• 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!
185
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)

Survivorship care after testicular cancer

New insights in late effects of treatment and

approaches to shared-care follow-up

H. Boer

Survivorship care after testicular cancer

New insights in late effects of treatment and

approaches to shared-care follow-up

(3)

The research presented in this thesis was conducted at the Department of Medical Oncology, University of Groningen, University Medical Center Groningen. This research project was financially supported by the Dutch Cancer Society (project number RUG 2011-5267) and the Netherlands Comprehensive Cancer Organisation.

Layout Bianca Pijl, www.pijlldesign.nl

Groningen, the Netherlands

Cover design Sam Maldonado, wwwanted.nl,

Amsterdam, the Netherlands

Printed by Ipskamp Printing

Enschede, the Netherlands

ISBN 978-94-034-1332-7 (print)

978-94-034-1331-0 (digital)

© Copyright, 2019, H. Boer, Groningen, the Netherlands

All rights reserved. No part of this thesis may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, without prior written permission of the author, or when appropriate, of the publishers of the publications included in this thesis.

The printing of this thesis was financially supported by the Stichting Werkgroep Interne Oncologie and the Graduate School of Medicals Science University of Groningen, University Medical Center

(4)

Survivorship care after testicular cancer

New insights in late effects of treatment and

approaches to shared-care follow-up

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de

rector magnificus prof. dr. E. Sterken

en volgens besluit van het College voor Promoties De openbare verdediging zal plaatsvinden op

maandag 25 februari 2019 om 16.15 uur

door

Hindrik Boer

geboren op 17 december 1983 te NietapRoden

(5)

Promotores

Prof. dr. J.A. Gietema Prof. dr. S. Siesling

Copromotor

Dr. A.J. Berendsen

Beoordelingscommissie

Prof. dr. W.T.A. van der Graaf Prof. dr. G.H. de Bock Prof. dr. D.J. Touw

(6)
(7)

Paranimfen

Dr. R.R.H. van den Brom Mr. dr. F.J. Vonck

(8)

Chapter 1 Chapter 2 Chapter 3 Chapter 4 Chapter 5 Chapter 6 Chapter 7 Chapter 8 Chapter 9

Contents

General introduction

Long-term and late effects of germ-cell testicular cancer treatment and implications for follow-up

Journal of Clinical Oncology, 2012

Long-term exposure to circulating platinum is associated with late effects of treatment in testicular cancer survivors

Annals of Oncology, 2015

Single nucleotide polymorphism in the 5-α-reductase gene (SRD5A2) is associated with increased prevalence of metabolic syndrome in chemotherapy-treated testicular cancer survivors

European Journal of Cancer, 2016

Vascular fingerprint and vascular damage markers associated with vascular events in testicular cancer patients during and after chemotherapy

European Journal of Cancer, 2016

Easy navigating through the forest of survivorship care Journal of Clinical Oncology, 2014

The next step in survivorship care plans: In control with an easy to use mobile application

Submitted

Shared survivorship care after testicular cancer Submitted

Summary and future perspectives

Summary in Dutch / Nederlandse samenvatting Acknowledgements / Dankwoord 9 19 47 69 89 109 117 133 155 171 179

(9)
(10)

General introduction

Chapter 1

(11)
(12)

11 Introduction and aim

Background

Testicular cancer is a disease that primarily aff ects young men between the age of 15 and 35 years. In this age group, it is the most common type of cancer in men. The incidence of testicular cancer is steadily increasing (Figure 1).1 The incidence rates in the northern and western part of

Europe are the highest worldwide.2 Since the introduction of platinum-based chemotherapy in

the late seventies, metastatic testicular cancer has become a curable disease with survival rates around 80%.3 Approximately 95% of testicular cancers are germ cell tumors. Germ cell tumors

are subdivided based on histology in two categories: seminoma (55%) and non-seminoma (45%). Treatment strategy for metastatic disease depends on histology, location of disease and classifi cation in prognosis groups based on the International Germ Cell Cancer Collaboration Group criteria.4 In general, in case of metastatic disease, the treatment consists of surgery with

subsequent platinum-based combination chemotherapy.5 In case of limited disseminated

seminoma, radiotherapy is a treatment option.5 In stage I disease, watchful-waiting or active

surveillance has become the main strategy instead of adjuvant chemotherapy or radiotherapy.6

Figure 1. Incidence (blue line) and mortality (green line) of testicular cancer in The Netherlands (Per 1:100.000 males, ESR).

As a result of both the increasing incidence and high cure rates, the number of survivors of testicular cancer is increasing. In the Netherlands, in 2017 the estimated number of testicular cancer patients diagnosed with testicular cancer in the prior 20 years was approximately 11,300 (Figure 2).7 In 2015, there were an estimated 257,800 men living with a history of testicular cancer

in the United States (data from Surveillance, Epidemiology, and End Results (SEER)). Due to the successful treatment, testicular cancer could serve as a model for the care for long-term cancer survivors: survivorship care. It has become increasingly clear that potential harmful long-term and late side eff ects of the treatment can have a major impact on the health-related quality of life after treatment. Long-term eff ects are side eff ects that appear during treatment and persist when the treatment is fi nished. In contrast, late eff ects are side eff ects that develop gradually and become

0 3 6 9 12 1989 1992 1995 1998 2001 2004 2007 2010 2013 2016

(13)

0 2000 4000 6000 8000 10000 12000 2009 2010 2011 2012 2013 2014 2015 2016 2017

Figure 2. 20-years prevalence of testicular cancer in The Netherlands (data Figure 1 and 2 from the Netherlands Comprehensive Cancer Organization).

manifest months to years after treatment. The health status of survivors should be monitored for long-term and late eff ects and, if indicated, preventive strategies and/or therapeutic options should be explored.

Figure 2. 20-years prevalence of testicular cancer in The Netherlands

(data Figure 1 and 2 from The Netherlands Comprehensive Cancer Organization).

Long-term and late toxicity in testicular cancer survivors

In general, testicular cancer survivors have a very good prognosis as demonstrated by SEER data; the life-expectancy of a testicular cancer patients diagnosed at age 30 years was estimated as 45 years; two years less than a man of the same age without cancer.8 However, after the treatment

a signifi cant proportion of testicular cancer survivors is aff ected by long-term and late toxicity of the treatment.9 The most threatening late eff ects are second cancers and cardiovascular disease.

Other long-term and late eff ects of treatment that are frequently observed after platinum-based chemotherapy are neurotoxicity, renal toxicity and hypogonadism.10

Early observations of acute cardiovascular toxicity during platinum-based chemotherapy were reported in the eighties.11 In the nineties, an increased prevalence of several cardiovascular

risk factors was noticed.12 In 2006, the 20-year risk for cardiovascular disease was estimated

to be 18.1% in a large Dutch cohort of 2500 testicular cancer survivors.13 Cardiovascular risk

factors, sometimes clustered in the metabolic syndrome, were more frequently observed in comparison with age-matched controls.14 Patients treated with the standard chemotherapy

regime consisting of bleomycin, etoposide, and cisplatin have a hazard ratio for coronary artery disease of 5.7 (95% confi dence interval [CI], 1.9–17.1) compared to age-matched controls.15 Early

onset cardiovascular disease was also found in a population-based study with more than 15,000 patients with a standardized mortality ratio of 1.36 (95% CI, 1.03–1.78).16 Another distinct example

(14)

patients is Raynaud’s phenomenon, a typical discoloration of the fingers after exposure to cold temperatures or emotion.17

The etiology of late cardiovascular side-effects of chemotherapy is not yet fully elucidated. Firstly, it may be that chemotherapeutic agents directly induce vascular damage or toxic changes. There are signs of vascular damage observed, such as microalbuminuria.18 It is

known that residuals of the platinum component of the treatment is circulating for many years after the treatment.19 It is unknown whether this circulating platinum is causally related to late

cardiovascular toxicity.

Secondly, risk factors may play a role in the development of cardiovascular disease. Hypogonadism is often observed in testicular cancer survivors and is considered to be a contributing factor to the risk at cardiovascular disease. Low testosterone is in part the result of the treatment, but it could also be influenced by obesity and altered androgen metabolism. Because it is observed that not all survivors are susceptible for developing both short-term and long-term effects, identifying genetic susceptibility could help in indicating specific subgroups that are particularly at risk for late effects. Specific variations in genes (single nucleotide polymorphisms (SNPs)) may identify patients who are especially vulnerable for developing long-term toxicity. Such a group of selected patients are candidates to develop and test interventional therapies to manage and prevent long-term and late toxicity. This method was for example used to evaluate the influence of a SNP in the gene encoding bleomycin hydrolase, BLMH, on outcome of testicular patients treated with bleomycin-cisplatin combination chemotherapy. Survival of patients was associated with the SNP in this bleomycin hydrolase gene, whereas bleomycin lung toxicity itself was not.20

Implications for the organization of survivorship care

The growing population of cancer survivors demands new ways to organize the care after cancer. Collaboration between medical oncologists and primary care physicians may improve early detection and management of, cardiovascular risk factors and lead to complementary psychosocial support close to where patients live. The high prevalence of cardiovascular disease risk factors and the metabolic syndrome in testicular cancer survivors makes collaboration between primary care and oncology especially important.14 If the primary care physician is involved in early

follow-up after completion of treatment, the long-term care after completion of the follow-follow-up is better secured. Because most of the patients treated for testicular cancer will not develop a relapse, they will be discarded from oncologic follow-up after ten years. However, especially this patient group stays at an increased risk for cardiovascular events for several decades after the treatment.

Since patients are often more familiar with their own primary care physician, the primary care physician is well-equipped to manage psychosocial issues of the patients, as well as their family members, both early and late after the cancer diagnosis. Also, after follow-up, the primary care physician is the most approachable and primarily the coordinating care provider. In an observational study by Dahl, testicular cancer survivors had a higher chance to seek help from the primary care physician.21 A study by De Padova et al. described the opinion of patients and care

(15)

providers. It was observed in this study that among patients and care providers uncertainties exist about the roles and responsibilities of the different physicians, supporting the need for adequate survivorship care planning.22

Primary care-led follow-up was successfully implemented for different types of cancer, e.g. breast cancer, colon cancer and melanoma.23-25 Shared-care follow-up for childhood cancer

survivors was studied in our center by Blaauwbroek et al.26,27 They concluded that both cancer

survivors and primary care physicians are willing to participate in shared-care follow-up. However, currently, in the case of testicular cancer, the collaboration between oncologists and primary care physicians is poorly coordinated and organized. As an example of shared-care follow-up, the study by Blaauwbroek was organized with a web-based survivorship care plan, accessible for patients and health care providers. Participation and satisfaction levels for both patients and primary care physicians were high. The development of a web-based communication system made it feasible to organize follow-up appointments in primary care and secondary care units.

Currently, standard follow-up care after testicular cancer takes place exclusively in secondary and tertiary care such as a comprehensive cancer center. Therefore, the safety of shared-care follow-up, with narrow collaboration between primary and secondary health care providers, has to be evaluated before other aspects and potential benefits can be explored. A new model of shared survivorship care will demand a more active role of the patients. Also, clear communication between the different care providers and the patient will be important. Exploratory studies are needed to advance the methods of follow-up and further optimize the quality of survivorship care.

Aim of the thesis

The studies described in this thesis, aim to improve our understanding of the mechanisms of treatment-related morbidity in testicular cancer patients and to develop and evaluate a new model of shared-care survivorship care after testicular cancer.

Outline of the thesis

The focus of the first part of this dissertation lies on cardiovascular late toxicity of testicular cancer survivors. The presented studies evaluate short- and long-term markers for and development of late toxicity.

In chapter 2, a review is presented of late toxicity of platinum-based treatment after testicular cancer. Among the most frequently occurring adverse conditions are cardiovascular disease and secondary neoplasms. Recommendations are given for survivorship care.

In chapter 3, serum platinum (Pt) decay after chemotherapy was assessed and modelled and the relationship between long-term circulating Pt levels and known late effects was Chapter 1

(16)

a population pharmacokinetic model, measured Pt data were simultaneously analysed, together with cisplatin dose, age, weight and height using NONMEM software. Based on this model, area under the curve 1–3 years after treatment was calculated for each patient. Predicted long-term Pt exposure was related to renal function and to late effects of treatment assessed median 9 (3–15) years after treatment.

The indirect effects of chemotherapy regarding cardiovascular disease risk factors, such as changes in metabolic processes, have to be taken into account too and are perhaps more important that direct toxic effects of the cancer treatment on vascular structures. Genetic variations resulting in changes in susceptibility for late toxicity can provide insight in which patients are particularly at risk for late effects and can also provide insight in the pathways along which late effects tend to develop. Genetic variation in androgen metabolism could in turn be associated with variation in adverse cardiometabolic changes after treatment. In chapter 4, a functional SNP in the gene SRD5A2 is described. The gene SRD5A2 encodes 5-alpha-reductase, the enzyme that converts testosterone into the more potent metabolite dihydrotestosterone.

Vascular events during treatment occur in a subgroup of patients. An intervention to prevent these events is not yet available. Acute changes during the period of active chemotherapy may predict how and in what subpopulation of patients late effects will develop. In chapter 5, acute changes during the chemotherapy are described. Changes in vascular markers could be used as biomarkers for early damage as a result of the treatment. The aim of this study is to identify risk factors for early cardiovascular damage.

In the second part of this dissertation, research on the organization of survivorship care is presented. A clear follow-up plan is an essential part in the organization of shared-care follow-up after treatment, during which visits alternate between oncologist and primary care physician. In the seminal report “Lost in transition”, it was argued that each patient should receive a survivorship care plan in order to receive the appropriate after care.26 Although this report was published more

than a decade ago, the implementation of this recommendation is limited. Since then, research has lost focus of the central idea, namely the plan itself. In chapter 6, a letter to the editor is presented that was written as part of a discussion on survivorship care planning. The use and development of a mobile application for generating survivorship care plans is explained in details in chapter 7. The purpose of this mobile application is to provide patients with a concise and simple care plan during survivorship care. The aim is to develop a tool that is available for other hospitals, both in the Netherlands and in other countries, treating patients with disseminated testicular cancer. With this care plan containing tool, patients can become more in control and navigation of his own care.

In chapter 8 a shared-care survivorship care study is described. The study is designed as an observational cohort study with a stopping rule to check for the safety of follow-up. Safety boundaries are defined for the occurrence of failed responses to signals indicating cancer recurrence. Secondary outcomes are feasibility, cardiovascular risk management, psychosocial status and experiences of patients and primary care physicians, as measured with an evaluation questionnaire.

Finally, data from this thesis are summarized in chapter 9. Findings from the current studies are placed into perspective and recommendations are given for future research.

(17)

References

Trabert B, Chen J, Devesa SS, et al: International patterns and trends in testicular cancer incidence, overall and by histologic subtype, 1973-2007. Andrology 3:4-12, 2015

Rosen A, Jayram G, Drazer M, et al: Global trends in testicular cancer incidence and mortality. Eur Urol 60:374-9, 2011

Hanna N, Einhorn LH: Testicular cancer: a reflection on 50 years of discovery. J Clin Oncol 32:3085-92, 2014

International Germ Cell Consensus Classification: a prognostic factor-based staging system for metastatic germ cell cancers. International Germ Cell Cancer Collaborative Group. J Clin Oncol 15:594-603, 1997 Feldman DR, Bosl GJ, Sheinfeld J, et al: Medical treatment of advanced testicular cancer. JAMA 299:672-84, 2008

Nichols CR, Roth B, Albers P, et al: Active surveillance is the preferred approach to clinical stage I testicular cancer. J Clin Oncol 31:3490-3, 2013

IKNL: http://www.cijfersoverkanker.nl, 2018

Capocaccia R, Gatta G, Dal Maso L: Life expectancy of colon, breast,

and testicular cancer patients: an analysis of US-SEER population-based data. Ann Oncol 26:1263-8, 2015 Kerns SL, Fung C, Monahan PO, et al: Cumulative Burden of Morbidity Among Testicular Cancer Survivors After Standard Cisplatin-Based Chemotherapy: A Multi-Institutional Study. J Clin Oncol 36:1505-1512, 2018

Haugnes HS, Bosl GJ, Boer H, et al: Long-term and late effects of germ cell testicular cancer treatment and implications for follow-up. J Clin Oncol 30:3752-63, 2012

Stefenelli T, Kuzmits R, Ulrich W, et al: Acute vascular toxicity after combination chemotherapy with cisplatin, vinblastine, and bleomycin for testicular cancer. Eur Heart J 9:552-6, 1988

Gietema JA, Sleijfer DT, Willemse PH, et al: Long-term follow-up of cardiovascular risk factors in patients given chemotherapy for disseminated nonseminomatous testicular cancer. Ann Intern Med 116:709-15, 1992

van den Belt-Dusebout AW, Nuver J, de Wit R, et al: Long-term risk of cardiovascular disease in 5-year survivors of testicular cancer. J Clin Oncol 24:467-75, 2006

de Haas EC, Altena R, Boezen HM, et al: Early development of the metabolic syndrome after chemotherapy for testicular cancer. Ann Oncol 24:749-55, 2013

Haugnes HS, Wethal T, Aass N, et al: Cardiovascular risk factors and morbidity in long-term survivors of testicular cancer: a 20-year follow-up study. J Clin Oncol 28:4649-57, 2010

Fung C, Fossa SD, Milano MT, et al: Cardiovascular Disease Mortality After Chemotherapy or Surgery for Testicular Nonseminoma: A Population-Based Study. J Clin Oncol 33:3105-15, 2015

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:2229-38, 1995

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 40:701-6, 2004

Gietema JA, Meinardi MT, Messerschmidt J, et al: Circulating plasma platinum more than 10 years after cisplatin treatment for testicular cancer. Lancet 355:1075-6, 2000

de Haas EC, Zwart N, Meijer C, et al: Variation in bleomycin hydrolase gene is associated with reduced survival after chemotherapy for testicular germ cell cancer. J Clin Oncol 26:1817-23, 2008

Dahl CF, Haugnes HS, Bremnes R, et al: A controlled study of risk factors for disease and current problems

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Chapter 1

(18)

De Padova S, Rosti G, Scarpi E, et al: Expectations of survivors, caregivers and healthcare providers for testicular cancer survivorship and quality of life. Tumori 97:367-73, 2011

Grunfeld E, Levine MN, Julian JA, et al: Randomized trial of long-term follow-up for early-stage breast cancer: a comparison of family physician versus specialist care. J Clin Oncol 24:848-55, 2006

Murchie P, Nicolson MC, Hannaford PC, et al: Patient satisfaction with GP-led melanoma follow-up: a randomised controlled trial. Br J Cancer 102:1447-55, 2010

Wattchow DA, Weller DP, Esterman A, et al: General practice vs surgical-based follow-up for patients with colon cancer: randomised controlled trial. Br J Cancer 94:1116-21, 2006

Blaauwbroek R, Tuinier W, Meyboom-de Jong B, et al: Shared care by paediatric oncologists and family doctors for long-term follow-up of adult childhood cancer survivors: a pilot study. Lancet Oncol 9:232-8, 2008

Blaauwbroek R, Barf HA, Groenier KH, et al: Family doctor-driven follow-up for adult childhood cancer survivors supported by a web-based survivor care plan. J Cancer Surviv 6:163-71, 2012

22 23 24 25 26 27 General introduction

(19)
(20)

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

(21)

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.

(22)

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.

(23)

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.

(24)

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). Abbr 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

Ta

bl

es

c

ha

pt

er

2

Ta 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 er s 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 in g 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 er (R R 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 nt ed 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 te r 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 ce rs ra ng ed b et w ee n 1 an d 2, a nd in cre as ed w ith in cre as in g 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 s fo llo w -up . T he ri sk o f s ec on d ca nc er w as 2 .6 (1 .7 -4. 0) 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 ith a n 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 te my el oi d le uk emi a an d ac ut e ly mp ho bl as tic le uk emi a. Ex ce ss c um ul at iv e le uk em ia ri sk w as 0 .2 3% b y 30 y ea rs af te r T C di ag no sis . Ab 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 lu te ris k

(25)

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.

(26)

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

as the key predictor of RP.36, 37, 39 However, the cumulative cisplatin dose may also contribute.37

(27)

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) . 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, 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. si on c rit er io n: a ge ≤ 50 at ti m e of a na ly sis . 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 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. t 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 in ar di 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. ar t 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 . den sebo ut 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) ne s 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 . 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). 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 ; 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 . erion: age ≤ 50 at time of analysis . 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 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 .

(28)

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

be limited by vigorous hydration during treatment, but it cannot be completely avoided.70

(29)

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

(30)

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,

(31)

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.

(32)

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

post-chemotherapy RPLND.120

(33)

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 cluded)* 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 , ap y; R T, r adiother ap y; FSH, follicle stim ulating hormone . tilit y tr eatment. T he use of cr yopr eser ved semen w as not speci fied . ap y gr

oups included patients additionally tr

eat ed with R T or RPLND . es r efer t o 15-y actuarial r at es . 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 at io n ex cl ud ed )* . 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 m on (20 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 . he (2 00 4) ) 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 ). ar t ( 20 05 ) 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 ) øy (2 00 5) ) 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) 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 , 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. 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. 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 . s re fe r to 1 5-y ac tu ar ia l r at e

(34)

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,

133 but not others.132

(35)

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

Referenties

GERELATEERDE DOCUMENTEN

Survivorship care after testicular cancer: New insights in late effects of treatment and approaches to shared-care follow-up..

Haugnes HS, Wethal T, Aass N, et al: Cardiovascular risk factors and morbidity in long-term survivors of testicular cancer: a 20-year follow-up study. J Clin Oncol

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 renal parameters, and comparison of Pt exposure in groups based on serum creatinine change. Median change in highest quartile: +26 µmol/L; median change in lower three

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