• No results found

University of Groningen Survivorship care after testicular cancer Boer, Hindrik

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Survivorship care after testicular cancer Boer, Hindrik"

Copied!
29
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Survivorship care after testicular cancer

Boer, Hindrik

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Boer, H. (2019). Survivorship care after testicular cancer: New insights in late effects of treatment and approaches to shared-care follow-up. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Long-term and late effects of germ-cell

testicular cancer treatment and

implications for follow-up

Chapter 2

2

Hege S. Haugnes1,2, George J. Bosl3, Hink Boer4, Jourik A. Gietema4, Marianne Brydøy5,6,

Jan Oldenburg7, Alv A. Dahl7,8, Roy M. Bremnes1,2, Sophie D. Fosså7,8.

1Department of Oncology, University Hospital of North Norway, Tromsø, Norway 2Institute of Clinical Medicine, University of Tromsø, Norway 3Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, USA 4Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands 5Section of Oncology, Institute of Medicine, University of Bergen, Bergen, Norway 6Department of Oncology, Haukeland University Hospital, Bergen, Norway 7Division of Cancer Medicine and Radiotherapy, Oslo University Hospital, Norway 8Medical Faculty, University of Oslo, Norway

(3)

Abstract

Germ-cell testicular cancer (TC) represents a malignancy with very high cure rates. After the introduction of cisplatin-based chemotherapy in the late 1970s, the 5-year survival rate has increased considerably, and is currently above 95%. Since TC usually is diagnosed before the age of 40, these men can expect to live for another 40-50 years after being successfully treated. This success, however, is hampered by an increased risk of long-term and late effects of treatment. Secondary malignant neoplasms and cardiovascular disease represent the most common potentially life-threatening late effects typically occurring more than 10 years after treatment. Other long-term effects include pulmonary toxicity, nephrotoxicity, neurotoxicity, decreased fertility, hypogonadism and psycho-social problems. The incidence and time to onset of these various side-effects vary according to treatment type and intensity. There is still little knowledge about underlying mechanisms and genetic susceptibility of the various adverse effects. Apart from treatment burden, it is not yet possible to identify patients who are at high risk for certain late effects after TC treatment.

In this clinical review, we present the current status regarding different somatic and psychosocial long-term late effects after treatment for TC, based on Medline searches and our own research. Moreover, we postulate recommendations for general medical evaluations that should begin after treatment is completed and continue during follow-up.

(4)

Introduction

The treatment of germ-cell (GC) testicular cancer (TC) represents a medical success story with today’s 5-year survival rates exceeding 95%.1 The improved prognosis is primarily due to the

effectiveness of cisplatin-based chemotherapy in the treatment of advanced disease, timing of surgery to remove residual disease, and the establishment of effective salvage regimens.2

TC affects mostly young men at their peak of family life, reproduction, education and career, with a life expectancy of 40-50 years after successful cancer treatment. Consequently, there has been an increased attention towards long-term effects of cancer treatment during the last decade, which has resulted in attempts to decrease treatment-related toxicity while maintaining the high cure rates.3 Despite these attempts, treatment-related toxicity has emerged

as an important issue for this young patient population. But since studies of late effects of cancer treatment are retrospective in nature, it should be pointed out that the current knowledge represents complications of treatment administered several years to decades ago.4

Long-term survivors are defined as individuals who are disease-free 5 years or more after primary treatment.4 Some side-effects of cancer treatment develop during treatment

and may persist during follow-up, e.g. peripheral neuropathy. These side-effects are referred to as long-term effects, while late effects become manifest months to years after completion of treatment.4 Secondary non-GC cancers and cardiovascular disease are examples of late effects.

These represent the most serious and potentially life-threatening effects of cancer treatment. Apart from treatment burden, it is presently not possible to identify patients at high risk for certain somatic long-term and late effects of TC treatment. A recent review paper on TC survivorship has recommended specific future research directions to elucidate the underlying mechanisms of the long-term and late effects.5

Based on Medline searches and our own research, we herein summarize the current knowledge regarding the different somatic and psychosocial long-term and late effects after TC treatment. We postulate recommendations for maintaining health and general medical evaluations that should begin after treatment and continue beyond the 10-year follow-up. Methods

Medline databases were used to identify original articles and reviews on long-term and late effects of TC treatment. The phrases used during the search process included “testicular cancer” or “germ cell cancer” combined with “survivors”, “late effects”, “morbidity”, or “long-term effects” and phrases reflecting the different toxicities (e.g. “myocardial infarction”, “cardiovascular disease”, “hypertension”). Articles published during the past two decades were preferentially included. Only articles published in English until 31st of December 2011 were reviewed. The majority of

included publications were based on participants who were in a durable complete remission, preferably long-term survivors as previously defined. The list of references was reviewed by all authors of this review.

(5)

Most studies on late effects of cancer treatment have been retrospective in nature.4 Thus, no

prospective randomized trials on late effects from testicular cancer treatment have been published.

Secondary malignant neoplasms

The development of a non-GC second malignancy represents one of the most severe late effects following treatment for TC. In addition,1-5% of TC survivors (TCSs) develop a contralateral TC6

which probably reflects common etiologic factors. A contralateral TC is not regarded a late effect of treatment and we make no further comment on its origin or management.

In a series of 40,576 TCSs, the overall post-TC observed/expected (O/E) ratio for developing a solid second cancer was 1.55 (95% confidence interval [CI] 1.48-1.62) in 10-year survivors,7 which is in

agreement with other large population-based studies (Table 1).8, 9 The O/E ratio was 2.6 (95% CI

2.1-3.2) for leukemia, mostly acute myeloid leukemias (AML) and lymphoblastic leukemias.(10) Concern has been raised regarding the association between etoposide and AML. However, the absolute risk of etoposide-induced AML after four cycles or less of standard chemotherapy is very low, and was reported to be less than 0.2% in chemotherapy-treated patients.10 The risk

of etoposide-induced AML is probably dose-dependent, and after high-dose etoposide the risk of AML has been ranging from 0.5% to 2.6% in several studies.11-13 Therapy-related leukemia is

usually diagnosed within the first 10 years after treatment for TC.14 Solid second malignancies are

generally diagnosed after a latency of at least 10 years and may be induced by both radiotherapy and chemotherapy (Table 1).7-9

Most radiation-induced malignancies are located within or close to initial abdominal radiation fields (bladder, stomach, pancreas and colon cancer). The statistically increased relative risks for solid cancers after cytotoxic treatment (Table 1) are to a great extent based on “old fashioned” cytotoxic treatment with larger radiotherapy fields and higher radiation and chemotherapy doses than used today.15 The role of modern risk-adapted treatment on the late development of a solid

second cancer is less clear.

The radiation exposure from repeated CT scans during follow-up could theoretically contribute to an increased risk for solid cancers.16, 17 Theoretical estimates of lifetime cancer risk range from

1.9% for an 18-year old to 1.2% for a 40-year old patient during surveillance for stage I TC,16 but

these are estimated phantom data, not patient data, and should be interpreted with caution. These hypotheses, based on extrapolations from Hiroshima and other data, are not supported at this time by clinical studies that demonstrate that such risk exists. A recent publication by van Walraven et al did not find any association between radiation exposure and risk of second cancers in 2569 men after treatment for low-grade TC, but the follow-up time was only median 11.2 years.18 Hence, given the very low risk, the relative risk/benefit should be considered when

making decisions about the use of CT in patient care. Some guidelines recommend MRI instead of CT during follow-up to reduce the risk of radiation-induced cancer, but there is no definitive study at this time.

(6)

Table 1.

Sec

ond c

anc

ers (solid tumors and leuk

emias) in long-t erm t esticular c anc er sur viv ors ( TCSs). eviations: No , n

umbers; gen pop

, gener al population; y , y ears; R T, r adiother ap y; CT , chemother ap y; RR, r elativ

e risk SIR, standar

diz ed incidenc e r atio; EAR, ex cess absolut e risk

bl

es

c

ha

pt

er

2

bl e 1. Se co nd c an ce rs (s ol id tu m or s an d le uk em ia s) in lo ng -te rm te sti cu la r c an ce r s ur vi vo rs (T CS s) . Fi rs t a ut ho r Ye ar o f di ag no si s No . o f p at ie nt s Pa ti en t c ha ra ct er is ti cs Ri sk o f s ec on d ca nc er co m pa re d w it h ge n po p Ma in re su lts Tr av is (2 00 5) (7 ) 1943 -20 01 40 57 6 20 98 4 (1 0-y su rvi vo rs ) Ch em ot he ra py Ra di ot he ra py CT a nd R T Da ta fr om po pu la tio n-ba se d c an ce r r eg is tr ie s. 1. 8 (1 .3 -2. 5) (R R) 2. 0 (1 .9 -2. 2) 2. 9 (1 .9 -4. 2) TC Ss a re a t s ig ni fic ant ly inc re as ed ri sk o f s ol id c anc fo r a t le as t 3 5 ye ar s af te r t re at m en t. The ri sk fo r s ec on d ca nc er d ec re as ed w ith inc re as ag e at te st ic ul ar c an ce r d iag no sis . Ca nc er s of th e lu ng (R R 1. 5) , c ol on (R R 2. 0) , b la dd 2. 7) p an cr ea s (R R 3. 6) a nd s to m ac h (R R 4. 0) a cc ou fo r 6 0% o f t he to ta l e xc es s. Ri ch ia rd i ( 20 06 ) (8 ) 1943 -20 00 29 51 1 Da ta fr om po pu la tio n-ba se d c an ce r r eg is tr ie s. No tr ea tm en t d et ail s w er e in cl ud ed . Ov er al l S IR w as 1 .6 5 ( 1. 57 -1. 73) fo r s ol id tu m or s af me di an 8 .3 y ea rs fo llo w -up . S IR s fo r m os t s ol id c an ra ng ed b et w ee n 1 an d 2, a nd in cre as ed w ith in cre fo llo w -up . Va n den B el t-Du se bo ut (2 00 7) (9 ) 1965 -19 95 2707 (5 -y su rvi vo rs ) Su rg er y on ly Ra di ot he ra py Ch em ot he ra py RT a nd C T 0. 7 (0 .4 -1. 3) (SI R) 1. 7 (1 .5 -2. 0) 1. 4 (0 .9 -2. 1) 3. 0 (2 .0 -4. 4) Ov er al l S IR w as 1 .7 (1 .5 -1. 9) a fte r m ed ia n 17. 6 ye ar fo llo w -up . T he ri sk o f s ec on d ca nc er w as 2 .6 (1 .7 -4. af te r i nf rad iap hr ag m at ic R T an d 2. 1 (1 .4 -3. 1) a fter ch em ot he ra py in co m pa ris on to s ur ge ry o nl y. Ho w ar d (2 00 8) (1 0) 1943 -20 01 42 72 2 (1 -y su rvi vo rs ) Su rg er y on ly Ra di ot he ra py Ch em ot he ra py RT a nd C T Da ta fr om po pu la tio n-ba se d c an ce r r eg is tr ie s. 1. 6 (0 .8 -2. 9) (O /E ) 2. 7 (1 .9 -3. 6) 3. 9 (1 .8 -7. 3) 4. 5 (0 .7 -13. 8) Se co nd ar y le uk em ia d ev el op ed in 89 pa tie nt s, w EAR a t 1 0. 8. p er 10 0. 000 pe rs on ye ar s. St at is tic al ly si gn ifi ca ntl y el eva te d ris ks w er e ob se rve d fo r a cu my el oi d le uk emi a an d ac ut e ly mp ho bl as tic le uk emi Ex ce ss c um ul at iv e le uk em ia ri sk w as 0 .2 3% b y 30 af te r T C di ag no sis . br ev ia tio ns : N o, n um be rs ; g en p op , g en er al p op ul at io n; y , y ea rs ; R T, ra di ot he ra py ; C T, c he m ot he ra py ; R R, re la tive ri sk S IR , s ta nd ar di ze d in ci de nc e ra tio ; E A R, e xc es s ab so k

(7)

Cardiovascular disease and Raynaud’s phenomenon Cardiovascular disease

Case reports on potentially life-threatening cardiovascular disease (CVD) during or shortly after treatment date back to the 1980s.19 The frequency of late cardiac morbidity among TCSs was

reported for the first time in 2000 by Meinardi et al. among 87 patients aged ≤50 years at the time of analysis.20 Subsequent reports on CVD in TCSs estimated the magnitude of the problem. Zagars

et al. reported on cardiac mortality in survivors of testicular seminoma treated with radiotherapy only.21 Of note, the cardiac-specific standardized mortality ratio (SMR) was first after 15 years of

follow-up significantly elevated at 2.02 (99% CI 1.22-3.16).21 A large international study by Fosså et

al. in 38,907 at least one-year TCSs reported a SMR for all circulatory diseases of 1.58 (95% CI 1.25-2.01) in men who received chemotherapy after 1975.22

Table 2 summarizes the prevalence of coronary artery disease (CAD) found in recent studies, subdivided in different treatment groups.20, 23-25 In a large nationwide cohort study

performed by Van den Belt-Dusebout et al,24 non-seminoma survivors younger than 45 years

had a significantly increased standardized incidence ratio for myocardial infarction (MI) at 2.06 (95% CI 1.15-3.41). A substantial part of the analysis was based on patients treated with regimens no longer used, thus supplementary studies are needed to ascertain the burden of the current standard regimens. A recent Norwegian study reported a significantly increased risk of CAD after bleomycin, etoposide and cisplatin (BEP) in comparison to age-matched controls, but this publication did not include mortality.25

Mechanisms behind treatment-induced CVD are not yet accurately elucidated. Both direct vascular damage and indirect effects through gradual development of CVD risk factors26

may contribute to the eventual cardiovascular damage. A significantly higher prevalence of hypertension was found in chemotherapy-treated TCSs, while diabetes was more prevalent after radiotherapy alone or in combination with chemotherapy, as compared with patients treated with other modalities and healthy controls.25, 27 In particular, the clustering of CVD risk factors into

the metabolic syndrome28 might be the mediating link between cytotoxic treatment and CVD.

Patients treated with cisplatin-based chemotherapy more often develop the metabolic syndrome in comparison to controls or other treatment groups.29, 30

Inflammation and endothelial dysfunction play an important role in the pathogenesis of atherosclerosis.31 Microalbuminuria, and circulating levels of von Willebrand factor and

plasminogen activating inhibitor 1 (PAI-1), markers representing endothelial dysfunction, are all significantly more prevalent in chemotherapy-treated TCSs.32 Cisplatin is detectable in serum

several years after administration,33, 34 and it is hypothesized that small amounts of cisplatin may

continuously stimulate the endothelium. However, there are no data to support that hypothesis. C-reactive protein was recently proposed as a potential marker for cardiovascular disease in TCSs,35 suggesting chronic endothelial activation.

(8)

Raynaud’s phenomenon

Many patients experience Raynaud’s phenomenon (RP) both during and after treatment.36 This

typical white discoloration of the digits and subsequent pain and redness is prevalent in 15-45% in chemotherapy-treated TCSs several years after treatment.36-39 The phenomenon is thought to

be a consequence of direct vascular damage in the digital arteries,40 but neurologic effects may

also be involved in the abnormal vasoregulation causing vasospasms.41 Bleomycin is considered

(9)

Ta bl e 2. R isk o f c or on ar y ar te ry d is ea se (C A D ) i n lo ng -te rm te sti cu la r c an ce r s ur vi vo rs (T CS s) . Ab br ev ia tio ns : N o, n um be rs ; f u, fo llo w -up ; y , y ea rs ; SI R, s ta nd ar di ze d in ci de nc e ra tio ; M I, m yo ca rd ia l i nf ar ct io n; A P, a ng in a pe ct or is ; C T, c he m ot he ra py ; R T, ra di ot he ra py ; R R, re la tiv e ris k; C VB , c is pl at in , v in bl as tin e an d bl eo my ci n; B EP , b le omy ci n, e to po si de a nd c isp la tin ; H R, h az ard ra tio ; n s, no t s ig ni fic an t; CV D , c ar di ov as cu la r d ise as e. *I nc lu si on c rit er io n: a ge ≤ 50 at ti m e of a na ly sis . †T he e nd po in t w as “c ar di ac e ve nt ” d ef in ed a s (1 ) p at ie nt d ie d fro m a M I o r s imi la r c ar di ac re la te d ep iso de ; ( 2) a ng in a or M I re po rt ed o n G P fo rm s; (3 ) c ar di ac a bn or m al ity o n th e as se ss m en t f or m at th e lo ng -te rm fo llo w -up c lin ic v isi t; (4 ) a ng ina o r c he st p ai n re po rt ed o n cl ini c vi si t; (5 ) c ar di ac su rg er y fo r c or ona ry a rt er y di se as e. Fi rs t au th or Ye ar o f di ag no si s No . o f pa tie nt s Me dia n fu (r an ge ), y Tr ea tm en t c ha ra ct er is tic s St at is tic al me th od Ri sk o f C A D (9 5% C I) Co m m en ts Me in ar di (2 00 0) (1 6) 1977 -19 87 87* (1 0-y su rvi vo rs ) 14 (10 -20 ) A ll tr ea te d w ith c isp la tin -co nt ai ni ng th er ap y SI R 7. 1 (1 .9 -18. 3) Tw o TC Ss w ith M I a nd thr ee pa tie nt s w ith A P w ith pr ov en my oc ard ia l i sc he mi a. Hu dd ar t (2 00 3) (1 9) 1982 -19 92 992 10 (0 -20) Su rg er y on ly Ra di ot he ra py C he m ot he ra py CT a nd R T RR 1. 0 (re fe re nc e)† 2. 4 (1 .0 -5. 4) † 2. 6 (1 .2 -5. 8) † 2. 8 (1 .1 -7. 1) † No re st ric tio ns re ga rd in g fo llo w -up du ra tio n. R R adj us te d fo r a ge . “C ar di ac e ve nt ” a s m ea sur ed en dp oi nt , s ee def in iti on b el ow . Va n den Be lt-Du se bo ut (2 00 6) (2 0) 1965 -19 95 2339 (5 -y su rvi vo rs ) 18 (5 -38) Su rg er y on ly RT in fra di ap hr ag m at ic (i nf) RT m ed ia st in um (m ed ) Ch em ot he ra py CT a nd R T in f CT a nd R T m ed SI R Treatm ent gr ou ps 0. 8 (0 .5 -1. 2) 0. 7 (0 .6 -0. 9) 1. 6 (1 .1 -2. 4) 1. 2 (0 .8 -1. 6) 1. 3 (0 .7 -2. 3) 2. 3 (1 .2 -3. 9) MI e ve nt s > 5 ye ar s af te r d ia gn osi s. Ca rd io va sc ul ar d at a w er e ob ta in ed fro m m ed ic al re co rd s an d th ro ug h qu es tio nn air es s en t t o pa tie nt s' ge ne ra l p ra ct iti on er s. Co m pa re d wi th a ge -ma tc he d co nt ro ls . <4 5 ye ar s 45 -54 ye ar s ≥ 54 ye ar s SI R Attain ed ag e 1. 6 (1 .0 -2. 5) 1. 5 (1 .1 -2. 0) 0. 9 (0 .7 -1. 1) Ha ug ne s (2 01 0) (2 1) 1980 -19 94 990 (1 3-y su rvi vo rs ) 19 (13 -28 ) Su rg er y on ly Ra di ot he ra py Ch em ot he ra py (CV B al on e) Ch em ot he ra py (B EP a lo ne ) CT a nd R T HR ns ; no t r ep or te d ns ; no t r ep or te d ns ; no t r ep or te d 3. 1 (1 .2 -7. 7) 4. 8 (1 .6 -13. 9) MI > 2 ye ar s af te r di agn os is . F at al ev en ts ex cl ud ed . A ll sel f-re po rt ed CV D v al id at ed b y us e of m ed ic al re co rd s. Co mp are d w ith a ge -ma tc he d co nt ro ls . Table 2. R isk of c or onar y ar ter y disease ( CAD) in long-t erm t esticular c anc er sur viv ors ( TCSs). Abbr eviations: No , n umbers; fu , follo w -up; y, years; SIR, standar diz ed incidenc e ratio; MI, my oc ar dial inf ar ction; AP , angina pect oris; CT , chemother ap y; RT , r adiother ap y; RR, relativ e risk ; CVB

, cisplatin, vinblastine and bleomy

cin; BEP

, bleomy

cin, et

oposide and cisplatin; HR, haz

ar d r atio; ns , not signi fic ant; C VD , c ar dio vascular disease . *Inclusion crit erion: age ≤ 50 at time of analysis . †T he endpoint w as “c ar diac ev ent ” de fined as (1) patient died from a MI or similar car diac relat ed episode; (2) angina or MI repor ted on GP forms; (3) car diac abnormalit y on the assessment

form at the long-t

erm follo

w

-up clinic visit; (4) angina or chest pain r

epor

ted on clinic visit; (5) c

ar diac sur ger y for c or onar y ar ter y disease .

(10)

Pulmonary toxicity

Pulmonary toxicity was identified as the major dose-limiting side-effect of bleomycin treatment already in the 1970s.42, 43 Bleomycin may cause pneumonitis, occasionally progressing to

pulmonary fibrosis during or shortly after administration of bleomycin. As there are no agreed criteria to define bleomycin pulmonary toxicity (BPT), the long-term prevalence of patients with non-fatal BPT varies between 7% and 21%.44-47 Fatal BPT has been reported to occur in 1-3% of

patients treated with bleomycin.44, 48 In a British study where 835 patients were evaluated median

7.4 years following treatment, multivariate analyses showed that decreased renal function (hazard ratio [HR] 3.3), age above 40 years (HR 2.3), initial stage IV disease (HR 2.6) and cumulative bleomycin dose >300 000 IU (300 units) were associated with increased risks of BPT.44 Interestingly,

the bleomycin hydrolase genotype was not found to be associated with the development of BPT or with changes in pulmonary function tests.47

Prior studies evaluating pulmonary function after treatment for TC concluded that possible reductions in the pulmonary function during or shortly after treatment were normalized during follow-up.43, 49-52 In a Norwegian study reporting long-term spirometry data from 1.049

TC survivors after median 11 years follow-up, men treated with large cumulative cisplatin doses and/or pulmonary surgery had significantly decreased spirometry variables compared with men treated with surgery only.53 Only cisplatin dose (p=0.007) and age (p=0.008) were significantly

associated with restrictive lung disease in multivariate analyses that included cumulative bleomycin (maximum: 360 000 IU/360 units), etoposide and vinblastin doses. The reduced pulmonary function was subclinical in the majority of these patients.

Population-based epidemiological studies have shown an association between pulmonary function and all-cause mortality.54, 55 Furthermore, TCSs cured by chemotherapy

after 1975 are at a 2.5-fold increased risk to die of respiratory disease compared with the normal population 22. The lungs are highly vascularized organs, and chemotherapy-induced endothelial

dysfunction is a possible mechanism that is involved in the reduced pulmonary function.56

Nephrotoxicity

Nephrotoxicity is a well-recognized acute and long-term effect of both radio- and chemotherapy for TC.57, 58 Studies in animals and humans have shown that cisplatin damages the proximal and

distal tubular epithelium and collecting ducts, and in the rat it also produces glomerular damage at the highest doses.59, 60 A statistically significant increase in serum creatinine and a decrease

in serum magnesium (Mg) from baseline measurements are often detectable after 4 cycles of standard cisplatin-based therapy.61, 62 Mg loss may lead to hypomagnesemia, hypokalemia,

and hypocalcemia. Less commonly, cisplatin may also induce sodium wasting and proteinuria, particularly after high cumulative cisplatin doses.63-67 Studies also show that neither serum

creatinine nor calculated creatinine clearance based on 24h urine collections is a sensitive measure of renal damage.66, 68, 69 The severity of renal damage during cisplatin treatment can

(11)

Pre-existing intrinsic renal disease augments cisplatin nephrotoxicity, as does the co-administration of other nephrotoxic drugs.

The acute nephrotoxic effects of cisplatin may be partly, but not completely, reversible. Long-term studies of renal function show persistent changes from baseline values years after completion of treatment, with little evidence of improvement over time.57, 58, 71, 72 The increased prevalence

of loss of albumin in the urine (microalbuminuria) with increasing follow-up duration may be considered a sign of vascular damage within the kidney.32 The clinical implications of these

changes are unclear, but they may possibly contribute to a reported increase in cardiovascular toxicity, including hypertension and myocardial infarction observed in patients a decade or more after cisplatin-based treatment.23-25 Renal artery stenosis several years after para-aortic irradiation,

resulting in severe hypertension, has been reported in some case reports.73, 74 Thus, men who

develop severe hypertension after abdominal irradiation should be investigated for renal artery stenosis.

In one report, significant increases in serum renin and aldosterone levels were reported in a majority of normotensive patients at a median of 26 months (range 9-54 months) after treatment.71

More recent data have shown a relationship between reduced glomerular filtration rate and the presence of microalbuminuria, with an increased risk of cardiovascular and all-cause mortality in a general population, and support an association between decreased renal function and risk of sudden cardiac death.75-77

Neuro- and ototoxicity Peripheral neuropathy

Cisplatin-induced neuropathy is typically sensory neuropathy of the distal limbs, with paresthesias being the core symptom. Cisplatin causes degeneration of large dorsal root ganglia neurons with loss of myelinated fibres, both distally in the limbs and proximally in the spinal cord, although other mechanisms may be involved as well.78, 79

The reported incidence of persistent peripheral neuropathy varies according to applied treatment and dose as well as method of assessment. A frequency of 20-40% has typically been stated.80 Three large studies with long follow-up have been published during recent years.37-39

In the Norwegian cohort, 28% of TCSs who received ≤4 cycles of cisplatin-based chemotherapy reported paresthesias, as opposed to 46% following ≥5 cycles and 10% after orchiectomy alone.37

Rossen et al reported similar findings (chemotherapy: 31%; surveillance: 9%),38 while peripheral

neuropathy was somewhat less frequent in the study by Glendenning et al. where 1/3 had received carboplatin-based chemotherapy.39 Paresthesias of the lower limbs may also be associated with

abdominal radiation.37 In severe cases, symptomatic treatment might be tried, but an effective

treatment has not been documented.81

Ototoxicity

Cisplatin-induced ototoxicity represents a distinct feature of cisplatin’s side effects and is presumably caused by selective damage to the outer hair cells of the cochlea,82 leading to tinnitus Chapter 2

(12)

and hearing impairment, typically affecting the high frequencies. Reported persistent ototoxicity varies widely with method of investigation, and applied treatment. Bokemeyer et al. reported persistent ototoxic symptoms in 5 to 65%, depending on cisplatin dose.83, 84 Ototoxic symptoms

beyond 5 years following cisplatin-based treatment have been reported in 21-24 % of TCSs.37, 38

The five-day BEP regimen seems preferable to the three-day regimen in regard of minimizing ototoxicity, particularly if 4 cycles are administered.37, 85

The considerable inter-individual variations observed may be due to polymorphisms in glutathione S-transferases,86, 87 or other candidate genes associated with cisplatin-induced

neuro-and ototoxicity.88, 89

Cognitive function

Cognitive function in TCSs has been addressed in a few studies 1-11 years following TC treatment.85, 90-93 Although some report cognitive complaints (self-reported) to be rather common irrespective

of cancer treatment or to increase following chemotherapy (≤4 cycles),92, 93 a permanent decline in

neuropsychological performance has not been documented in any study. Cognitive complaints have rather been related to emotional distress and fatigue.

Avascular necrosis

Testicular cancer survivors treated with chemotherapy seem to have an increased risk of developing avascular necrosis (osteonecrosis) compared to patients treated for other solid tumors (REF Shim Drug Safety 2008).94 The exact reason for this vulnerability is not known, but

is probably multifactorial. Most reported cases have also received corticosteroids. The femoral head is most commonly affected, often bilaterally.94, 95 Based on three reports, Winquist et al

estimated an overall crude incidence of 1.5% following chemotherapy for testicular cancer. Cook et al reported an overall prevalence of 3,8% in 103 consecutive patients invited to attend an MRI scan of the hips.96 Avascular necrosis should be suspected in patients presenting with pain in the

inguinal region (or upper thigh/buttocks). A limp and decreased hip motion may also be present. If suspected, an MRI should be performed in case of normal plain x-ray, and the involvement of an orthopedic surgeon in the management of these patients is imperative.94

Hypogonadism

Subnormal testosterone values have been reported in TCSs treated with chemotherapy compared to healthy controls,30 and following more than four chemotherapy cycles,97, 98 or combination of

chemotherapy and RT when compared to men treated with surgery only.99 Subclinical Leydig cell

dysfunction with increased LH has been found in several studies.30, 84, 97, 98, 100-102

In long-term follow-up studies where similar reference levels for testosterone were applied (<10nmol/l or <2.6ng/ml), overall 12%-16% had subnormal testosterone values or used androgen replacement therapy (ART).97-99 Similar estimates were found in the Norwegian cohort,

(13)

Figure 1. Fraction of men with “manifest or subclinical” hypogonadism according to

treatment in the Norwegian cohort. Dark blue (lower): Used androgen replacement therapy (ART) at follow-up (manifest hypogonadism); Medium blue (middle): low s-testosterone (<10nmol/l) at follow-up (manifest hypogonadism); Light blue (upper): Elevated s-LH (≥12 IU/l) and normal s-testosterone at follow-up (subclinical hypogonadism). (P= .014 for manifest hypogonadism (ART or low testosterone) versus normal testosterone, P<.001 for any hypogonadism (ART, low testosterone and/or elevated LH) versus no hypogonadism). RT, radiotherapy; ≤850 mg, cumulative cisplatin dose ≤850 mg; >850 mg, cumulative cisplatin dose >850 mg. Surgery RT £ 850 mg > 850 mg Le ve ls of h yp og on ad is m (% ) 0 5 10 15 20 25 30 35

where overall 15%, ranging from 8% to 18% depending on treatment, of men younger than 65 years had hypogonadism by this definition (testosterone <10 nmol/l or used ART) (Fig 1, p=.014) (personal communication, M Brydøy).

Chapter 2

Figure 1. Fraction of men with “manifest or subclinical” hypogonadism according to treatment in the Norwegian cohort. Dark blue (lower): Used androgen replacement therapy (ART) at follow-up (manifest hypogonadism); Medium blue (middle): low s-testosterone (<10nmol/l) at follow-up (manifest hypogonadism); Light blue (upper): Elevated s-LH (≥12 IU/l) and normal s-testosterone at follow-up (subclinical hypogonadism). (P= .014 for manifest hypogonadism (ART or low testosterone) versus normal testosterone, P<.001 for any hypogonadism (ART, low testosterone and/or elevated LH) versus no hypogonadism). RT, radiotherapy; ≤850 mg, cumulative cisplatin dose ≤850 mg; >850 mg, cumulative cisplatin dose >850 mg.

(14)

Low testosterone levels are significantly associated with increased prevalence of the metabolic syndrome(103) and an increased risk for CVD mortality as well as respiratory disease mortality in epidemiological studies.104, 105 Subclinical hypogonadism might also enhance the risk of

osteoporosis, although data on this subject are inconclusive.106, 107

The relatively high frequency of Leydig cell dysfunction implies that the hormonal status of TC survivors should be regularly assessed. However, there is currently no evidence supporting testosterone replacement therapy to prevent late effects such as CVD, and treatment decisions should be guided by clinical symptoms.(108)

Fertility and sexuality Fertility

At long-term follow-up of cohorts of TC survivors with known intentions to conceive a child, overall post-treatment conception and paternity rates vary from 49% to 82% (Table 3).99, 109-112

Some investigators report an association between the likelihood of paternity with cumulative chemotherapy dose,110, 113 but preservation of antegrade ejaculation following RPLND is the most

critical factor for conception without use of cryopreserved sperm.110, 111 After primary RPLND,

Beck et al. recently reported that 99% preserved antegrade ejaculation after nerve-sparing surgery,114 higher than 91-93% previously reported.115, 116 After nerve-sparing post-chemotherapy

RPLND, 71-89% retain antegrade ejaculation,116-119 compared to 25% following full bilateral

(15)

Table 3. Post tr eatment conc eption and pat ernit y r at es among testicular cnc er (T C) sur viv ors with kno wn int entions to conc eiv e a child follo wing TC tr eatment (donor insemination ex cluded)* *Numbers ar e post-tr eatment when not speci fied as pr e-tr eatment (pr e). Abbr eviations: No , n umbers; fu , follo w -up; y, years; RPLND , r etr operit oneal lymph node dissection; CT , chemother ap y; R T, r adiother ap y; FSH, follicle stim ulating hormone . †Without infer tilit y tr eatment. T he use of cr yopr eser ved semen w as not speci fied . ‡Both chemother ap y gr

oups included patients additionally tr

eat ed with R T or RPLND . § R at es r efer t o 15-y actuarial r at es . Ta bl e 3. Po st tr eat m en t c on ce pt io n an d pat er ni ty rat es am on g te st ic ul ar c anc er (T C) s ur vi vo rs w ith kno w n int ent io ns to c onc ei ve a c hi ld fo llo w ing T C tr ea tm ent (do no r in se m in at io n ex cl ud ed )* . Fi rs t a ut ho r No . o f T C su rv iv or s in cl ud ed Me dia n fu (r an ge ), y Tr ea tm en t c ha ra ct er is tic s Co nc ep tio n/ pa te rn ity ac co rd in g to tr eat m en t Ma in re su lts Sp er m on (20 03) (1 03 ) 88 (12 0 pr e) 7 (2 -19) Su rv ei lla nc e RP LN D RT ≤4 CT c yc le s ≤ 4 CT c yc le s + R PL N D (p rim ar y or s ec on da ry ) No s ig nif ic an t d iff er en ce s be tw ee n t re at m en t g ro ups . 59% (52/ 88 ) f at he re d fo llo w in g TC tr ea tm en t, in cl ud ing cr yo pr es er ve d se m en in 7 . 4 3% (3 8/8 8) co nce iv ed w ith in on e ye ar of a tt em pt s. Pr ior to TC tr ea tm en t, 78 % (9 3/ 12 0) fa th er ed , 6 6% (7 9/ 12 0) c on ce iv ed w ith in o ne y ea r o f atte m pts . Hu yg he (2 00 4) (1 00 ) 16 4 (2 28 pr e) 8 (3 -27) RT 2-4 ch em ot her ap y cy cl es 67% (11 0/ 164) c on ce iv ed fo llo w ing T C tr ea tm ent co m pa re d to 9 1% (2 08 /2 28 ) p rio r t o TC . Cu m ul at iv e co nce pt io n ra te s lo w er fo llo w in g RT th an c he m ot he ra py . (P < 0. 01 ). Hu dd ar t ( 20 05 ) (9 0) 207 A t l ea st 5 y Su rv ei lla nc e Ch em ot he ra py RT Ch em ot he ra py /R T 88% (8 5% †) co nc ei ve d 75% (71% †) c on ce iv ed 85% (82% †) c on ce iv ed 83% (67% †) c on ce iv ed 82% (16 9/ 207) c on ce iv ed fo llo w ing T C tr ea tm ent (in cl ud in g in fe rt ili ty tr ea tm en t i n 10 ). 77 % (1 59 /2 07 ) c on ce ive d w ith ou t i nfe rti lity tr ea tm en t (p = .0 3 fo r c he m ot he ra py v s su rv ei lla nc e) Me n w ith e le va te d FS H le ss lik el y to h av e co nc ei ve d (6 8% ) t ha n m en w ith n or m al F SH le ve ls (9 1% ). (P < .0 01 ) Br yd øy (2 00 5) (1 01 ) 552 11 (4 -21) Su rv ei lla nc e RP LN D RT Cisp la tin ≤ 8 50 m g‡ Ci sp la tin > 85 0 m g‡ 81% fa th er ed (92% 15 -y§ ) 77% fa th er ed (7 8% 1 5-y§ ) 65% fa th er ed (72% 15 -y§ ) 62% fa th er ed (64% 15 -y§ ) 38% fa th er ed (48% 15 -y§ ) 65% (36 1/ 552) h ad fa th er ed b y fo llo w -up (w ith out cr yo pr es er ve d sp er m , b ut in cl ud in g ot he r i nf er til ity tr ea tm en ts in 3 6) 71% o ve ra ll 15 -y ac tu ar ia l p os t-tr ea tm en t pa te rn ity ra te s (p < .0 01 a cc or di ng to tr ea tm en t g ro up ) Dr y ej ac ul at io n w as th e m os t n eg at iv e pr ed ic to r. Me n di ag no se d 19 89 -1994 w er e m or e lik el y to h av e fa th er ed th an m en d ia gn os ed 1 98 0-1988. (P < .001) *N um be rs a re p os t-tr ea tm en t w he n no t s pe ci fie d as pr e-tr ea tm en t ( pr e) . A bb re vi ati on s: N o, n um be rs ; f u, fo llo w -up ; y , y ea rs ; R PL N D , r et ro pe rit on ea l l ym ph n od e di ss ec tio n; CT , ch em ot he ra py ; R T, ra di ot he ra py ; F SH , f ol licl e st im ul at in g ho rm on e. †W ith ou t i nf er til ity tr ea tme nt . T he u se o f c ry op re se rv ed s eme n w as n ot s pe ci fie d. ‡B ot h ch emo th er ap y gr ou ps in cl ud ed p at ie nt s ad di tio na lly tr ea te d w ith R T or R PL N D . § Ra te s re fe r to 1 5-y ac tu ar ia l r at e

(16)

Lampe et al. defined prognostic groups for spermatogenic recovery based on repeated sperm analyses pre- and post-chemotherapy in 178 men.121 Overall, 80% recovered

some spermatogenesis by 4 years after chemotherapy, if not azoospermic (<1 mill/ml) prior to treatment.121 Carboplatin-based regimens and ≤4 cycles were positively related to recovery.

Petersen et al. found that 19% were azoospermic 5 years after 4 BEP cycles, and 47% after ≥3cycles of a more dose intensive regimen.122 Gandini et al. reported that 94% and 97% had recovered

some spermatogenesis two years after RT and 2 to 4 cycles of chemotherapy, respectively.123

Information regarding fertility following more recently introduced chemotherapy regimens is sparse.124, 125 Recovery of spermatogenesis following high-dose chemotherapy with

stem cell support has also been reported (5/10 patients).126 TC patients should be informed that

the recovery from chemotherapy-induced azoospermia may take several years. Furthermore, offering cryopreservation is obligatory prior to chemotherapy.

Sexuality

Besides impaired ejaculatory function, groups of TCSs are reported to experience decreased drive and increased erectile dysfunction when compared to controls.99, 127, 128 Some also report

decreased sexual satisfaction.129 Dahl et al found that overall sexual problems were expressed

by 39% of TCSs versus 36% in controls.128 Interestingly, among men aged 20-39 years, sexual

satisfaction was significantly better compared to controls. Tuinman et al. showed that couples established after a TC diagnosis were less sexually satisfied than couples founded pre-diagnosis.129

However, data within this field are conflicting. Psychosocial health

Fatigue

A significantly higher frequency of chronic (> 6 months’ duration) cancer-related fatigue (CRF) was reported among long-term TCSs in Norway compared with the normative male population (17% and 10%, respectively).130 Significantly higher levels of interleukin-1 receptor antagonist and

C-reactive protein were observed in TCSs with CRF compared to TCSs without CRF, indicating an association between fatigue and inflammation.131

Mental health

Compared to samples of men from the general population, significantly increased levels of anxiety among Norwegian TCSs were associated with peripheral neuropathy, fear of recurrence, economic concerns, alcohol abuse, sexual difficulties, younger age, and a history of treatment for mental problems.132 It is not clear whether TCSs experience more depressive disorders than the

general population, since a significantly increased prevalence has been reported in some studies,

(17)

Lifestyle

A recent American study confirmed a previous Norwegian finding of high prevalence of current smoking and problem drinking among TCSs. Their intake of fruit and vegetables was low. However, TCSs were more likely to engage in regular exercise that cancer-free controls.134

Employment

In the U.S., levels of unemployment among TCSs are similar to men in the general population.135

Similarly, Norwegian TCSs reported comparable levels of work engagement as age-matched men in the general population.136 Fleer et al. highlighted the importance of employment on

health-related quality of life in TCSs.137 No data are available with regard to work ability 10 or more years

after diagnosis of TC.

Health-related quality of life (HRQoL)

In most studies, overall HRQoL of TCSs, as assessed by validated questionnaires, has been similar to that of normative samples of age-matched men.138, 139 Although findings may accurately reflect

HRQoL status, these results may also be due to either “response shift” 140 or the use of instruments

that are not targeted to the concerns of TCSs. In one investigation, a subgroup of approximately 15% TCSs reported decreased HRQoL.138

Impact of lifestyle factors

Cancer survivors, in general, are high health care utilizers with several distinct health care issues.4, 141, 142 For TCSs, in particular, the young age at diagnosis and high cure rate, lead to

long-term consequences of therapy which have a greater impact on their lives, families, and society at large than the acute complications from the cytotoxic therapies.22, 25, 143, 144

While treatment-associated long-term risks and complications are brought on by cytotoxic treatment, genetic predisposition and/or common lifestyle factors,145-148 lifestyle is the only variable

subjected to change. In line with this, Aziz argue that the follow-up care of cancer patients should provide the opportunity for maintaining health by lifestyle interventions.4

Prevention such as promoting smoking cessation, better nutrition, and promoting a non-sedetary lifestyle may play key roles in reducing the impact of potential adverse effects of previous cancer therapy.149-151 In a large Dutch population of TCSs, the relative risk of CVD attributed to recent

smoking was 2.6, while the corresponding factor for cisplatin-based chemotherapy was 1.9.24 In

a Norwegian population of long-term TCSs, smoking was associated with decreasing pulmonary function as well as increasing neurological side-effects.37, 53 Additionally, smoking increases the

overall risk of a second malignancy.9 Thus, lifestyle interventions may be particularly important for

TCSs due to the long life expectancy and their increased vulnerability. These interventions may start early in the follow-up period. Demark-Wahnefried and coworkers focus on the “teachable moment” which the cancer diagnosis provides and argue that oncology care providers should not only lead their patients away from disease, but also promote lifestyle changes that may improve the length and quality of life of their patients.142 However, of 160 German TC patients who did

smoke at the time of diagnosis, most changed their tobacco habit, but only 29% managed to quit smoking within 10 years post diagnosis.152 In a randomized physical training intervention in Chapter 2

(18)

a Norwegian population of long-term TCSs, training had significant effect on cardiorespiratory fitness among the young and middle-aged, but had no favorable effect on patients’ experience of fatigue, mental distress, or health-related quality of life.153 Hence, substantial intervention efforts

may be required for limited effects, but the potential for improved health and reduced morbidity in long-term TCSs is definitely present.

Conclusions and recommendations for follow-up

In this review we have presented the current status regarding different somatic and psychosocial effects after TC treatment. Current knowledge about adverse effects is based on therapy administered years to decades ago. Although cisplatin-based chemotherapy still is the cornerstone in the treatment of TC, we do not have long-term data regarding the consequences from receiving one or two cycles of BEP or carboplatin in the adjuvant setting, or from receiving other chemotherapy agents such as paclitaxel. Thus, there is clearly a need for ongoing research to elucidate long-term and late effects from current treatment strategies and to minimize the long-term morbidity after TC treatment. To achieve this goal, better knowledge about the pathophysiology behind the term complications, defining risk factors for developing long-term morbidity and identification of predictive factors for adverse effects is required.5

During follow-up of TC patients, there is a gradual shift of focus from detection of tumor recurrence to identification of late effects of treatment and promotion of general health in TCSs. We should recommend our patients to maintain a healthy life-style to reduce the risk of serious late effects as second cancers and CVD. Every cancer patient should have an informative end-of-treatment summary at completion of the treatment together with a survivorship care plan. The focus of the survivorship care plan will change with increasing follow-up duration from early detection of a relapse to measures to maintain health. This survivorship care plan includes tools for acquiring and maintaining a healthy life style and a formal regular check on cardiovascular risk factors and gonadal status. A survivorship care plan can be implemented in addition to the routine oncological follow-up or when the routine follow-up with the oncologist is terminated and taken over by another healthcare giver. If so, there has to be a proper transition of further follow-up with a solid and clear survivorship plan (Fig 2). Currently, the distribution of survivorship plans is recommended in Norway, Sweden and the Netherlands. There are so far no data evaluating the use of a survivorship plan.

In conclusion, the treatment success of men with TC is hampered by the emergence of long-term and late effects of treatment. Knowledge about adverse effects is crucial for all physicians involved in treatment and follow-up of TC patients, and attention towards prevention and identification of adverse effects is essential for these young men.

(19)

Figure 2. Survivorship care plan to be delivered to the patient at completion of treatment. The plan includes a treatment summary and short information about side-eff ects from testicular cancer treatment, focusing on modifi able cardiovascular risk factors.

(20)

References

Verdecchia A, Francisci S, Brenner H, et al: Recent cancer survival in Europe: a 2000-02 period analysis of EUROCARE-4 data. Lancet Oncol 8:784-796, 2007

Feldman DR, Bosl GJ, Sheinfeld J, et al: Medical Treatment of Advanced Testicular Cancer. Jama 299:672-684, 2008

Abouassaly R, Fossa SD, Giwercman A, et al: Sequelae of treatment in long-term survivors of testis cancer. Eur Urol 60:516-526, 2011

Aziz NM: Cancer survivorship research: state of knowledge, challenges and opportunities. Acta Oncol 46:417-432, 2007

Travis LB, Beard C, Allan JM, et al: Testicular cancer survivorship: research strategies and recommendations. J Natl Cancer Inst 102:1114-1130, 2010

Fossa SD, Chen J, Schonfeld SJ, et al: Risk of contralateral testicular cancer: a population-based study of 29,515 U.S. men. J Natl Cancer Inst 97:1056-1066, 2005

Travis LB, Fossa SD, Schonfeld SJ, et al: Second cancers among 40,576 testicular cancer patients: focus on long-term survivors. J Natl Cancer Inst 97:1354-1365, 2005

Richiardi L, Scelo G, Boffetta P, et al: Second malignancies among survivors of germ-cell testicular cancer: a pooled analysis between 13 cancer registries. Int J Cancer 120:623-631, 2007

van den Belt-Dusebout AW, de WR, Gietema JA, et al: Treatment-specific risks of second malignancies and cardiovascular disease in 5-year survivors of testicular cancer. J Clin Oncol 25:4370-4378, 2007 Howard R, Gilbert E, Lynch CF, et al: Risk of Leukemia Among Survivors of Testicular Cancer: A Population-based Study of 42,722 Patients. Ann Epidemiol 18:416-421, 2008

Kollmannsberger C, Beyer J, Droz JP, et al: Secondary leukemia following high cumulative doses of etoposide in patients treated for advanced germ cell tumors. J Clin Oncol 16:3386-3391, 1998

Houck W, Abonour R, Vance G, et al: Secondary leukemias in refractory germ cell tumor patients undergoing autologous stem-cell transplantation using high-dose etoposide. J Clin Oncol 22:2155-2158, 2004

Wierecky J, Kollmannsberger C, Boehlke I, et al: Secondary leukemia after first-line high-dose chemotherapy for patients with advanced germ cell cancer. J Cancer Res Clin Oncol 131:255-260, 2005 Travis LB, Andersson M, Gospodarowicz M, et al: Treatment-associated leukemia following testicular cancer. J Natl Cancer Inst 92:1165-1171, 2000

Fossa SD, Aass N, Kaalhus O: Radiotherapy for testicular seminoma stage I: treatment results and long-term post-irradiation morbidity in 365 patients. Int J Radiat Oncol Biol Phys 16:383-388, 1989

Tarin TV, Sonn G, Shinghal R: Estimating the risk of cancer associated with imaging related radiation during surveillance for stage I testicular cancer using computerized tomography. J Urol 181:627-632, 2009

Hall EJ, Brenner DJ: Cancer risks from diagnostic radiology. Br J Radiol 81:362-378, 2008

van WC, Fergusson D, Earle C, et al: Association of diagnostic radiation exposure and second abdominal-pelvic malignancies after testicular cancer. J Clin Oncol 29:2883-2888, 2011

Doll DC, List AF, Greco FA, et al: Acute vascular ischemic events after cisplatin-based combination chemotherapy for germ-cell tumors of the testis. Ann Intern Med 105:48-51, 1986

Meinardi MT, Gietema JA, van der Graaf WT, et al: Cardiovascular morbidity in long-term survivors of metastatic testicular cancer. J Clin Oncol 18:1725-1732, 2000

Zagars GK, Ballo MT, Lee AK, et al: Mortality after cure of testicular seminoma. J Clin Oncol 22:640-647, 2004 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

(21)

Fossa SD, Gilbert E, Dores GM, et al: Noncancer causes of death in survivors of testicular cancer. J Natl Cancer Inst 99, 2007

Huddart RA, Norman A, Shahidi M, et al: Cardiovascular disease as a long-term complication of treatment for testicular cancer. J Clin Oncol 21:1513-1523, 2003

van den Belt-Dusebout A, Nuver J, de Wit R, et al: Long-Term Risk of Cardiovascular Disease in 5-Year Survivors of Testicular Cancer. J Clin Oncol 24:467-475, 2006

Haugnes HS, Wethal T, Aass N, et al: Cardiovascular risk factors and morbidity in testicular cancer survivors: A 20 year follow-up study. J Clin Oncol 28:4649-4657, 2010

Yusuf S, Hawken S, Èunpuu S, et al: Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study. The Lancet 364:937-952, 2004 Sagstuen H, Aass N, Fosså SD, et al: Blood pressure and body mass index in long-term survivors of testicular cancer. J Clin Oncol 23:4980-4990, 2005

Alberti KGM, Zimmet P, Shaw J: The metabolic syndrome--a new worldwide definition. The Lancet 366:1059-1062, 2005

Haugnes HS, Aass N, Fossa SD, et al: Components of the metabolic syndrome in long-term survivors of testicular cancer. Ann Oncol 18:241-248, 2007

Nuver J, Smit AJ, Wolffenbuttel BH, et al: The Metabolic Syndrome and Disturbances in Hormone Levels in Long-Term Survivors of Disseminated Testicular Cancer. J Clin Oncol 23:3718-3725, 2005

31. Libby P, Ridker PM, Hansson GrK: Inflammation in Atherosclerosis: From Pathophysiology to Practice. J Am Coll Cardiol 54:2129-2138, 2009

Nuver J, Smit AJ, Sleijfer DT, et al: Microalbuminuria, decreased fibrinolysis, and inflammation as early signs of atherosclerosis in long-term survivors of disseminated testicular cancer. Eur J Cancer 40:701-706, 2004

Sprauten M, Darrah TH, Peterson DR, et al: Impact of long-term platinum concentrations on neuro- and ototoxicity in cisplatin-treated testicular cancer survivors, in (ed 30), 2012, pp 300-307

Gietema JA, Meinardi MT, Messerschmidt J, et al: Circulating plasma platinum more than 10 years after cisplatin treatment for testicular cancer. Lancet 355:1075-1076, 2000

Wethal T, Haugnes HS, Kjekshus J, et al: C-reactive protein; a potential marker of second cancer and cardiovascular disease in testicular cancer survivors?, in (ed 46), 2010, pp 3425-3433

Berger CC, Bokemeyer C, Schneider M, et al: Secondary Raynaud’s phenomenon and other late vascular complications following chemotherapy for testicular cancer. Eur J Cancer 31A:2229-2238, 1995

Brydoy M, Oldenburg J, Klepp O, et al: Observational study of prevalence of long-term Raynaud-like phenomena and neurological side effects in testicular cancer survivors. J Natl Cancer Inst 101:1682-1695, 2009

Rossen PB, Pedersen AF, Zachariae R, et al: Health-Related Quality of Life in Long-Term Survivors of Testicular Cancer. J Clin Oncol 27:5993-5999, 2009

Glendenning JL, Barbachano Y, Norman AR, et al: Long-term neurologic and peripheral vascular toxicity after chemotherapy treatment of testicular cancer. Cancer 116:2322-2331, 2010

Hansen SW, Olsen N, Rossing N, et al: Vascular toxicity and the mechanism underlying Raynaud’s phenomenon in patients treated with cisplatin, vinblastine and bleomycin. Ann Oncol 1:289-292, 1990 Block JA, Sequeira W: Raynaud’s phenomenon. Lancet 357:2042-2048, 2001

Carter SK, Blum RH: New Chemotherapeutic Agents... Bleomycin and Adriamycin. CA Cancer J Clin 24:322-331, 1974

Comis RL, Kuppinger MS, Ginsberg SJ, et al: Role of Single-Breath Carbon Monoxide-diffusing Capacity in Monitoring the Pulmonary Effects of Bleomycin in Germ Cell Tumor Patients. Cancer Res 39:5076-5080, 1979 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 Chapter 2

(22)

O’Sullivan JM, Huddart RA, Norman AR, et al: Predicting the risk of bleomycin lung toxicity in patients with germ-cell tumours. Ann Oncol 14:91-96, 2003

Van Barneveld PW, van der Mark TW, Sleijfer DT, et al: Predictive factors for bleomycin-induced pneumonitis. Am Rev Respir Dis 130:1078-1081, 1984

Dearnaley DP, Horwich A, A’Hern R, et al: Combination chemotherapy with bleomycin, etoposide and cisplatin (BEP) for metastatic testicular teratoma: long-term follow-up. Eur J Cancer 27:684-691, 1991 Nuver J, Lutke Holzik MF, Van ZM, et al: Genetic variation in the bleomycin hydrolase gene and bleomycin-induced pulmonary toxicity in germ cell cancer patients. Pharmacogenet Genomics 15:399-405, 2005 Simpson AB, Paul J, Graham J, et al: Fatal bleomycin pulmonary toxicity in the west of Scotland 1991-95: a review of patients with germ cell tumours. Br J Cancer 78:1061-1066, 1998

Van Barneveld PW, Sleijfer DT, van der Mark TW, et al: Natural course of bleomycin-induced pneumonitis. A follow-up study. Am Rev Respir Dis 135:48-51, 1987

Lucraft HH, Wilkinson PM, Stretton TB, et al: Role of pulmonary function tests in the prevention of bleomycin pulmonary toxicity during chemotherapy for metastatic testicular teratoma. Eur J Cancer Clin Oncol 18:133-139, 1982

Osanto S, Bukman A, Van Hoek F, et al: Long-term effects of chemotherapy in patients with testicular cancer. J Clin Oncol 10:574-579, 1992

Hansen SW, Groth S, Sorensen PG, et al: Enhanced pulmonary toxicity in smokers with germ-cell cancer treated with cis-platinum, vinblastine and bleomycin: a long-term follow-up. Eur J Cancer Clin Oncol 25:733-736, 1989

Haugnes HS, Aass N, Fossa SD, et al: Pulmonary Function in Long- Term Survivors of Testicular Cancer. J Clin Oncol 27:2779-2786, 2009

Mannino DM, Buist AS, Petty TL, et al: Lung function and mortality in the United States: data from the First National Health and Nutrition Examination Survey follow up study. Thorax 58:388-393, 2003

Schunemann HJ, Dorn J, Grant BJB, et al: Pulmonary Function Is a Long-term Predictor of Mortality in the General Population : 29-Year Follow-up of the Buffalo Health Study. Chest 118:656-664, 2000

Nuver J, de Haas EC, Van ZM, et al: Vascular damage in testicular cancer patients: a study on endothelial activation by bleomycin and cisplatin in vitro. Oncol Rep 23:247-253, 2010

Hansen SW, Groth S, Daugaard G, et al: Long-term effects on renal function and blood pressure of treatment with cisplatin, vinblastine, and bleomycin in patients with germ cell cancer. J Clin Oncol 6:1728-1731, 1988

Fossa SD, Aass N, Winderen M, et al: Long-term renal function after treatment for malignant germ-cell tumours. Ann Oncol 13:222-228, 2002

Vickers AE, Rose K, Fisher R, et al: Kidney slices of human and rat to characterize cisplatin-induced injury on cellular pathways and morphology. Toxicol Pathol 32:577-590, 2004

Tanaka H, Ishikawa E, Teshima S, et al: Histopathological study of human cisplatin nephropathy. Toxicol Pathol 14:247-257, 1986

Hodgkinson E, Neville-Webbe HL, Coleman RE: Magnesium depletion in patients receiving cisplatin-based chemotherapy. Clin Oncol (R Coll Radiol ) 18:710-718, 2006

Willox JC, McAllister EJ, Sangster G, et al: Effects of magnesium supplementation in testicular cancer patients receiving cis-platin: a randomised trial. Br J Cancer 54:19-23, 1986

Dentino M, Luft FC, Yum MN, et al: Long term effect of cis- diamminedichloride platinum (CDDP) on renal function and structure in man. Cancer 41:1274-1281, 1978

Schilsky RL, Anderson T: Hypomagnesemia and renal magnesium wasting in patients receiving cisplatin. Ann Intern Med 90:929-931, 1979

44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64

(23)

Vogelzang NJ, Torkelson JL, Kennedy BJ: Hypomagnesemia, renal dysfunction, and Raynaud’s phenomenon in patients treated with cisplatin, vinblastine, and bleomycin. Cancer 56:2765-2770, 1985 Schilsky RL, Barlock A, Ozols RF: Persistent hypomagnesemia following cisplatin chemotherapy for testicular cancer. Cancer Treat Rep 66:1767- 1769, 1982

Hamdi T, Latta S, Jallad B, et al: Cisplatin-induced renal salt wasting syndrome. South Med J 103:793-799, 2010

Daugaard G, Abildgaard U: Cisplatin nephrotoxicity. A review. Cancer Chemother Pharmacol 25:1-9, 1989 Daugaard G, Rossing N, Rorth M: Effects of cisplatin on different measures of glomerular function in the human kidney with special emphasis on high- dose. Cancer Chemother Pharmacol 21:163-167, 1988 Hartmann JT, Kollmannsberger C, Kanz L, et al: Platinum organ toxicity and possible prevention in patients with testicular cancer. Int J Cancer 83:866-869, 1999

Bosl GJ, Leitner SP, Atlas SA, et al: Increased plasma renin and aldosterone in patients treated with cisplatin-based chemotherapy for metastatic germ-cell tumors. J Clin Oncol 4:1684-1689, 1986

Meijer S, Mulder NH, Sleijfer DT, et al: Nephrotoxicity of cis- diamminedichloride platinum (CDDP) during remission-induction and maintenance chemotherapy of testicular carcinoma. Cancer Chemother Pharmacol 8:27-30, 1982

Bali L, Silhol F, Kateb A, et al: [Renal artery stenosis after abdominal radiotherapy]. Ann Cardiol Angeiol (Paris) 58:183-186, 2009

Mulla MG, Ananthkrishnan G, Mirza MS, et al: Renal artery stenosis after radiotherapy for stage I seminoma, a case report. Clin Oncol (R Coll Radiol ) 19:209, 2007

Peacock JM, Ohira T, Post W, et al: Serum magnesium and risk of sudden cardiac death in the Atherosclerosis Risk in Communities (ARIC) Study. Am Heart J 160:464-470, 2010

Astor BC, Hallan SI, Miller ER, III, et al: Glomerular filtration rate, albuminuria, and risk of cardiovascular and all-cause mortality in the US population. Am J Epidemiol 167:1226-1234, 2008

Gansevoort RT, Matsushita K, van d, V, et al: Lower estimated GFR and higher albuminuria are associated with adverse kidney outcomes in both general and high-risk populations. A collaborative meta-analysis of general and high-risk population cohorts. Kidney Int 80:93-104, 2011

Krarup-Hansen A, Helweg-Larsen S, Schmalbruch H, et al: Neuronal involvement in cisplatin neuropathy: prospective clinical and neurophysiological studies. Brain 130:1076-1088, 2007

Hausheer FH, Schilsky RL, Bain S, et al: Diagnosis, management, and evaluation of chemotherapy-induced peripheral neuropathy. Semin Oncol 33:15-49, 2006

Kollmannsberger C, Kuzcyk M, Mayer F, et al: Late toxicity following curative treatment of testicular cancer. Semin Surg Oncol 17:275-281, 1999

Wolf S, Barton D, Kottschade L, et al: Chemotherapy-induced peripheral neuropathy: prevention and treatment strategies. Eur J Cancer 44:1507- 1515, 2008

Bauer CA, Brozoski TJ: Cochlear structure and function after round window application of ototoxins. Hear Res 201:121-131, 2005

Bokemeyer C, Berger CC, Hartmann JT, et al: Analysis of risk factors for cisplatin-induced ototoxicity in patients with testicular cancer. Br J Cancer 77:1355-1362, 1998

Bokemeyer C, Berger CC, Kuczyk MA, et al: Evaluation of long-term toxicity after chemotherapy for testicular cancer. J Clin Oncol 14:2923-2932, 1996

Fossa SD, de Wit R, Roberts JT, et al: Quality of life in good prognosis patients with metastatic germ cell cancer: a prospective study of the European Organization for Research and Treatment of Cancer Genitourinary Group/Medical Research Council Testicular Cancer Study Group (30941/TE20). J Clin Oncol 21:1107-1118, 2003 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 Chapter 2

(24)

Oldenburg J, Kraggerud SM, Cvancarova M, et al: Cisplatin-induced long-term hearing impairment is associated with specific glutathione s-transferase genotypes in testicular cancer survivors. J Clin Oncol 25:708-714, 2007

Oldenburg J, Kraggerud SM, Brydoy M, et al: Association between long-term neuro-toxicities in testicular cancer survivors and polymorphisms in glutathione-s-transferase-P1 and -M1, a retrospective cross sectional study. J Transl Med 5:70, 2007

Ross CJ, Katzov-Eckert H, Dube MP, et al: Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy. Nat Genet 41:1345-1349, 2009

Riedemann L, Lanvers C, Deuster D, et al: Megalin genetic polymorphisms and individual sensitivity to the ototoxic effect of cisplatin. Pharmacogenomics J 8:23-28, 2008

Skoogh J, Steineck G, Stierner U, et al: Testicular-cancer survivors experience compromised language following chemotherapy: Findings in a Swedish population-based study 3-26 years after treatment. Acta Oncol 51:185-197, 2012

Pedersen AD, Rossen P, Mehlsen MY, et al: Long-term cognitive function following chemotherapy in patients with testicular cancer. J Int Neuropsychol Soc 15:296-301, 2009

Schagen SB, Boogerd W, Muller MJ, et al: Cognitive complaints and cognitive impairment following BEP chemotherapy in patients with testicular cancer. Acta Oncol 47:63-70, 2008

Skaali T, Fossa SD, Andersson S, et al: Self-reported cognitive problems in testicular cancer patients: relation to neuropsychological performance, fatigue, and psychological distress. J Psychosom Res 70:403-410, 2011

Shim K, MacKenzie MJ, Winquist E: Chemotherapy-associated osteonecrosis in cancer patients with solid tumours: a systematic review. Drug Saf 31:359-371, 2008

Winquist EW, Bauman GS, Balogh J: Nontraumatic osteonecrosis after chemotherapy for testicular cancer: a systematic review. Am J Clin Oncol 24:603- 606, 2001

Cook AM, Dzik-Jurasz AS, Padhani AR, et al: The prevalence of avascular necrosis in patients treated with chemotherapy for testicular tumours. Br J Cancer 85:1624-1626, 2001

Gerl A, Muhlbayer D, Hansmann G, et al: The impact of chemotherapy on Leydig cell function in long term survivors of germ cell tumors. Cancer 91:1297- 1303, 2001

Wiechno P, Demkow T, Kubiak K, et al: The quality of life and hormonal disturbances in testicular cancer survivors in Cisplatin era. Eur Urol 52:1448-1454, 2007

Huddart RA, Norman A, Moynihan C, et al: Fertility, gonadal and sexual function in survivors of testicular cancer. Br J Cancer 93:200-207, 2005

Palmieri G, Lotrecchiano G, Ricci G, et al: Gonadal function after multimodality treatment in men with testicular germ cell cancer. Eur J Endocrinol 134:431-436, 1996

Hansen SW, Berthelsen JG, Von Der MH: Long-term fertility and Leydig cell function in patients treated for germ cell cancer with cisplatin, vinblastine, and bleomycin versus surveillance. J Clin Oncol 8:1695-1698, 1990

Nord C, Bjoro T, Ellingsen D, et al: Gonadal hormones in long-term survivors 10 years after treatment for unilateral testicular cancer. Eur Urol 44:322- 328, 2003

Brand JS, van dT, I, Grobbee DE, et al: Testosterone, sex hormone- binding globulin and the metabolic syndrome: a systematic review and meta-analysis of observational studies. Int J Epidemiol 40:189-207, 2011 Araujo AB, Kupelian V, Page ST, et al: Sex Steroids and All-Cause and Cause-Specific Mortality in Men. Arch Intern Med 167:1252-1260, 2007

Laughlin GA, Barrett-Connor E, Bergstrom J: Low Serum Testosterone and Mortality in Older Men. J Clin Endocrinol Metab 93:68-75, 2008 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105

(25)

Brown JE, Ellis SP, Silcocks P, et al: Effect of chemotherapy on skeletal health in male survivors from testicular cancer and lymphoma. Clin Cancer Res 12:6480-6486, 2006

Ondrusova M, Ondrus D, Dusek L, et al: Damage of hormonal function and bone metabolism in long-term survivors of testicular cancer. Neoplasma 56:473- 479, 2009

Bhasin S, Cunningham GR, Hayes FJ, et al: Testosterone therapy in men with androgen deficiency syndromes: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 95:2536-2559, 2010 Huyghe E, Matsuda T, Daudin M, et al: Fertility after testicular cancer treatments: results of a large multicenter study. Cancer 100:732-737, 2004

Brydoy M, Fossa SD, Klepp O, et al: Paternity following treatment for testicular cancer. J Natl Cancer Inst 97:1580-1588, 2005

Matos E, Skrbinc B, Zakotnik B: Fertility in patients treated for testicular cancer. J Cancer Surviv 4:274-278, 2010

Spermon JR, Kiemeney LA, Meuleman EJ, et al: Fertility in men with testicular germ cell tumors. Fertil Steril 79 Suppl 3:1543-1549, 2003

Brydoy M, Fossa SD, Klepp O, et al: Paternity and Testicular Function Among Testicular Cancer Survivors Treated With Two to Four Cycles of Cisplatin- Based Chemotherapy. Eur Urol 58:134-141, 2010

Beck SD, Bey AL, Bihrle R, et al: Ejaculatory status and fertility rates after primary retroperitoneal lymph node dissection. J Urol 184:2078-2080, 2010

Heidenreich A, Albers P, Hartmann M, et al: Complications of primary nerve sparing retroperitoneal lymph node dissection for clinical stage I nonseminomatous germ cell tumors of the testis: experience of the German Testicular Cancer Study Group. J Urol 169:1710-1714, 2003

Subramanian VS, Nguyen CT, Stephenson AJ, et al: Complications of open primary and post-chemotherapy retroperitoneal lymph node dissection for testicular cancer. Urol Oncol 28:504-509, 2010 Jacobsen KD, Ous S, Waehre H, et al: Ejaculation in testicular cancer patients after post-chemotherapy retroperitoneal lymph node dissection. Br J Cancer 80:249-255, 1999

Pettus JA, Carver BS, Masterson T, et al: Preservation of ejaculation in patients undergoing nerve-sparing postchemotherapy retroperitoneal lymph node dissection for metastatic testicular cancer. Urology 73:328-331, 2009

Coogan CL, Hejase MJ, Wahle GR, et al: Nerve sparing post- chemotherapy retroperitoneal lymph node dissection for advanced testicular cancer. J Urol 156:1656-1658, 1996

Heidenreich A, Pfister D, Witthuhn R, et al: Postchemotherapy retroperitoneal lymph node dissection in advanced testicular cancer: radical or modified template resection. Eur Urol 55:217-224, 2009

Lampe H, Horwich A, Norman A, et al: Fertility after chemotherapy for testicular germ cell cancers. J Clin Oncol 15:239-245, 1997

Petersen PM, Hansen SW, Giwercman A, et al: Dose-dependent impairment of testicular function in patients treated with cisplatin-based chemotherapy for germ cell cancer. Ann Oncol 5:355-358, 1994 Gandini L, Sgro P, Lombardo F, et al: Effect of chemo- or radiotherapy on sperm parameters of testicular cancer patients. Hum Reprod 21:2882-2889, 2006

Reiter WJ, Kratzik C, Brodowicz T, et al: Sperm analysis and serum follicle-stimulating hormone levels before and after adjuvant single-agent carboplatin therapy for clinical stage I seminoma. Urology 52:117-119, 1998

Pectasides D, Pectasides E, Papaxoinis G, et al: Testicular function in poor-risk nonseminomatous germ cell tumors treated with methotrexate, paclitaxel, ifosfamide, and cisplatin combination chemotherapy. J Androl 30:280-286, 2009

Ishikawa T, Kamidono S, Fujisawa M: Fertility after high-dose chemotherapy for testicular cancer. Urology 63:137-140, 2004 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 Chapter 2

Referenties

GERELATEERDE DOCUMENTEN

Patients homozygous or heterozygous variant for SNP rs523349 have an odds ratio of 2.56 for the metabolic syndrome after treatment for metastatic testicular cancer compared

Metastatic TC patients with a high-risk vascular fingerprint before start of (B)EP chemotherapy developed arterial vascular events more often (19% vs. 2%).. During chemotherapy

By focusing too much on the care plan as a document summarizing the treatment we tend to forget the primary goal of a plan in general: guiding follow-up care to ensure that

We launched and made available a mobile application and the accompanying SCP generator, which can be easily used to offer survivors and their healthcare providers a clear

Therefore, we designed a survivorship care program for testicular cancer survivors to test the safety and feasibility of shared-care follow-up together with the PCP, and compared

Ideally, each cancer survivor should receive survivorship care that is adjusted to his or her diagnosis and treatment plan and personal risk profile, based on

Het is van potentieel belang om patiënten met een hoger risico op cardiovasculaire complicaties tijdens chemotherapie te identificeren, bij voorkeur voorafgaand aan de

Sjoukje Lubberts en Lars Steggink, dank voor de soepele samenwerking bij de verschillende studies en veel succes met de afronding van jullie proefschriften.. Nynke