. Sigbjørn Smeland Chairman of osteosarcoma studies/Principal InvestigatorScandinavian Sarcoma Group Euroboss I 3. Reporting serious adverse events
All serious adverse events, related or not to the study treatment, occurring during the treatment periodor within 30 days after the last protocol treatment administration, must be reported to the main studysecretariat: SSG secretariat
Regional Tumor Registry
Lund University Hospital
SE–221 85 Lund
Sweden

Tel. +46–46–17 75 55
Fax. +46–46–18 81 43
E-mail: [email protected]

This must be done by fax within 24 hours of the initial observation of the events. Details should bedocumented on the specified Serious adverse event form. The study secretariat will forward all Seriousadverse event reports within 24 hours of receipt to all appropriate persons. A complete report mustfollow the initial report within 10 days.
4. Management of methotrexate toxicity and delayed
4.1 General considerations
Prompt intervention will prevent severe toxicity. Severe toxicity is anticipated if there is a greater than100% rise in the serum creatinine level within 24 hours after start of the methotrexate infusion and/or theserum methotrexate levels are in the "toxicity range" on the MTX excretion curve (see below). Patientsin this situation should be treated by continued hydration and alkalinization of the urine with 3 000 ml/m2/24 h of 5% glucose with 40 mmol NaHCO /L and 20 mmol KCl/L. In this case, the minimum diuresis should be increased to 600 ml/m2/6 h. Increase the dose of leucovorin as described below.
The administration of potassium should be carefully monitored, depending on renal function. Bodyweight, fluid input and output and blood pressure should be monitored. Blood counts, serum creatinine,liver transaminases, ALP, bilirubin and serum methotrexate levels should be measured daily. If increasedserum-creatinine, kidney function should be evaluated with GFR. Records should be kept of the clinicalcourse. Always ensure that the patient is not taking other medications which interfere with methotrexatebinding or excretion. If stomatitis and myelosuppression are severe enough to delay subsequentchemotherapy courses, then rescue should be continued for one additional day in subsequent MTXcourses, i.e. 5 additional doses of leucovorin after the serum MTX is <0.2 µM.
Euroboss I 4.2 Methotrexate excretion curve
Adjustment of leucovorin dose during delayed methotrexate excretion Patient's actual serum MTX × standard daily dose of leucovorin of leucovorin (mg) = ———————————————————————— Upper limit of serum MTX for the actual day and time The upper limit of decline in serum MTX levels as a function of time is shown in theMTX excretion curve.
The upper limit of serum MTX Example:If the 48 hours methotrexate level was 40 µM, the leucovorin dose should be adjusted to: 32mg/m2 × 40 = 640 mg/m2/24 hours by continuous i.v. infusion 2 It is possible to reduce the dose of leucovorin on the following days in relation to the reduction inS-MTX.
When the S-MTX level is in the range of 0.9–0.2 µM, give leucovorin in doses of 8 mg/m2 orallyevery 6 hours until one dose after the serum level is <0.2 µm.
Note: Always continue to monitor urine pH and give more NaHCO if pH <7.
Euroboss I 5. Calculation of renal tubular reabsorption of phosphate
– TmP/GFR
The ratio of the renal tubular maximum reabsorption rate of phosphate to the glomerular filtration rate(TmP/GFR) is used to monitor renal tubular function after administration of potential toxicchemotherapeutics (i.e. ifosfamide) Calculation of TmP/GFR requires the calculation of the fraction tubular reabsorption of phosphate(TRP).
TRP = 1-{(U /P ) × (P /U )}
Up: urine phosphatePp: Pcr: plasma creatinineUcr: urine creatinine If TRP is < 0.86: Phosphate reabsorption is at its maximum and there is a linear relationship
between changes in plasma phosphate and excretion. TmP/GFR is therefore given by the relation:
TmP/GFR = TRP × Pp
If TRP is > 0.86: There is a curvilinear relationship between changes in plasma phosphate and
excreation which fits a rectangular hyperbola. TMP/GFR is given by the relation:
TmP/GFR = 0,3 × TRP/{1-(0,8 × TRP)} × Pp
TmP/GFR is measured in mmol/L. Reference ranges in patients 40–65 years are:Male 0,90–1,35 mmol/L 0,88–1,42 mmol/L References:1. Barth JH, Jones RG and Payne RB. Calculation of renal tubular reabsorption of phosphate: the algorithm performs better than the nomogram. Ann Clin Biochem 2000; 37: 79–81 2. Payne RB. Renal tubular reabsorption of phosphate (TmP/GFR): indications and interpretation.
Ann Clin Biochem 1998; 35: 201–206 Euroboss I 6. WHO performance status: ECOG
Able to carry out all normal activity without restriction Restricted in physically strenuous activity but ambulatory and able to carry out light work Ambulatory and capable of self-care but unable to carry out any work; up and about morethan 50 % waking hours Capable of only limited self-care; confined to bed or chair more than 50% of waking hours Completely disabled; cannot carry on any self-care; totally confined to bed or chair 7. Associated research projects
Micrometastases (only for patients with osteosarcoma)
MICROMETASTASIS IN PATIENTS WITH OSTEOSARCOMA
Coordinator: Professor Ø.S. Bruland, Oslo 7.1 Aims
1. To collect samples of mononuclear cells derived from peripheral blood and bone marrow aspirates
of patients with various biological subtypes and clinical stages of osteogenic sarcoma (OS).
2. To evaluate the feasibility of immunomagnetic positive tumor cell isolation, using monoclonal antibo- dies TP–3 and 9.2.27 in combination with Dynabeads®.
3. To generate hypotheses in relation to the use of such methods as a prognostic tool in OS patients on the basis of findings in this study.
4. To explore the biological role of circulating tumor cells in OS.
7.2 Background
The current management of OS is fraught with difficult dilemmas (22). In particular, overtreatment and
long-term sequelae among cured patients are of major concern.
OS is considered a systemic disease characterized by the presence of sub-detectable tumor spread atprimary diagnosis in the vast majority of patients. Adjuvant chemotherapy is a cornerstone in the currentmulti-modal management. However, OS displays considerable heterogeneity with regard to metastaticcapacity and chemosensitivity. The current outcome may well be further improved by individualizing thetreatment. Up to now it has not been possible to identify subgroups with different prognosis of patientsand better methods for "biological staging" are needed. The presence of circulating tumor cells maywell be a new prognostic factor. If consensus with regard to prognostic factors can be achieved, thestage might be set for exploring "risk-adapted therapy" in OS (22).
A method for immunomagnetic isolation and detection of tumor cells present in mononuclear cellsuspensions from bone marrow (BM) aspirates and peripheral blood (PBL) will be applied. Twomonoclonal antibodies, known to be reactive with osteosarcoma cells, but not with normal hematopoieticcells are used in this rosetting technique. Based on the cumulative experience (now 43 OS-patients)from The Norwegian Radium Hospital over a ten years period, it was decided to include"micrometastases" as one of the research projects in the protocols ISG/SSG I and II. It has been Euroboss I decided to continue this study also in the SSG XIV protocol. The aim is to increase the number ofsamples so that any conclusion regarding the potential clinical significance of micrometases can bedrawn faster.
In ISG/SSG I and II each center was encouraged to freeze down mononuclear cells from BM and PBLblood to be sent to Oslo for subsequent testing. "Rizzoli" supplied samples for analyses from 29 OS-patients – "Lund" four; samples that now are under analysis.
The specimens from patients treated at The Norwegian Radium Hospital were analyzed directly afterthe sampling, whereas frozen materials have been used from the two other institutions. Using freshmaterials 61% (11/18 pts) of "classical" OS-patients were positive in BM, only one positive in PBL.
Interestingly, none of the BM-negative patients have suffered a relapse with a mean follow-up of 47months, whereas three did so in the BM-positive cohort (one dead of disease, one alive with multiplemetastases and one in CR, salvaged with thoracotomy) – mean follow-up 45 months. Two out of fourwith axial primary tumors had tumor cells in BM, none in PBL. Among patients with overt metastasesat primary diagnosis 92% (11/12) had positive BM-samples, and usually a very high number of rosetteswere recovered. Five of the nine patients with relapsed metastatic disease (56%) had positive BM.
Two of the four negative patients had several years of disease free interval from primary treatment anddetection of a solitary lung metastasis, and both are alive NED at six years follow-up (25).
The analysis of frozen samples seemingly give conflicting results in as much as fewer of the patientsscore positive. The number of mononuclear cells recovered after thawing are generally much lowermaking a direct comparison difficult.
7.3 Organization
Data in the study will be collected by the Clinical Research Branch / Department of Oncology, Norwegian
Radium Hospital; it will be based on information given in the enclosed registration form and data from
the central register of the protocol. The Department of Tumor Biology, Norwegian Radium Hospital,
will perform the tumor cell isolation assays without knowledge of clinical information.
7.4 Clinical material
Peripheral blood (20 ml heparinized blood) and bone marrow aspirates (10–20 ml) will be collected
into tubes containing heparin (heparin coated Vacutainer etc.). Material will then be transferred to tubes
for shipment, as overnight express postal service. This material will be supplied each center. The prepa-
ration of mononuclear cells and testing with the rosetting assay will be performed at the Department of
Tumor Biology, Norwegian Radium Hospital.
7.5 Practical aspects of sampling
Samples will be obtained from each patient, preferably at 1) initial diagnosis, 2) at the time of primary
surgery following preoperative chemotherapy, 3) at the end of treatment.
If a relapse is diagnosed and surgical removal of metastases is planned, renewed sampling is highlyrecommended.
Samples can often be obtained simultaneously with a surgical procedure. This is an advantage especiallyin younger patients. Blood and bone marrow should be drawn before manipulating the tumor. If possible,fresh tumor material should also be frozen for subsequent comparative studies.
Mark each tube with: * patient's initials* birth date * BM (bone marrow), PB (peripheral blood) Euroboss I 7.6 Future prospects
A relationship seemingly exists between the presence and number of isolated tumor cells and
clinical stage and progression of the disease. The results obtained are promising for the use
of this sensitive method in selecting patients to receive individualized therapeutic interven-
tion, for monitoring effects of systemic treatment, and as part of patient follow-up programs.

The extra samples of cryo-preserved material are available for exploring improved bead-technologyand/or PCR assays. Plasma samples are collected for evaluating the usefulness of novel "tumor-markers".
7.7 Shipment of samples
It is mandatory to contact Prof. Bruland before sending the material!
Prof. Øyvind BrulandDept. of OncologyNorwegian Radium HospitalNO–0310 OsloNorwayTel +47–22–93 40 00, Fax +47–22–52 55 59Email: [email protected] 7.8 Informed consent for patients regarding the research project
"DETECTION OF MICROMETASTASIS IN OSTEOSARCOMA"
You have recently been diagnosed with a type of bone cancer, osteosarcoma. Modern treatment of this
disease has indeed improved the cure rates. Before effective cytostatics were available, approximately
20% of the patients without detectable spread at the time of operation were cured. In contrast today
close to 70% of osteosarcoma patients are cured.
Modern combination chemotherapy given to osteosarcoma patients is very intensive. In addition to theacute adverse effects there is also a significant risk for the development of late complications. An aim inthe international research community is to optimise the combination of drugs and their doses (so called"risk adapted therapy") so that the most aggressive combinations are reserved for those patients thatreally need it. Presence of metastatic disease is a known risk factor and will imply more aggressivechemotherapy. We know from historical data that most patients with osteosarcoma have metastaticspread at the time of primary diagnosis. The problem is that the majority of these patients only havemicrometastasis, that is lesions that are so small that they can not be identified with current diagnosticmodalities (X-rays, etc.). We are with the current methods not able to distinguish between patient withmicrometastatic spread and patients without metastatic disease. The consequence is that all patientsreceive the similar intensive chemotherapy. In an aim to identify patients with microscopic spread wehave embarked upon a common Scandinavian research protocol linked to our current osteosarcomatreatment; "The Micrometastasis Project".
The Norwegian Radium Hospital has developed special techniques to disclose single tumor cells thatare spread from the primary tumor in blood and bone marrow. The aim of the research project is tocollect materials from a number of patients so that we can learn if the detection of these tumor cells is ofprognostic value and could aid our treatment of the individual patients.
Your participation in the project implies that we need to draw a bone marrow aspirate and a sample ofperipheral blood. These will be done before the start of chemotherapy, at the time of definite surgery for Euroboss I the primary tumor and when the adjuvant chemotherapy is completed. Also patients experiencing relapseswill be candidates. We will aim to do these procedures when you are under general anaesthesia forprimary biopsy, implantation of venous access port, etc. In cases where general anaesthesia will not begiven, you will receive local anaesthesia, as is the routine procedure for several other groups of patients.
You and your relatives should decide your participation in this study. If you do not want to participate,this will of course have no consequences for the treatment you will receive. You are also free to withdrawfrom the study, at any time, without any further justification. You and your relatives accept your willingnessto participate and confirm that information has been given by signing the formula below.
Øyvind S. Bruland Sigbjørn Smeland Professor of Clinical Oncology Research Project Leader Protocol chairman 8. Guidelines for pathology
Chairman:
Prof. Lars Gunnar Kindblom
Dept. of Pathology
Sahlgrenska Hospital
SE-413 45 Gothenburg
Tel +46-31-342 10 00
[email protected]
Coordinators:
Dr. Tom Böhling
Dr. Anna Elisabeth Stenwig Dept. of Pathology Dept. of Pathology Haartman Institute Norwegian Radium Hospital Helsinki University Montebello, NO-0310 Oslo FI-00014 Helsinki Tel +47-22-93 40 00 Tel +358-9-191 24 19 Fax +47-22-93 54 26 Dr. Bodil Bjerkehagen Dr. Henryk Domanski Dept. of Pathology Dept. of Pathology Dept. of Pathology Norwegian Radium Hospital Karolinska Hospital Lund University Hospital Montebello, NO-0310 Oslo SE-171 76 Stockholm Tel +47-22-93 40 00 Tel +46-8-517 751 59 Tel +46-46-17 34 02 Dr. Teddy Holmström Dr. Jeanne M. Meis-Kindblom Dept. of Pathology Dept. of Pathology Helsinki University Sahlgrenska Hospital FI-00014 Helsinki SE-413 45 Gothenburg Tel +358-9-310 66 11 Tel +46-31-342 38 69 Fax +358-9-310 653 42 Euroboss I Handling of specimens
In the case of amputation specimens of skin and soft tissue not infiltrated by the tumor should beremoved and larger blood vessels should be examined for possible tumor thrombosis. In all types ofspecimens, note the relationship between tumor, bone, articular cartilage, epiphysis, surrounding softtissues, and resection margins. Make a statement about the margins (intralesional, marginal, wide,radical) and give measurements in cm. Sample bone marrow from the resection site.
The bone containing the tumor should be sectioned in a plane that will optimally identify residual viable
tumor tissue. The bone should be cut longitudinally into halves with a band saw. Saw an additional
3–4 mm thick longitudinal slice from one of the halves and an additional slice from the other half for a
large section. Take photographs of the sawing lines and slices (Figs. 1–3). Saw the remaining two
halves into 3–4 mm thick slices perpendicular to the above planes and take additional photographs.
Choose the two slides that contain most tumor tissue. If the tumor tissue is very soft, it is better to precut
it with a large knife as deeply as possible before sawing.
Fix the slices overnight and divide the tumor areas into pieces of appropriate size (Fig. 4). Use an ink
pen for drawing the lines on the tissue. Take photographs and make a drawing and number the separate
areas. Saw along the lines and put the pieces in formalin for 1–2 days. Decalcify, if necessary, the
pieces using fast decalcifiers for preservation of tissue. Note that the decalcifying time varies greatly
from piece to piece. Avoid overdecalcification. The pieces should be processed, embedded, cut and
stained using routine methods. The total number of sections depends on the size of the tumor.
Euroboss I Grading of tumor response:
Grading of tumor response by assessment of treatment induced tumor necrosis is mainly based onexamination of tumor specimen from children or young adults treated with neoadjuvant chemotherapyfor highly malignant osteosarcoma. For most trials the cut-off between good and poor responders hasbeen set at around 90% necrosis. In the current protocol less intensive neoadjuvant chemotherapy isgiven and different types of bone sarcomas with an expected poorer response to chemotherapy areincluded. For the actual age group (41–65 years) a potential highly toxic salvage therapy (high-dosemethotrexate) is prescribed. Therefore, the definition of poor histologic response in this protocol isdefined as little or no effect of chemotherapy identified.
Evaluation of prognostic impact of histologic response to preoperative chemotherapy is a secondaryaim of this study. For SSG, tumor necrosis should be assessed according to SSG's modification ofHuvos system.
Four histological grades are used: Unaffected tumor with little or no identified effects Euroboss I Areas of affected tumor tissue detected and one or both of the following criteria:> 10% of examined tumor area reveal unquestionable viable tumor orone or more areas of unaffected viable tumor exceed 2.5 mm in largest diameter < 10% of examined tumor area reveal unquestionable viable tumor and no single area of unaffected viable tumor exceed 2.5 mm in largest diameter No histologic evidence of viable tumor identified within the specimen.
With unaffected means a morphologic appearance closely resembling that of the pre-treatment biopsy.
With unquestionable viable means various degrees of response, including decreased cellularity, and
signs of maturation with bone and cartilage matrix production, but with remaining clearly viable tumor
cells.
Poor response:
Poor response will correspond to a grade I response as defined above.
In the case of several tumor manifestations (i.e. metastatic disease), all resected specimens should beassessed for histologic response. The overall response will be decided by the poorest response reportedat any site.
The location of residual tumor (intramedullary, periostal, soft tissue) should be indicated in the report.
9. Guidelines for surgery
Because of the complexity of the current protocol (aggressive combination chemotherapy and surgery),the whole treatment program should be planned in tumor board, with a pathologist and/or a cytologist,a diagnostic radiologist, an orthopedic surgeon, and an oncologist. Follow-up discussions during thetreatment are necessary.
Surgery is carried out either at the start of treatment or after one cycle of chemotherapy, and within 21days after completion of preoperative chemotherapy, as soon as possible after leucocyte (neutrophiles>1.0 × 109/L and platelets (> 100 × 109/L) recovery. Note that re-evaluation before surgery with chestX-ray and X-ray + CT/MRI of the involved bone are mandatory before surgery.
Biopsy
The location of the diagnostic biopsy is crucial. At later surgery the biopsy tract must be included en
bloc
with the specimen. A misplaced biopsy may greatly complicate the definitive surgery, Hence the
diagnostic biopsy should be planned and performed by the surgeon who will be responsible for the
definitive surgery.
If a coarse needle biopsy is done, location of the biopsy tract has to be tattooed.
Euroboss I Surgery
The surgical planning should be based on clinical data and radiographic examinations (plain radiographs,
scintigraphy, CT, MRI, and possibly angiography) performed before start of chemotherapy and just
before surgery. The surgeon should try to assess the tumor response, feasibility of a limb-saving surgical
procedure, and need for bone-, joint-, vascular-, and soft tissue reconstruction. No fixed guidelines
can be claimed for either the choice or extent of local tumor surgery, nor type of reconstruction.
A conclusion that a tumor is inoperable must always be done at a referral center.
The tumor should preferably be excised with a wide surgical margin. If a limb-saving procedure withwide surgical margin is not possible, an amputation has to be considered. However, a marginal marginmay be acceptable in good responders. Amputation is a surgical procedure and has to be classifiedaccording to the obtained surgical margin.
Surgery of primary tumor: Localized tumor. The aim of the surgery is the removal of the tumor withwide surgical margins. The surgical planning is based on clinical data and radiographic examinationsperformed before start of chemotherapy and before surgery. If a limb salvage surgical procedure withwide surgical margins is not feasible, an amputation is recommended. No fixed guidelines can be claimedfor the type of reconstruction.
Patients with metastases at presentation: In case of unresectable metastatic disease, a surgical rem-oval of primary tumor with inadequate surgical margins can be accepted. When the metastases areconsidered resectable, the surgical procedure must be planned to achieve a wide removal of primarytumor.
Surgery of the metastases: Surgical resection of the metastatic lesions is always recommended whena complete surgery (removal of all evident metastatic disease, with no tumor tissue at the resectionmargins on histologic examination) can be planned on the basis of the radiographic examinations per-formed before surgery. Whenever possible, surgical removal of the primary tumor and metastasesshould be performed at the same time to achieve an early removal of the macroscopic disease, and toreduce the delay in delivering chemotherapy after surgery.
For lung surgery, sternotomy or lateral thoracotomy is left to the discretion of the thoracic surgeon. Incase of unilateral metastases on CT scan, it is recommended to surgically explore the contralateral side.
In case of complete remission of lung metastases on CT scan after preoperative chemotherapy, the lungshould nevertheless be explored surgically.
Evaluation of surgical margin
After intra- or postoperative macroscopical examination of the specimen, where any area with supposed
inadequate margin is marked, the specimen should be cross sectioned serially and margins examined
microscopically (see "Guidelines for pathology").
Surgical margins are defined according to Enneking.
Radical margin: The whole compartment is removed.
Wide margin: A cuff of healthy tissue all around the tumor is included in the specimen.
Marginal margin: The excision is performed close to the tumor capsule and through the reactivezone in one or several planes.
Intralesional margin: ("Debulking surgery"). The excision is in one or several planes performed through Euroboss I 10. Guidelines for radiotherapy
Surgical removal of all tumor tissue at any site should always be attempted and is the only proven wayto achieve local control. In selected cases, however, radiotherapy plays an important role; i.e when thesurgical procedure causes unacceptable mutilation, when the tumor is unresectable for technical and/ormedical reasons, or when the resection margins following surgery are involved judged by histopathologicalexamination. Only tumors of malignancy grade 3 and 4 are included in this treatment proposal.
The patients treated with postoperative irradiation should receive an eq. total dose of 56–62 Gy in2 Gy's fraction where margins are microscopically involved, and 64–70 Gy where macroscopic tumortissue is left behind. For definitive radiotherapy of an inoperable osteosarcoma a radiation dose of70 Gy or more should always be attempted.
Selected institutions may advocate the use of intraoperative electron boost irradiation or brachytherapyby high-dose rate after loading techniques in cases where macroscopic tumor tissues are left behind orwhere the surgical margins obviously are inadequate.
10.1 Practical guidelines for delivery of radiotherapy
10.1.1 Treatment equipment High-energy photons generated by a linear accelerator should be used to treat deep-seatedtumors; with a recommended radiation quality between 5 and 15 MV. If the target volumeexceeds to the surface of the patient, adequate dose coverage should be obtained using bolusmaterial rather than a mixed beam technique.
A three-dimensional computerised dose planning with inhomogeneity corrections shall be per-formed based on CT-scans performed on a flat table top. The CT-accusition must cover allorgans at risk as well as the entire target volume.
A multiple field technique, with optimal beam entry directions and beam weights, should beapplied to obtain a homogeneous dose to the PTV while at the same time minimise the dose toorgans at risk. Normal tissue sparing should be attempted by field shaping using customisedblocks (individually cut) or with a multileaf collimator.
Virtual or conventional simulation procedures are mandatory for all fields. The position of allshielding blocks should be indicated on the simulation films or on BEV-plots as overlays onDDR's.
10.1.2 Treatment equipment and verification • Simulator port films or Beams Eye View-plots (BEV-plots) as overlay on Digital Reconstructed Radiographs (DDR).
• Treatment verification (portfilm or Electronic Portal Imaging (EPI) at the beginning, in the middle, and at the end of the treatment.
• Measurement of entrance dose or other equivalent methods to ensure that the correct calculated dose is given to the patient.
• Radiation treatment charts as well as the dose plan and port films shall be saved.
Euroboss I 10.1.3 Abbreviations (Source: ICRU Report 50) GTV (Gross Tumor Volume) is the gross tumor extention; either palpable or visible/demonstrableby imaging techniques. Its delineation should preferentially be done in collaboration with aradiologist.
CTV (Clinical Target Volume) contains a demonstrable GTV and/or sub-clinical microscopicmalignant disease. Original tumor extension should guide the delineation of CTV. Ideally thisshould be done in collaboration with the treating surgeon. In axial tumors a safety margin of 2cm added to GTV should be attempted. For an extremity osteosarcoma a margin of 4–5 cmmay be advisable.
PTV (Planning Target Volume) is a geometrical concept defined to select appropriate beamsizes and beam arrangements, taking into consideration the net effect of all the possible geometricalvariations. Margins, 0.5–1.0 cm, should be added to the CTV to take into account the effectsof organ and patient movements and inaccuracies in beam and patient set-up in order to ensurethat the prescribed dose is actually absorbed within the CTV.
10.1.4 Fractionation 1.8–2.0 Gy/fraction given as 1 fraction per day and 5 fractions per week. In case of holidaysor machine breakdowns, the overall treatment time might be extended where the indication isadjuvant, whereas in case of macroscopic tumor tissue an accelerated compensation should beconsidered.
10.1.5 Dose homogeneity A maximum dose variation within the PTV between 95–105% according to the dose planshould be attempted. Hot spots outside the PTV with a maximum of 110% are acceptable onlyif total volume is less than 10 cm3. Moreover, single hot spots should not exceed 5 cm3.
10.1.6 Organs at risk & Tolerance doses for normal tissues All structures that may be associated with serious late toxicity should be delineated and dose-volume-histograms generated. The following maximum radiation doses should obtain: Medulla spinalis (more than 5 cm) 45 GyMedulla spinalis (below 5 cm) 50 GyBrain tissue 60 GyN.opticus/chiasma 50 GyIntestine 50 Gy, depending on partial volumeLiver 20 Gy total – 50 Gy if less than ¼ volume is irradiatedKidney 20 Gy (> 1/3)Heart 30 GyLung 20 – 60 Gy depending on partial volumeUrinary bladder 60 Gy Euroboss I 10.2 Follow-up
The standard guidelines pointed elsewhere in this protocol should be followed. Particular emphasis
should be placed on establishing whether a local recurrence represents an "in field" central relapse or
develops outside field or as an "edge" reminence.
CTC-score / RTOG-score vs. SOMA-lent guidelines should preferientally be used for scoring normaltissue complication rates.
10.3 Quality assurance (QA)
Cooperative group multicentre clinical trials require means to guarantee uniformity of treatment procedures
and QA is a prerequisite to evaluate patients entered from various institutions. A major deviation from
the protocol may jeopardise the ability to answer questions addressed in the trial.
10.3.1 Objectives The aim of this QA part of the protocol is to ensure uniformity of all radiotherapy data for eachpatient. Specifically, this means; • To establish a uniform description of the radiation therapy given in terms of volumes, doses and fractionation • To evaluate compliance with the radiotherapy instructions • To help enable a correct evaluation of the endpoints in the trial • To create a platform for evaluation of reactions in healthy tissue, to radiation alone or in combination with the chemotherapy administered 10.3.2 Elements of the QA A final treatment evaluation of each single patient after completion of radiotherapy shall beperformed. A responsible physicist will check the following parameters: • Patient immobilisation • 3D dose planning • Dose prescription according to ICRU 50 and beam calibrations according to the IAEA Technical Report No. 277 or TRS 398 • Fractionation • Volumes of GTV, CTV and PTV • Field data for all fields • Dose distribution in a transversal plane in or close to the centre of GTV • Dose distribution in transversal, sagittal and frontal planes in or close to the centre of PTV • Dose-volume histogram for all defined volumes of interest • Copy of the treatment chart.
A questionnaire shall be filled in by the radiation oncologist at the completion of the treatmentand sent to the Study Secretariat (Oncologic Centre, Lund for SSG-patients).
Name (first & family name)Date of birth (day, month, year) Send this form one week after start of chemotherapy to: SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden This form is a prerequisite for patient eligibility in Euroboss I and should be completed and sent to thesecretariat, together with the following: 1. Anteroposterior and lateral MR of the primary tumor-involved bone.
2. Representative histological slides of the primary tumor.
The above named institution and department(s) commit themselves to participate in the clinicalEuroboss I study and will comply with the scheduled investigations, treatment and follow-up.
The above named institution and department(s) agree to participate in the following research projectsassociated with the Euroboss I protocol: Name and signature of the responsible principal investigator Name (first & family name) Date of birth (day, month, year) Send this form one week after start ofchemotherapy to: SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Hospital and department Enclosed are: 1. MR of primary tumour involved bone
2. Histological representative slides of diagnostic material 3. Institution´s commitment form Histology: .
Start of chemotherapy Name (first & family name)Date of birth (day, month, year) Pathology report I
Send to:SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Fine needle aspiration biopsy Classic osteoblastic osteosarcoma Name (first & family name) Date of birth (day, month, year) Send to:SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Hospital and department Patient data
Duration of symptoms, months (Time interval from first symptom to pathologic confirmation of diagnosis) Metastatic site(s): Soft tissue involvement: Investigations prior to treatment
CT of involved bone: MRI of involved bone: Alkaline phosphatase . . . . . (specify unit) . . . . . (specify unit) Neoadjuvant chemotherapy Adjuvant chemotherapy Type of reconstruction
Vascularized graft Other, specify; .
Type of metastasectomy (lung) bilateral separate through sternotomy Number of metastases removed All known metastases removed Resection (other than lung) No metastasectomy performed Name (first & family name)Date of birth (day, month, year) Pathology report II
Final report including photograph(Polaroid picture or slide) Send to: SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Surgically free of disease Classic osteoblastic osteosarcoma Chemotherapy response: Number of blocks: . . . . . . .
Whole tumor section available: Chemotherapy response: Number of metastases removed: Name (first & family name) Date of birth (day, month, year) Submit this form to: SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Hospital and department Start of treatment, date End of treatment, date Target absorbed dose(s)
metastase(s), specify; .
metastase(s), specify; .
Radiation quality: Specified target dose Number of fractions Dose to clinical organs
Dose modification factors yes, specify; .
Deviation from plan
yes, specify; .
Name (first & family name) Date of birth (day, month, year) Send to:SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Hospital and department • Preoperative chemotherapy or postoperative chemotherapy with
good histopathology response
• Adjuvant chemotherapy
Body surface: . . . . . . . .
100 mg/m2, 48 hours iv 60 mg/m2 , 24 hours iv Ifosfamide 3 000 mg/m2/day, 2 days iv Name (first & family name) Chemotherapy toxicity flow-sheet
Date of birth (day, month, year) Submit this form together with Form 6A to: SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden • Preoperative chemotherapy or postoperative chemotherapy with
good histopathology response
• Adjuvant chemotherapy
Preoperative cycle No: Postoperative cycle No: . . . . . . . .
Urinary electrolytewasting (grade) Depressed level ofconsciousness * According to NCIC CTG expanded common toxicity criteria. See reverse side.
* According to NCIC CTG expanded common toxicity criteria
Urinary electrolyte asymptomatic, not irreversible requiring requiring treatment and managable with requiring i. v.
continued replacement painful erythema, painful erythema, mucosal necrosis and/or erythema, or mild edema, or ulcers, req parenteral or enteral Depressed level of none somnolence or sedation interfering stupor; difficult to with function, but not arouse; interfering with activities of activities of daily Neuropathy-sensory loss of deep tendom objective sensory permanent sensory reflexes or paresthesia loss or paresthesia loss that interferes but not interfering function, but not activities of daily activities of daliyliving Name (first & family name) Date of birth (day, month, year) Send to:SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Hospital and department • Postoperative chemotherapy with poor histological response
Body surface: . . . . . . . .
only cycle 3
Delayed MTX excretion: no, if yes 48 hours MTX serum level only in case of delayed MTX excretion 100 mg/m2, 48 hours iv 60 mg/m2 , 24 hours iv 3 000 mg/m2/day, 2 days iv 8 000 mg/m2, 4 hours iv Name (first & family name) Chemotherapy toxicity flow-sheet
Date of birth (day, month, year) Submit this form together with Form 6A to: SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden • Postoperative chemotherapy with poor histological response
only cycle 3
Urinary electrol. 0 1 2 3 4 0 1 2 3 4 Stomatitis*Depressed level 0 1 2 3 4 0 1 2 3 4 * According to NCIC CTG expanded common toxicity criteria. See reverse side.
* According to NCIC CTG expanded common toxicity criteria
Urinary electrolyte asymptomatic, not irreversible requiring requiring treatment and managable with requiring i. v.
continued replacement painful erythema, painful erythema, mucosal necrosis and/or erythema, or mild edema, or ulcers, req parenteral or enteral Depressed level of none somnolence or sedation interfering stupor; difficult to with function, but not arouse; interfering with activities of activities of daily Neuropathy-sensory loss of deep tendom objective sensory permanent sensory reflexes or paresthesia loss or paresthesia loss that interferes but not interfering function, but not activities of daily activities of daliyliving Name (first & family name) Date of birth (day, month, year) Send to:SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Hospital and department Date of evaluation Physical examination X-ray of the primary tumor site No evidence of disease No evidence of disease after previous relapse Distant metastases Alive with disease Treatment related death Secondary malignancy Dead of other causes If yes, histology .
No. of metastases Treatment for relapse
Palliative intent Name (first & family name) Serious adverse event report
Date of birth (day, month, year) Send to:SSG secretariatRegional Tumor RegistryLund University HospitalSE-221 85 LUNDSweden Hospital and department Physician/investigator Primary diagnosis: Category of event: Description of event (including relevant history/lab data/test): Start of infusion Treatment for event:

Source: http://radium.no/bruland/PDF/Euroboss1.pdf

1301_eam_newsletter_v1_en.indd

ERSTE The ESG Letter January, February/2013 Edition RESPONSIBLE RETURN INVESTMENT BOARDHealthcare provision – a basic need not always met ENGAGEMENTAccess to Medicine COMPANY OF THE MONTHGlaxoSmithKline PLC PIN IT DOWNBitter pills & healthy capitalists? ERSTE ASSET MANAGEMENT 02 When we wish each other a Happy New Year – as was the case just a few days ago – then the topic of health will almost always be at the forefront of our minds. As human beings, being healthy and staying healthy is accorded a high priority. The necessary healthcare provision is good to very good in large parts of Europe, despite the fact that even in these countries a debate about a „two-tier society in medical care" keeps emerging from time to time.

Lc/ms/ms analysis of the endogenous dimethyltryptamine hallucinogens, their precursors, and major metabolites in rat pineal gland microdialysate

Received: 20 March 2013, Revised: 20 May 2013, Accepted: 23 May 2013 Published online in Wiley Online Library (wileyonlinelibrary.com) DOI 10.1002/bmc.2981 LC/MS/MS analysis of the endogenousdimethyltryptamine hallucinogens, theirprecursors, and major metabolites in ratpineal gland microdialysate Steven A. Barkera*, Jimo Borjiginb, Izabela Lomnickaa and Rick Strassmanc