100% found this document useful (1 vote)
80 views224 pages

Acute Side Effects of Radiation Therapy - A Guide To Management (PDFDrive)

The document is a guide on the acute side effects of radiation therapy, detailing their management and implications for cancer treatment. It emphasizes the importance of understanding these side effects to improve patient quality of life and treatment outcomes. The guide includes information on various acute side effects, their mechanisms, symptoms, prevention, and management strategies.

Uploaded by

decafisajoke59
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
100% found this document useful (1 vote)
80 views224 pages

Acute Side Effects of Radiation Therapy - A Guide To Management (PDFDrive)

The document is a guide on the acute side effects of radiation therapy, detailing their management and implications for cancer treatment. It emphasizes the importance of understanding these side effects to improve patient quality of life and treatment outcomes. The guide includes information on various acute side effects, their mechanisms, symptoms, prevention, and management strategies.

Uploaded by

decafisajoke59
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 224

Ainaz Sourati · Ahmad Ameri

Mona Malekzadeh

Acute Side Effects of


Radiation Therapy

A Guide to Management

123
Acute Side Effects of Radiation Therapy
Ainaz Sourati • Ahmad Ameri
Mona Malekzadeh

Acute Side Effects of


Radiation Therapy
A Guide to Management
Ainaz Sourati Ahmad Ameri
Department of Clinical Oncology Department of Clinical Oncology
Imam Hossein Educational Hospital, Shahid Imam Hossein Educational Hospital, Shahid
Beheshti University of Medical Sciences Beheshti University of Medical Sciences
(SBMU), Shahid Madani Street (SBMU), Shahid Madani Street
Tehran Tehran
Iran Iran

Mona Malekzadeh
Department of Radiotherapy and Oncology
Shohadaye Tajrish Educational Hospital
Shahid Beheshti University of Medical
Sciences (SBMU)
Tehran
Iran

ISBN 978-3-319-55949-0    ISBN 978-3-319-55950-6 (eBook)


DOI 10.1007/978-3-319-55950-6

Library of Congress Control Number: 2017944188

© Springer International Publishing AG 2017


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of
the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recita-
tion, broadcasting, reproduction on microfilms or in any other physical way, and transmission or infor-
mation storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar
methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publica-
tion does not imply, even in the absence of a specific statement, that such names are exempt from the
relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, express or implied, with respect to the material contained herein or for any errors
or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims
in published maps and institutional affiliations.

Printed on acid-free paper

This Springer imprint is published by Springer Nature


The registered company is Springer International Publishing AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
To my dear mother who I will forever be
beholden to her.
To my loving husband, Arash, who I really
appreciate his support and endurance
through my most trying times.
Ainaz Sourati
To my love Zhila.
Ahmad Ameri
To my dear parents and my beloved son,
Hooman.
Mona Malekzadeh
Foreword

Dichotomies in medicine are common, but it is nowhere more clearly demonstrated


than in radiation exposure either used in diagnoses or in treatment as compared to
excessive or unwanted radiation exposure either locally or systematically. Other
dichotomies demonstrated in radiation injury include the separation between acute
and late radiation damage, locally applied as contrasted to systemic or total body
exposure, one or few radiation exposures vs. fractionated radiation exposures, and
use of preventive or ameliorating agents applied before exposure in contrast to post-­
exposure treatments. All of these are important to our understanding of the effective,
appropriate use of radiation as a treatment modality. This can only be ameliorated
by accurate information about the causes, frequency, severity, and prevention or
treatment of these radiation-associated conditions.
In the clinic, observed reactions to radiotherapy range from minor to serious and
from temporary to permanent. Physician concern for these potential debilitating
effects permeates the clinical practice of radiation and influences the selection of
treatments on a day-to-day basis. Optimal utilization of radiation as a cancer ther-
apy requires a clear understanding of the range of the major tissue sequela that can
occur, including the risk of occurrence, natural history, and their management. This
understanding leads to better clinical practice: greater confidence in recommending
aggressive radiation treatment programs when appropriate or selection of other
therapies when they are safer and equally effective.
Acute reactions of radiation therapy, the inflammatory reactions that occur as
part of normal tissue reaction to radiotherapy, are often considered as an expected
part of therapy, but they also may become debilitating to the patient during the treat-
ment course. Understanding of the mechanism, timing of occurrence, preventable
measures, and management of these side effects warrant discussion of their success-
ful management, including ways to facilitate more comfortable therapy course for
the patient.
In the era of multimodality therapy, it’s also important to understand the risk fac-
tors of radiation therapy when combined with other modalities, i.e., systemic che-
motherapy or biological agents. It is also important to realize the impact of other
modalities as sole cause of side effect or as a co-positive impact along with radia-
tion. Therefore, clinicians must be aware of these alternative possibilities and the
method diagonals and modify them.

vii
viii Foreword

In this volume, the clinician-scientists of Shahid Beheshti University and Dr.


Sourati and her associates have successfully provided to the readers the plethora of
these concerns in a comprehensive yet relatively straightforward and simple text for
day-to-day use of clinicians. Organ-specific effects are detailed as appropriate to
understanding, minimizing, and managing radiation injuries. I congratulate them in
their successful effort, and I am certain that many clinicians and their patients would
significantly benefit from the information provided by these authors.

Maywood, IL, USA Bahman Emami, MD, FACR, FASTRO, FACRO


Introduction

Cancer is one of the leading causes of death worldwide [1]. Currently, cancer
patients survive longer than they did more than two decades ago, and the population
living with cancer is increasing. Understanding more about cancer radiobiology and
major developments in radiation therapy technology play a critical role in increas-
ing the life expectancy of these patients. Radiation therapies are used with three
different concepts in cancer that are definitive, adjuvant, or palliative. It has been
estimated that more than 50% of all cancer patients receive radiation therapy
throughout the course of their disease [2]. With new technological advancements in
imaging and radiation delivery, radiation therapy has been possible for more com-
plicated cases, increasing the rate of radiation therapy in cancer treatments.
In addition to the tumor sterility and tumoricidal effects of radiation therapy,
radiation has the potential to affect normal tissues, which includes tissue damage in
radiation therapy fields or systemic side effects (e.g., hematologic side effects,
fatigue). Physicians try to lower radiation therapy side effects by defining smaller
targets and using more dedicated machines, but side effects continue to occur and
their spectrum and severity are changing. As these patients live longer, more atten-
tion is directed on their quality of life.
Radiation therapy side effects are divided into acute and late. Late side effects
occur 3 months after treatment completion, and acute side effects occur during
treatment or within 3 months after treatment.
Late side effects can develop without dose thresholds, which include stochastic
side effects, or with distinctive thresholds, which include deterministic side effects
(e.g., neurologic effects, cardiac effects). Deterministic late side effects are dose-­
limiting and can be the result of the healing process following severe acute side
effects. This kind of late side effects could be predictable, and efforts should be
made to protect normal tissues [3].
Acute side effects are a type of deterministic effects, which have distinctive
thresholds, and there is a direct relation with delivered dose and severity of resultant
damage. These effects usually cause no limit in treatment dose because they occur
in rapidly dividing cells, which have rapid cell repopulation; as a result, they are
reversible. The main point in acute side effects is necessary actions for prevention
and proper treatment in accordance with their severity to prevent radiation therapy
disruption because treatment disruption can affect treatment results, particularly in
tumors with rapid cell growth, which intensify cell growth inversely. Severe acute

ix
x Introduction

side effects can also result in consecutive late effects. Using new techniques in radi-
ation therapy (e.g., 3D-conformal radiation therapy, intensified modulated radiation
therapy, image-guided radiation therapy) reduces normal tissue dose and conse-
quently reduces treatment complications. However, the percentage of acute side
effects is still noticeable because the dose threshold in these side effects’ occurrence
is less than late side effects. Furthermore, in some treatments, it is necessary to
increase radiation dose to reach a definite complication (e.g., in locally advanced
breast cancer after radical mastectomy, enough radiation must be delivered to reach
dermatitis). We have to know the proper actions in approaching the complications
in order to inhibit unwanted treatment disruption. Acute side effects may increase
risk of late side effects, and prevention of these effects should be considered.
With respect to all points, acute side effects of radiation therapy are one of the
most important issues in the treatment of cancer patients. Approach and manage-
ment of these side effects are substantial to improve treatment results and increase
patient compliance with treatment. We attempt to assemble the most common of
these acute side effects in this book. In each part, we describe a summary of inci-
dence, mechanism, symptoms, grading, and management of distinct acute side
effects based on available evidence. Our main goal is gathering articles about each
acute side effect for convenient and practical use for all involved in the radiation
therapy process.

References

1. Stewart BW, Wild C (2014) World Cancer Report 2014. International Agency for Research on
Cancer. World Health Organization. 505
2. Slotman BJ, Cottier B, Bentzen SM, Heeren G, Lievens Y, Van den Bogaert W (2005) Overview
of national guidelines for infrastructure and staffing of radiotherapy. ESTRO-QUARTS: work
package 1. Radiother Oncol 75(3):349. E1–E6
3. ICRP (1977) Recommendations of the International Commission on Radiological Protection.
Ann ICRP. Sect. ICRP Publication 26
Contents

1 Radiation Dermatitis ���������������������������������������������������������������������������������� 1


1.1 Mechanism�������������������������������������������������������������������������������������������� 1
1.2 Timing�������������������������������������������������������������������������������������������������� 2
1.3 Sign and Symptoms������������������������������������������������������������������������������ 3
1.4 Scoring�������������������������������������������������������������������������������������������������� 4
1.5 Risk Factors������������������������������������������������������������������������������������������ 4
1.5.1 Treatment-Related Factors Including���������������������������������������� 4
1.5.2 Patient-Related Factors Include������������������������������������������������ 5
1.6 Diagnosis���������������������������������������������������������������������������������������������� 6
1.7 Prevention �������������������������������������������������������������������������������������������� 7
1.7.1 General Measures��������������������������������������������������������������������� 7
1.7.2 Intensity-Modulated Radiation Therapy (IMRT)���������������������� 8
1.7.3 Low-Level Laser Therapy�������������������������������������������������������� 8
1.7.4 Amifostine�������������������������������������������������������������������������������� 8
1.7.5 Ascorbic Acid��������������������������������������������������������������������������� 8
1.7.6 Oral Zinc ���������������������������������������������������������������������������������� 8
1.7.7 Silver Leaf Dressing ���������������������������������������������������������������� 9
1.7.8 MAS065D �������������������������������������������������������������������������������� 9
1.7.9 Soft Dressing or Barrier Film �������������������������������������������������� 9
1.7.10 Hyaluronic Acid������������������������������������������������������������������������ 9
1.7.11 Trolamine���������������������������������������������������������������������������������� 9
1.7.12 Topical Sucralfate ������������������������������������������������������������������ 10
1.7.13 Theta Cream���������������������������������������������������������������������������� 10
1.7.14 Dexpanthenol (Provitamin B5) ���������������������������������������������� 10
1.7.15 Calendula�������������������������������������������������������������������������������� 10
1.7.16 Pentoxifylline�������������������������������������������������������������������������� 10
1.7.17 Aloe Vera�������������������������������������������������������������������������������� 10
1.7.18 Deodorant�������������������������������������������������������������������������������� 11
1.7.19 Laughter Therapy�������������������������������������������������������������������� 11
1.8 Management���������������������������������������������������������������������������������������� 11
1.8.1 Grade 1 Dermatitis������������������������������������������������������������������ 11
1.8.2 Grade 2–3 Dermatitis�������������������������������������������������������������� 12
1.8.3 Grade 4 Dermatitis������������������������������������������������������������������ 13
References���������������������������������������������������������������������������������������������������� 13

xi
xii Contents

2 Hair Loss���������������������������������������������������������������������������������������������������� 21
2.1 Mechanism������������������������������������������������������������������������������������������ 22
2.2 Timing������������������������������������������������������������������������������������������������ 22
2.2.1 Risk Factors���������������������������������������������������������������������������� 22
2.2.2 Symptoms ������������������������������������������������������������������������������ 22
2.3 Scoring������������������������������������������������������������������������������������������������ 23
2.4 Prevention ������������������������������������������������������������������������������������������ 23
2.5 Management���������������������������������������������������������������������������������������� 24
References���������������������������������������������������������������������������������������������������� 24
3 Radiation Brain Injury������������������������������������������������������������������������������ 27
3.1 Mechanism������������������������������������������������������������������������������������������ 28
3.2 Timing������������������������������������������������������������������������������������������������ 28
3.3 Risk Factors���������������������������������������������������������������������������������������� 28
3.3.1 Treatment-Related Factors������������������������������������������������������ 28
3.3.2 Patient-Related Factor������������������������������������������������������������ 29
3.4 Symptoms ������������������������������������������������������������������������������������������ 29
3.5 Diagnosis�������������������������������������������������������������������������������������������� 30
3.6 Scoring������������������������������������������������������������������������������������������������ 31
3.7 Prevention and Management�������������������������������������������������������������� 31
3.7.1 Dexamethasone Prescription�������������������������������������������������� 32
References���������������������������������������������������������������������������������������������������� 33
4 Radiation Orbital Toxicity������������������������������������������������������������������������ 39
4.1 Blepharitis and Eyelid Dermatitis������������������������������������������������������ 40
4.2 Eyelash Loss �������������������������������������������������������������������������������������� 40
4.3 Conjunctivitis�������������������������������������������������������������������������������������� 40
4.4 Xerophthalmia������������������������������������������������������������������������������������ 41
4.5 Corneal Toxicity���������������������������������������������������������������������������������� 43
4.5.1 Treatment�������������������������������������������������������������������������������� 43
4.6 Iris Toxicity ���������������������������������������������������������������������������������������� 44
References���������������������������������������������������������������������������������������������������� 44
5 Ear Toxicity������������������������������������������������������������������������������������������������ 47
5.1 Mechanism������������������������������������������������������������������������������������������ 47
5.2 Timing������������������������������������������������������������������������������������������������ 48
5.3 Risk Factors���������������������������������������������������������������������������������������� 48
5.4 Symptoms and Diagnosis�������������������������������������������������������������������� 49
5.5 Scoring������������������������������������������������������������������������������������������������ 49
5.6 Management���������������������������������������������������������������������������������������� 50
References���������������������������������������������������������������������������������������������������� 51
6 Oral Mucositis�������������������������������������������������������������������������������������������� 53
6.1 Mechanism������������������������������������������������������������������������������������������ 53
6.2 Timing������������������������������������������������������������������������������������������������ 54
6.3 Risk Factors���������������������������������������������������������������������������������������� 55
6.3.1 Tumor Site������������������������������������������������������������������������������ 55
6.3.2 Concomitant Systemic Therapy���������������������������������������������� 55
Contents xiii

6.3.3 Radiation Dose������������������������������������������������������������������������ 55


6.3.4 Radiation Fractionation Schedule ������������������������������������������ 56
6.3.5 Patient-Related Factors ���������������������������������������������������������� 56
6.4 Symptoms ������������������������������������������������������������������������������������������ 57
6.5 Scoring������������������������������������������������������������������������������������������������ 58
6.6 Prevention ������������������������������������������������������������������������������������������ 59
6.6.1 Prophylactic Interventions������������������������������������������������������ 59
6.6.2 Prophylactic Intervention Under Evaluation�������������������������� 60
6.7 Management���������������������������������������������������������������������������������������� 63
6.7.1 Patient Assessment������������������������������������������������������������������ 63
6.7.2 Mouth Care ���������������������������������������������������������������������������� 64
6.7.3 Management of Oral Pain ������������������������������������������������������ 64
6.7.4 Antifungals������������������������������������������������������������������������������ 68
6.7.5 Antivirals�������������������������������������������������������������������������������� 68
6.7.6 Feeding Tube/Nutritional Support������������������������������������������ 69
References���������������������������������������������������������������������������������������������������� 69
7 Xerostomia�������������������������������������������������������������������������������������������������� 79
7.1 Mechanism������������������������������������������������������������������������������������������ 79
7.2 Timing������������������������������������������������������������������������������������������������ 81
7.3 Risk Factors���������������������������������������������������������������������������������������� 81
7.4 Symptoms ������������������������������������������������������������������������������������������ 82
7.5 Diagnosis�������������������������������������������������������������������������������������������� 83
7.6 Scoring������������������������������������������������������������������������������������������������ 83
7.7 Prevention ������������������������������������������������������������������������������������������ 84
7.7.1 Amifostin�������������������������������������������������������������������������������� 85
7.7.2  IMRT�������������������������������������������������������������������������������������� 86
7.7.3 Submandibular gland transfer ������������������������������������������������ 86
7.7.4 Pilocarpine ���������������������������������������������������������������������������� 87
7.7.5 Acupuncture���������������������������������������������������������������������������� 88
7.8 Management���������������������������������������������������������������������������������������� 89
7.8.1 Supportive Care���������������������������������������������������������������������� 89
7.8.2 Saliva Supplementation������������������������������������������������������������ 90
7.8.3 Acupuncture������������������������������������������������������������������������������ 89
7.8.4 Drugs���������������������������������������������������������������������������������������� 91
References���������������������������������������������������������������������������������������������������� 92
8 Loss of Taste����������������������������������������������������������������������������������������������� 97
8.1 Mechanism������������������������������������������������������������������������������������������ 97
8.2 Timing������������������������������������������������������������������������������������������������ 98
8.3 Risk Factors���������������������������������������������������������������������������������������� 99
8.4 Symptoms ������������������������������������������������������������������������������������������ 99
8.5 Diagnosis�������������������������������������������������������������������������������������������� 99
8.6 Scoring���������������������������������������������������������������������������������������������� 100
8.7 Prevention ���������������������������������������������������������������������������������������� 100
8.8 Management�������������������������������������������������������������������������������������� 100
References�������������������������������������������������������������������������������������������������� 102
xiv Contents

9 Laryngeal Edema ������������������������������������������������������������������������������������ 105


9.1 Mechanism���������������������������������������������������������������������������������������� 105
9.2 Timing���������������������������������������������������������������������������������������������� 105
9.3 Risk Factors�������������������������������������������������������������������������������������� 106
9.4 Symptoms ���������������������������������������������������������������������������������������� 106
9.5 Scoring���������������������������������������������������������������������������������������������� 106
9.6 Prevention ���������������������������������������������������������������������������������������� 107
9.7 Management�������������������������������������������������������������������������������������� 107
References ������������������������������������������������������������������������������������������������ 107
10 Radiation Pneumonitis���������������������������������������������������������������������������� 109
10.1 Mechanism�������������������������������������������������������������������������������������� 109
10.2 Timing�������������������������������������������������������������������������������������������� 110
10.3 Risk Factors������������������������������������������������������������������������������������ 110
10.3.1 Volume and Dose Parameters�������������������������������������������� 110
10.3.2 Fractionation Schedule������������������������������������������������������ 110
10.3.3 Chemotherapy/Hormone Therapy�������������������������������������� 111
10.3.4 Smoking ���������������������������������������������������������������������������� 111
10.3.5 Other Factors���������������������������������������������������������������������� 111
10.4 Symptoms �������������������������������������������������������������������������������������� 111
10.5 Diagnosis���������������������������������������������������������������������������������������� 112
10.6 Scoring�������������������������������������������������������������������������������������������� 112
10.7 Prevention �������������������������������������������������������������������������������������� 113
References�������������������������������������������������������������������������������������������������� 114
11 Pericarditis������������������������������������������������������������������������������������������������ 117
11.1 Mechanism�������������������������������������������������������������������������������������� 118
11.2 Timing�������������������������������������������������������������������������������������������� 118
11.3 Risk Factors������������������������������������������������������������������������������������ 118
11.4 Symptoms �������������������������������������������������������������������������������������� 119
11.5 Diagnosis���������������������������������������������������������������������������������������� 119
11.6 Scoring�������������������������������������������������������������������������������������������� 120
11.7 Prevention �������������������������������������������������������������������������������������� 121
11.8 Treatment���������������������������������������������������������������������������������������� 122
References�������������������������������������������������������������������������������������������������� 122
12 Esophagitis������������������������������������������������������������������������������������������������ 125
12.1 Mechanism�������������������������������������������������������������������������������������� 125
12.2 Timing�������������������������������������������������������������������������������������������� 125
12.3 Risk Factors������������������������������������������������������������������������������������ 126
12.4 Symptoms �������������������������������������������������������������������������������������� 127
12.5 Scoring�������������������������������������������������������������������������������������������� 127
12.6 Diagnosis���������������������������������������������������������������������������������������� 128
12.7 Prevention �������������������������������������������������������������������������������������� 128
12.8 Management������������������������������������������������������������������������������������ 128
References�������������������������������������������������������������������������������������������������� 129
Contents xv

13 Radiation Gastritis���������������������������������������������������������������������������������� 133


13.1 Mechanism�������������������������������������������������������������������������������������� 133
13.2 Risk Factors������������������������������������������������������������������������������������ 134
13.3 Timing�������������������������������������������������������������������������������������������� 134
13.4 Symptoms �������������������������������������������������������������������������������������� 135
13.5 Diagnosis���������������������������������������������������������������������������������������� 135
13.6 Prevention �������������������������������������������������������������������������������������� 135
13.7 Management������������������������������������������������������������������������������������ 135
References�������������������������������������������������������������������������������������������������� 136
14 Radiation-Induced Liver Disease ���������������������������������������������������������� 137
14.1 Mechanism�������������������������������������������������������������������������������������� 137
14.2 Risk Factors������������������������������������������������������������������������������������ 138
14.3 Timing�������������������������������������������������������������������������������������������� 138
14.4 Symptoms �������������������������������������������������������������������������������������� 139
14.5 Scoring�������������������������������������������������������������������������������������������� 139
14.6 Diagnosis���������������������������������������������������������������������������������������� 140
14.7 Prevention �������������������������������������������������������������������������������������� 140
14.8 Management������������������������������������������������������������������������������������ 141
References�������������������������������������������������������������������������������������������������� 142
15 Enteritis���������������������������������������������������������������������������������������������������� 145
15.1 Mechanism�������������������������������������������������������������������������������������� 145
15.2 Timing�������������������������������������������������������������������������������������������� 146
15.3 Risk Factors������������������������������������������������������������������������������������ 146
15.4 Symptoms and Diagnosis���������������������������������������������������������������� 147
15.5 Scoring�������������������������������������������������������������������������������������������� 147
15.6 Prevention �������������������������������������������������������������������������������������� 148
15.7 Management������������������������������������������������������������������������������������ 149
15.7.1 Dietary Modification�������������������������������������������������������� 150
15.7.2 Antidiarrheals�������������������������������������������������������������������� 150
15.8 Antispasmodics ������������������������������������������������������������������������������ 150
References�������������������������������������������������������������������������������������������������� 151
16 Radiation Cystitis������������������������������������������������������������������������������������ 155
16.1 Mechanism�������������������������������������������������������������������������������������� 155
16.2 Timing�������������������������������������������������������������������������������������������� 156
16.3 Risk Factors������������������������������������������������������������������������������������ 156
16.4 Symptoms and Diagnosis���������������������������������������������������������������� 157
16.5 Scoring�������������������������������������������������������������������������������������������� 157
16.6 Prevention �������������������������������������������������������������������������������������� 158
16.6.1 Bladder Sparing���������������������������������������������������������������� 158
16.6.2 Intravesical Instillation������������������������������������������������������ 158
16.7 Management������������������������������������������������������������������������������������ 158
16.7.1 Anticholinergics���������������������������������������������������������������� 159
16.7.2 Alpha-1 Blocker���������������������������������������������������������������� 159
16.7.3 Analgesics ������������������������������������������������������������������������ 160
16.7.4 Intravesical GAG�������������������������������������������������������������� 160
References�������������������������������������������������������������������������������������������������� 160
xvi Contents

17 Radiation Proctitis ���������������������������������������������������������������������������������� 165


17.1 Mechanism�������������������������������������������������������������������������������������� 165
17.2 Risk Factors������������������������������������������������������������������������������������ 165
17.3 Timing�������������������������������������������������������������������������������������������� 166
17.4 Symptoms �������������������������������������������������������������������������������������� 166
17.5 Scoring�������������������������������������������������������������������������������������������� 166
17.6 Prevention �������������������������������������������������������������������������������������� 167
17.7 Management������������������������������������������������������������������������������������ 168
References�������������������������������������������������������������������������������������������������� 169
18 Fatigue������������������������������������������������������������������������������������������������������ 173
18.1 Mechanism�������������������������������������������������������������������������������������� 173
18.2 Timing�������������������������������������������������������������������������������������������� 174
18.3 Risk Factors������������������������������������������������������������������������������������ 174
18.4 Symptoms �������������������������������������������������������������������������������������� 175
18.5 Diagnosis and Scoring�������������������������������������������������������������������� 176
18.6 Management������������������������������������������������������������������������������������ 178
18.6.1 Non-pharmacologic Interventions������������������������������������ 178
18.6.2 Exercise���������������������������������������������������������������������������� 179
18.6.3 Counseling and Education������������������������������������������������ 179
18.6.4 Optimize Sleep Quality���������������������������������������������������� 179
18.6.5 Complementary Therapies������������������������������������������������ 180
18.6.6 Other Psychosocial Interventions ������������������������������������ 180
18.6.7 Nutrition Counseling�������������������������������������������������������� 181
18.6.8 Pharmacologic Intervention���������������������������������������������� 181
References�������������������������������������������������������������������������������������������������� 182
19 Hematological Side Effects���������������������������������������������������������������������� 191
19.1 Mechanism�������������������������������������������������������������������������������������� 191
19.2 Timing�������������������������������������������������������������������������������������������� 192
19.3 Risk Factors������������������������������������������������������������������������������������ 193
19.4 Symptoms �������������������������������������������������������������������������������������� 193
19.5 Prevention �������������������������������������������������������������������������������������� 194
19.6 Treatment���������������������������������������������������������������������������������������� 195
19.6.1 Neutropenia���������������������������������������������������������������������� 195
19.6.2 Thrombocytopenia������������������������������������������������������������ 197
19.6.3 Anemia������������������������������������������������������������������������������ 198
References�������������������������������������������������������������������������������������������������� 199
20 Radiation-Induced Nausea and Vomiting (RINV)�������������������������������� 207
20.1 Mechanism�������������������������������������������������������������������������������������� 207
20.2 Timing�������������������������������������������������������������������������������������������� 208
20.3 Risk Factors������������������������������������������������������������������������������������ 208
20.4 Symptoms �������������������������������������������������������������������������������������� 208
20.5 Prevention and Management���������������������������������������������������������� 209
References�������������������������������������������������������������������������������������������������� 211
Index������������������������������������������������������������������������������������������������������������������ 213
Radiation Dermatitis
1

Anticancer properties of radiation were discovered by skin changes after exposure


to radiation; initially, radiation was used for the treatment of skin diseases such as
lupus erythematous. Skin cancer was the first cancer treated by radiation, and der-
matitis accompanies radiation therapy since its discovery.
Radiation dermatitis is one of the most common adverse effects of radiation
therapy. Approximately 90–95% [1, 2] of all patients treated with radiation ther-
apy will experience a skin reaction at the treated area. Radiation dermatitis is
especially seen in radiation therapy for breast cancer, head and neck cancer, lung
cancer, and sarcoma due to the superficial position of these cancers and higher
radiation doses to the skin. Various degrees of radiation dermatitis are experienced
by patients undergoing radiation therapy. In most patients, the radiation dermatitis
is mild to moderate (Grades 1 and 2); about 15–25% of patients experience severe
reactions [3–5].

1.1 Mechanism

Radiation to the skin results in direct cellular injury and inflammatory cell influx.
After the initial dose of radiation, cellular damage to epidermal basal cells, endothe-
lial cells, Langerhans cells, and vascular components occur, and an inflammatory
response in the epidermis and dermis develops following every subsequent fraction
of radiation [1].
Initial cellular injury results from short-lived free-radical production and irre-
versible double-stranded breaks in nuclear and mitochondrial DNA [6]. Clumping
of nuclear chromatin, swelling of the nucleus, nuclear disfiguration or loss of the
nuclear membrane, mitochondrial distortion, and degeneration of the endoplasmic

© Springer International Publishing AG 2017 1


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_1
2 1 Radiation Dermatitis

reticulum, as well as direct cellular necrosis and apoptosis, occur due to cellular
damage. Repetitive cellular damage caused from each fraction of treatment does not
allow time for cells to repair DNA damage [7–11].
Because of free-radical production, an inflammatory cytokine cascade is induced,
and several cytokines and chemokines are produced including interleukins 1 and 6,
tumor necrosis factor-α, and transforming growth factor-β [12]. These cytokines act
on the endothelial cells of local vessels, which express adhesion molecules consti-
tutively and induce migration of leukocytes and other immune cells from circulation
to irradiated skin [13].
Reduction and impairment of functional stem cells induced by radiation lead to
an alteration in the normal turnover of skin cells [6].
A prompt skin reaction may appear after exposure that is related to the inflamma-
tory response, release of histamine-like substances, permeability and dilation of capil-
laries, and subsequent dermal edema. It presents with early transient skin erythema
that can be seen within a few hours after radiation and subsides after 24–48 h [14].
After subsidence of initial erythema, the later phase includes additional edema,
an increased dilation in capillaries, and erythrocyte extravasation, which resulted in
an erythematous reaction. This phase is followed by thinning of the epidermis, dam-
age to epithelial cells, degeneration of glands, reduced secretion of sebum, and
sweat and clumps of exfoliated corneocytes (scales), which manifests as dry
desquamation.
If damage to the basal cells and glands is more severe, epidermal necrosis, fibrin-
ous exudates, and moist desquamation occur. The manifestation of moist desquama-
tion results from the formation of small blisters in and around the basal layer of the
epidermis that may also extend into the more superficial layers. The epidermis
sloughs when these blisters rupture and coalesce, denuding the dermis and causing
permanent epilation [15–17]. Finally, skin necrosis or ulceration of the full-­thickness
dermis may develop as the most severe damage of higher radiation doses [15, 16].

1.2 Timing

As noted above, a transient early erythema can be seen within a few hours of radia-
tion, even after a single fraction of radiation (2 Gy) and subsides after 24–48 h [14].
The main erythematous reaction appears at about the second to third week (within
1–4 weeks) of radiation. Earlier reaction can be seen in patients with particularly
sensitive skin. Effects typically continue to progress during treatment [18–20], and
subsequently dry desquamation can develop after the third week or after a cumula-
tive dose of 30 Gy, which is clinically characterized by dryness, scaling, and pruri-
tus. Following 4–5 weeks (45–60 Gy) of therapy, moist desquamation may occur,
which is characterized by serous oozing and exposure of the dermis [14]. Symptoms
persist for the duration of radiation therapy and peak 1–2 weeks after treatment
completion. At about 3–5 weeks after radiation, the skin begins to recover with
epidermal regeneration and within 1–3 months (depending on the severity of reac-
tion), complete healing occurs [1, 6, 18, 19, 21, 22]. Post-inflammatory
1.3 Sign and Symptoms 3

hyperpigmentation may persist for 5–7 weeks or even longer after radiation [20, 23].
Patient skin type may also influence timing and severity of radiation reactions,
reflected by interindividual variation in skin presentation of radiation damage.

1.3 Sign and Symptoms

Mild dermatitis (Grade I) presents with erythematous skin accompanied by mild


edema, pruritus, or pain. Dry, peeling skin (desquamation) and temporary epilation
due to involvement of adnexal structures may occur (Fig. 1.1).
Moderate dermatitis (Grades 2 and 3) consists of tender or edematous reactions
and moist desquamation (Fig. 1.2). The integrity of the dermis is impaired, and
secondary infection with Staphylococcus aureus may occur (Fig. 1.3).
Severe dermatitis (acute radionecrosis) is rarely seen and may occur in the set-
ting of very extensive superficial tumors treated with massive radiation doses over a
very short period. It manifests as a very painful inflammatory or hemorrhagic plaque
with deep necrosis, which can expose the muscles, tendons, and bones [19, 21, 22].

Fig. 1.1 Grade 1


dermatitis of neck after
radiation therapy for
laryngeal cancer

Fig. 1.2 Grade 2


dermatitis of neck after
radiotherapy for
nasopharyngeal cancer
with multiple node
involvement
4 1 Radiation Dermatitis

Fig. 1.3 Grade 3


dermatitis in
inframammary fold after
breast irradiation

Table 1.1 CTCAE v4.3 for dermatitis


Definition
Grade 1 Faint erythema or dry desquamation
Grade 2 Moderate to brisk erythema; patchy moist desquamation, mostly confined to skin
folds and creases; moderate edema
Grade 3 Moist desquamation in areas other than skin folds and creases; bleeding induced by
minor trauma or abrasion
Grade 4 Skin necrosis or ulceration of full-thickness dermis; spontaneous bleeding from
involved site

1.4 Scoring

The severity of radiation dermatitis is graded using several grading systems such as
Common Terminology Criteria for Adverse Event (NCI-CTCAE grading), Radiation
Therapy Oncology Group (RTOG) toxicity scoring system, and the Radiation-­
Induced Skin Reaction Assessment Scale (RISRAS).
Common Terminology Criteria for Adverse Event (NCI-CTCAE grading) is one of
the most commonly used, and NCI-CTCAE v4.3 has been shown in Table 1.1 [24].
RTOG scoring is quite similar to NCI-CTCAE grading.

1.5 Risk Factors

Treatment and patient-related factors may affect incidence and severity of


dermatitis.

1.5.1 Treatment-Related Factors Including

The higher total dose, higher dose per fraction, reduced overall treatment time,
beam energy (depend on type and quality of the beam), higher volume and surface
1.5 Risk Factors 5

area exposed, the junction between the electron and photon field, use of tissue
expander and bolus material [25–27], concurrent systemic therapy with radiation
(radiosensitizing drug such as paclitaxel, docetaxel, anthracyclines, dactinomycin,
methotrexate, 5-fluorouracil, hydroxyurea, bleomycin, and cetuximab) [6, 28] are
related to the development of radiation dermatitis.
Epidermal growth factor receptor (EGFR) is highly expressed in the epidermis,
particularly in the proliferative basal cell layers, and has a major role in the regula-
tion of multiple phases of epithelial biology, including cell cycle progression, dif-
ferentiation, cell movement, and cellular survival; it also has an essential impact on
the inflammatory reactions of the skin [29–31]. There is a reciprocal relationship
between radiation and epidermal growth factor receptor inhibitors’ (like cetuximab)
effects on skin.
More severe radiation dermatitis is seen in patients receiving cetuximab plus
radiation therapy than radiation therapy alone or even concurrent chemoradiother-
apy [5, 15]. There is a slightly longer duration of radiation dermatitis (11.1 versus
9.4 weeks) [32] and an earlier onset of radiation dermatitis (week 1 or 2 compared
with 3–5 weeks, respectively) [15], when cetuximab is added to radiation therapy.
Dermatitis usually reveals rapid recovery with no scarring following the end of
treatment of cetuximab plus radiation therapy [15]. A deferred cetuximab-induced
acne-like rash appears within irradiated fields in concurrent treatment of cetuximab
with radiation therapy (about 3–5 weeks after initiation of treatment versus
7–10 days, respectively). It seems that there is no significant relationship between
the severity of cetuximab-associated acne-like rash outside irradiated fields and the
severity of radiation dermatitis [32].

1.5.2 Patient-Related Factors Include

Demographic and behavioral factors, comorbid diseases, inherited disorders, and


site of radiation therapy can affect incidence and severity of radiation-induced der-
matitis [2, 33–41] (Table 1.2).

Table 1.2 Patient-related predisposing factors for radiation-induced dermatitis


Demographic and Site of radiation
behavioral factors Comorbid disease Inherited disorders therapy
Black race Actinic skin lesions Basal cell nevus Anterior neck
Obesity Seroma aspiration after disease Extremities
Female gender breast surgery Fanconi anemia Submammary
Advanced age Systemic lupus Bloom syndrome Skin folds
Breast implant erythematosus Xeroderma
Smoking Systemic sclerosis Pigmentosum
Poor nutrition Juvenile rheumatoid Ataxia-telangiectasia
arthritis
HIV infection
Diabetes mellitus
Hypertension
6 1 Radiation Dermatitis

Smoking seems to impair wound healing by cutaneous vasoconstriction [17].


The impact of increasing age on radiation dermatitis is related to decreased epi-
dermal turnover resulting in extended healing times. Coexisting diseases like hyper-
tension, diabetes, obesity, or malnutrition in older patients affect the severity and
resolution of radiation dermatitis [17].
It has been reported that aspiration of a seroma following breast surgery may
lead to damage to the lymphatic system, compromising wound healing and leading
to a severe skin reaction during radiation therapy [41].
Patients with systemic lupus erythematosus (SLE), juvenile rheumatoid arthritis
(JRA), and systemic sclerosis have significantly greater radiosensitivity and are at
greater risk for developing radiation side effects. The presence of this connective tis-
sue disease is considered as a relative contraindication to radiation therapy [42, 43].
Wound healing is complicated in patients with uncontrolled diabetes mellitus
due to poor macrophage function including phagocytic activity, prolonged inflam-
matory phase, and greater risk of infection. Diabetes’ effect on normal tissue reac-
tion during radiation therapy needs further study [17].
There are a few rare genetic mutations that may predispose patients to severe
radiation dermatitis, including mutations in the ataxia-telangiectasia gene. It has
been suggested that unanticipated severe radiation dermatitis may indicate the pres-
ence of undetected genetic abnormalities in the ATM gene (ataxia-telangiectasia
mutated) that can predispose patients to cutaneous complications [44, 45].
No studies have specifically investigated the effect of hemoglobin level on nor-
mal skin exposed to radiation therapy.

1.6 Diagnosis

Acute radiation dermatitis is a clinical diagnosis based on the finding of skin changes
that shows a sharp demarcation of the skin changes and their limitation to the irradi-
ated areas and the history of radiation therapy and its duration. A skin biopsy is
usually not necessary for the diagnosis of radiation dermatitis [13].
Two important differential diagnoses of acute radiation dermatitis are radiation
recall and cellulitis. Radiation recall is an acute inflammatory skin reaction limited
to the area that was previously irradiated and occurs following cytotoxic agents or
other drug administration. It occurs at least 7 days after radiation therapy and may
occur weeks to years after radiation therapy [46–48]. Cytotoxic drugs such as
docetaxel, doxorubicin, and paclitaxel have been associated with radiation recall.
Radiation recall manifestations include a rash, dry desquamation, erythema, ulcer-
ation, hemorrhage, or even necrosis. Radiation recall generally resolves within
1–2 weeks after drug discontinuation. Patient’s history of skin reaction following
radiation treatment and medications will help to reach an accurate diagnosis [49].
Cellulitis is a bacterial infection of skin and subcutaneous tissues. It is character-
ized by localized erythema, warmth, swelling, pain, tenderness, fevers, and even
purulent drainage. These clues can differentiate cellulitis from radiation dermatitis.
However, radiation dermatitis is a predisposing factor of secondary cellulitis, and it
1.7 Prevention 7

may be possible for both conditions to exist together. In this condition a culture
from the wound may confirm the diagnosis of cellulitis. Empiric antibiotic treat-
ment may also be helpful in these patients [49].
Eczema is a chronic, relapsing, inflammatory skin condition that is diagnosed by
clinical presentation of symmetrical, pruritic, dry skin that persists for <6 months.
Chronic eczema may exacerbate radiation dermatitis as both conditions are inflam-
matory in nature. Eczema may be distinguished from radiation dermatitis by its
symmetrical distribution. In contrast, radiation dermatitis will only appear within
radiation fields [49].
Recurrent or secondary tumors should be considered if atypical plaques and nod-
ules develop within the radiation field. Although irradiated skin may present some
difficulty in healing, a biopsy is useful in this clinical situation.
Atopic dermatitis [49], lichen planus [50], pemphigus [51, 52], and dermato-
phyte infection [53] rarely occur in the site of radiation therapy that clinical course,
lesion distribution, and laboratory studies are helpful to distinguish from other
diagnoses.

1.7 Prevention

1.7.1 General Measures

General recommendations for skin care in patients that are treated with radiation
therapy include [13, 54]:

Washing the irradiated area daily with water and mild soap (pH-neutral agents) or
normal saline then dry it and use a water-based moisturizer.
Wearing loose-fitting clothes.
Avoiding skin irritants such as perfumes and alcohol-based lotions and wax or other
depilatory creams.
Avoiding extremes of heat and cold.
Avoiding sun exposure and use of sunscreen with a minimum SPF 30.

Currently, application of topical steroids is the only prophylactic intervention


for radiation dermatitis protection suggested with a high level of evidence [55,
56]. Prophylactic application of topical steroids can reduce the severity of radia-
tion skin reaction because of their anti-inflammatory effects (i.e., reducing the
production of IL-1, IL-2, IL6 IFN-γ, TNF, and histamine and inhibiting leukocyte
migration) [14, 57].
Low- to medium-potency topical corticosteroids (groups 4–6) such as mometa-
sone furoate 0.1% or hydrocortisone 1% cream or methylprednisolone aceponate
cream 0.1% or beclomethasone dipropionate spray are applied to the treatment field
once or twice daily after each radiotherapy treatment [13, 58–61].
Treatment should be stopped if there is any exudate from the affected area [54]
or if a skin infection is suspected.
8 1 Radiation Dermatitis

Other protective options with primarily positive results that warrant further
research on their efficacy in preventing and managing acute radiation dermatitis
include:

1.7.2 Intensity-Modulated Radiation Therapy (IMRT)

There is some evidence suggesting that IMRT significantly reduces the severity and
duration of radiation dermatitis compared with standard conventional technique
[62–64].

1.7.3 Low-Level Laser Therapy

Low-level laser therapy is a form of phototherapy that induces the wound-healing


process [65]. It has generated great interest in preventing radiation side effects such
as dermatitis, xerostomia, or oral mucositis. The primary promising results of these
trials warrant further research on its efficacy in preventing and managing acute radi-
ation dermatitis [66].

1.7.4 Amifostine

Amifostine is a thiol that selectively protects normal cells from radiation damage by
scavenging oxygen-derived free radicals. It has been shown that the severity of radi-
ation dermatitis is significantly lower among patients receiving amifostine [67–69].
Further studies are needed to determine the impact and safety of amifostine as a
cytoprotective agent against acute radiation dermatitis.

1.7.5 Ascorbic Acid

Ascorbic acid has antioxidant capacity and anti-free-radical action. No benefit of its
topical application has been found for radiation dermatitis protection. However, oral
ascorbic acid in breast cancer radiation therapy with a volume lower than 500 mL
and in those that received a radiation dose between 107% and 110% of the pre-
scribed dose showed positive results [70, 71].

1.7.6 Oral Zinc

Zinc (Zn) as an antioxidant has been evaluated in normal tissue protection studies
against radiation injuries. There are primarily positive results for dermatitis that
need further evaluation [72, 73].
1.7 Prevention 9

1.7.7 Silver Leaf Dressing

Silver leaf dressing has antimicrobial activity and is another option proposed for
radiation-induced dermatitis prevention. There are positive results about its efficacy
[74–76] that should be confirmed in further studies.

1.7.8 MAS065D

MAS065D (Xclair) is a nonsteroidal water-in-oil cream that contains hyaluronic


acid, shea butter (emollient), glycyrrhetinic acid (licorice extract with anti-­
inflammatory properties), Vitis vinifera (antioxidant activity), and telmesteine
(anti-­elastase and anti-collagen activity in vitro) [77]. MAS065D seems to be
effective in the management of radiation dermatitis, but further studies are neces-
sary [78, 79].
Oral proteolytic enzymes (e.g., products containing papain, bromelain, trypsin,
and chymotrypsin) seem to be effective in protection against radiation dermatitis,
and further evaluation is recommended [72, 80].

1.7.9 Soft Dressing or Barrier Film

It seems that prophylactic use of friction protections with soft dressing or barrier
film in areas at risk such as the axilla, head and neck region, perineum, and skin
folds could be an effective intervention for reducing skin reaction severity [81].
Other options without well-designed trial support include:

1.7.10 Hyaluronic Acid

Hyaluronic acid cream appears to reduce inflammatory response and oxidative free-­
radical damage and have been evaluated in radiation therapy studies as a preventive
method for radiation dermatitis. Additional studies are required to clarify hyaluronic
acid role as a preventive option in radiation dermatitis [82, 83].

1.7.11 Trolamine

A topically applied salicylates analgesic, this agent has been shown to promote
the wound-healing process by recruitment of macrophages and enhancement of
granulation tissue formation, which has therapeutic applications in dermatology.
Current evidence does not support the use of trolamine for radiation dermatitis
and requires more research to clarify its efficacy in prevention of radiation derma-
titis [84–87].
10 1 Radiation Dermatitis

1.7.12 Topical Sucralfate

Sucralfate is a topical drug that has been applied for the treatment of several types
of epithelial wounds such as ulcers, inflammatory dermatitis, mucositis, and burn
wounds. Some studies have evaluated the efficacy of sucralfate cream for preven-
tion of radiation dermatitis with differing results. Current data do not support it to
prevent radiation dermatitis [87–90].

1.7.13 Theta Cream

It contains glucan, Hydroxyprolisilane, and matrixyl with the immune system-­


modulating effects and tissue regeneration properties [91]. Evidence from a limited
number of randomized trials does not support theta cream in radiation dermatitis
prevention [92].

1.7.14 Dexpanthenol (Provitamin B5)

A stable alcohol form of pantothenic acid, dexpanthenol is converted in tissues to


pantothenic acid, which is essential to normal epithelial function and enhances skin
regeneration and wound healing [93–95]. Current evidence does not support the use
of dexpanthenol for radiation dermatitis [92].

1.7.15 Calendula

Fabricated from a plant of the marigold family Calendula officinalis, Calendula has
antioxidant and anti-inflammatory properties. Studies evaluating the efficacy of
calendula in management and prevention of radiation dermatitis have mixed results
that warrant further investigation [96, 97].

1.7.16 Pentoxifylline

A useful drug in a variety of vaso-occlusive disorders, pentoxifylline has ant-­


inflammatory properties. The pentoxifylline effects on acute skin reactions need to
be determined in well-designed studies [98, 99].

1.7.17 Aloe Vera

An ancient herbal remedy and natural product, there are mixed results of its efficacy
in radiation dermatitis protection that warrants more research before its extensive
usage is recommended [100, 101].
1.8 Management 11

1.7.18 Deodorant

Using deodorant did not demonstrate any effect on preventing radiation dermatitis
development [102, 103].

1.7.19 Laughter Therapy

Laughter therapy may have a beneficial role in preventing radiation dermatitis. To


confirm laughter therapy effect on radiation dermatitis, well-designed randomized
studies with large sample sizes are required [104].

1.8 Management

Patients should be visited at least weekly during their radiation therapy course with
regard of the skin reaction at each visit. The management of radiation dermatitis is
guided by the severity of skin damage.

1.8.1 Grade 1 Dermatitis

For most patients, general skin care measures seem to be sufficient for ameliorating
patient’s symptoms. Patients should be educated on washing their skin gently with
a mild soap (pH-neutral agents) or normal saline as a supportive care for erythema-
tous skin. Washing reduces bacterial load and risk for infection.
Dry desquamation is managed by moisturizing the area with a thin layer of non-­
scented, hydrophilic, lanolin-free ointment or cream (e.g., Eucerin, Lubriderm) 2–4
times daily [49]. Topical moisturizers can act as a skin bolus and increase the radia-
tion skin dose, so patients should be informed not to apply these products shortly
before radiation treatment.
Pruritus or irritation can be controlled with mid-potency topical corticosteroids.
Although there is insufficient information to support using topical corticosteroids
(corticosteroids Group IV and V) in radiation-induced dermatitis, most practitioner
is prescribing steroid for it. Topical steroid which is usually prescribed in radiation-­
induced dermatitis is listed in Table 1.3 [105].

Table 1.3 Topical corticosteroids used in radiation dermatitis


Group IV Group V
Fluocinolone acetonide 0.01–0.2% Fluticasone propionate 0.05%
Hydrocortisone valerate 0.2% Desonide 0.05%
Hydrocortisone butyrate 0.1% Fluocinolone acetonide 0.025%
Flurandrenolide 0.05%
Triamcinolone acetonide 0.1%
Mometasone furoate 0.1%
12 1 Radiation Dermatitis

Antihistamines do not seem to be effective in reducing pruritus related to radia-


tion dermatitis [13].
Normal saline compress is applied up to four times daily and may help patients
to clean their skin better and ameliorate symptoms related to inflammation. Patients
are instructed to moisten gauze with warm or room-temperature saline solution and
apply moist gauze to the affected area for 10–15 min. The gauze is removed and the
wound is gently irrigated with normal saline and dried [54].

1.8.2 Grade 2–3 Dermatitis

In Grades 2 and 3 radiation dermatitis, in addition to general skin care measures, a


number of topical applications can be helpful to reduce symptoms.
Patients with moist desquamation are at increased risk of infection and should be
monitored for purulent drainage and fevers.
Moist desquamation typically is managed by wound dressings that provide the
ideal moist wound-healing environment allowing for faster reepithelization,
phagocytosis of bacteria, extracellular debris, and necrotic material, absorbing
wound secretions, diminishing pain, and protecting the wound from contamination
[6, 7, 106].
Dressings should be comfortable to increase patient compliance for prolonged
use. They should also be able to absorb large amounts of serous leakage from the
wound to prevent maceration of the surrounding healthy skin and must be removed
without disturbing granulation and new tissue.
Various forms of dressings are available in clinical practice with no sufficient
evidence to support particular types in the management of moist desquamation
[13, 75, 107, 108]. Simple dressings alone are not recommended, due to the pain
and trauma caused at dressing changes [41]. Hydrocolloids or hydrogels dressings
have a traditional interest in the use of moist desquamation. However, a recent
study that compared hydrogel versus dry dressing on healing time of moist desqua-
mation found that healing time was significantly prolonged in the hydrogel group
dressings without any improvement in patient comfort [109]. A new range of sili-
cone foam skin dressings (e.g., Mepilex Lite) is currently available commercially
with promising primary results [110].
Patients should be instructed to place normal saline compresses several times a
day, applying a dressing after cleaning the wound and changing dressings frequently
depending upon the severity of weeping.
When infection occurs, treatment should be done with topical and/or systemic
antibiotics. Silver-based dressings are antibacterial and have proven to be effective for
this purpose. Beta glucan or silver sulfadiazine cream may also be useful (but should
only be applied after radiation therapy and after cleaning the irradiated area; caution
use in patients with severe renal and hepatic impairment and patients with G6PD
deficiency) [111]. In patients treated with concomitant chemotherapy or at increased
risk of developing neutropenia, complete blood count should be checked. In patients
with neutropenia or signs of sepsis, blood cultures should be obtained [15].
References 13

Nonsteroidal anti-inflammatory drugs can often be effective for pain manage-


ment and can also relieve the discomfort associated with itching and swelling
around the skin reaction.
Treatment interruption may be needed in Grade 3 radiation dermatitis depending
on patient performance status, presence of sepsis or severe neutropenia, and the
radiation oncologist’s judgment [13].
Biologic preparations are investigated in some types of radiation dermatitis
including the use of calcineurin inhibitors (e.g., pimecrolimus cream or tacrolimus
ointment) for radiation dermatitis in patients receiving cetuximab [15] or topical
granulocyte-macrophage colony stimulating for vulvar radiation dermatitis [112].
The effects of foam dressing with human recombinant human epidermal growth
factor (rhEGF) [68] and granulocyte-colony stimulating factor (G-CSF) subcutane-
ously administered at the periphery of the wound [113] has been proposed in radia-
tion dermatitis with moist desquamation. Further studies are required to evaluate
optimal dosage, treatment scheduling, and confirming the mechanism of action of
these agents.
Lianbai liquid is a Chinese remedy that has been reported to be effective in the
treatment of Grade III acute radiation dermatitis and needs further evaluation [114].

1.8.3 Grade 4 Dermatitis

Grade 4 radiodermatitis is a rare complication. Surgical interventions are the treat-


ment backbone. This grade should be treated with discontinuation of radiation ther-
apy and surgical debridement, full-thickness skin graft, or myocutaneous or pedicle
flaps [13].

References
1. Salvo N, Barnes E, Draanen JV, Stacey E, Mitera G, Breen D, Giotis A, Czarnota G, Pang J,
Angelis CD (2010) Prophylaxis and management of acute radiation-induced skin reactions: a
systematic review of the literature. Curr Oncol 17(4):94–112
2. Brown KR, Rzucidlo E (2011) Acute and chronic radiation injury. J Vasc Surg
53(15):15S–21S
3. Pires AM, Segreto RA, Segreto HR (2008) RTOG criteria to evaluate acute skin reaction and
its risk factors in patients with breast cancer submitted to radiotherapy. Rev Lat Am
Enfermagem 16(5):844–849
4. KK F, Pajak TF, Trotti A et al (2000) A radiation therapy oncology group (RTOG) phase III
randomized study to compare hyperfractionation and two variants of accelerated fraction-
ation to standard fractionation radiotherapy for head and neck squamous cell carcinomas:
first report of RTOG 9003. Int J Radiat Oncol Biol Phys 48:7–16
5. Bonner JA, Harari PM, Giralt J et al (2006) Radiotherapy plus cetuximab for squamous-cell
carcinoma of the head and neck. N Engl J Med 354:567–578
6. Hymes SR, Strom EA, Fife C (2006) Radiation dermatitis: clinical presentation, pathophysi-
ology, and treatment 2006. J Am Acad Dermatol 54:28–46
7. Mendelsohn FA, Divino CM, Reis ED, Kerstein MD (2002) Wound care after radiation ther-
apy. Adv Skin Wound Care 15:216–224
14 1 Radiation Dermatitis

8. Malkinson FD, Keane JT (1981) Radiobiology of the skin: review of some effects on epider-
mis and hair. J Invest Dermatol 77:133–138
9. Hopewell JW (1990) The skin: its structure and response to ionizing radiation. Int J Radiat
Biol 57:751–773
10. Mettler FA, Upton AC (1995) Medical effects of ionizing radiation, 2nd edn. W.B. Saunders,
Philadelphia, pp 14–21
11. Hopewell JW, Nyman J, Turesson I (2003) Time factor for acute tissue reactions following
fractionated irradiation: a balance between repopulation and enhanced radiosensitivity. Int
J Radiat Biol 79:513–524
12. Muller K, Meineke V (2007) Radiation-induced alterations in cytokine production by skin
cells. Exp Hematol 35:96–104
13. Radiation dermatitis. Available from www.https://www.uptodate.com/contents/
radiation-dermatitis#H63768201
14. Meghrajani CF, Co HC, Ang-Tiu CM, Roa FC (2013) Topical corticosteroid therapy for the
prevention of acute radiation dermatitis. Expert Rev Clin Pharmacol 6(6):641–649
15. Bernier J, Russi EG, Homey B, Merlano M, Mesía R, Peyrade F, Budach W (2011)
Management of radiation dermatitis in patients receiving cetuximab and radiotherapy for
locally advanced squamous cell carcinoma of the head and neck: proposals for a revised grad-
ing system and consensus management guidelines. Ann Oncol 22(10):2191–2200
16. Madison KC (2003) Barrier function of the skin: “la raison d’etre” of the epidermis. J Invest
Dermatol 121:231–241
17. Porock D (1988) Predicting the severity of radiation skin reactions in women with breast
cancer. Edith Cowan University, Australia
18. Collen B, Mayer M (2006) Acute effects of radiation treatment: skin reactions. Can Vet
J 47(9):931–935
19. McQuestion M (2006) Evidence based skin care management in radiation therapy. Semin
Oncol Nurs 22:163–173
20. Frederick DM, Renato GP, Wolff K, Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell
DJ (2008) Fitzpatrick’s dermatology in general medicine. In: Radiobiology and radiation
effects, 7th edn. McGraw-Hill, New York, pp 858–862
21. Bray FN, Simmons BJ, Wolfson AH, Keyvan NK (2016) Acute and chronic cutaneous reac-
tions to ionizing radiation therapy. Dermatol Ther (Heidelb) 6(2):185–206
22. Mateus C, Thomas M (2014) Radiation dermatitis. Therapeutics in dermatology, 13 March
23. Lee JH, Kay CS, Maeng LS, Oh SJ, Lee AH, Lee DJ, Han CW, Cho SH (2009) The clinical
features and pathophysiology of acute radiation dermatitis in patients receiving tomotherapy.
Ann Dermatol 21(4):358–363
24. CTCAE v 4.03 can be downloaded without charge at http://evs.nci.nih.gov/ftp1/CTCAE/
CTCAE_4.03_2010-06-14_QuickReference_8.5x11.pdf. Accessed 6 Aug 2010
25. Hall EJ, Cox JD (2003) Physical and biological basis of radiation therapy. In: Cox JD, Ang
KK (eds) Radiation oncology, 8th edn. Mosby, St. Louis, pp 3–62
26. Fernando IN, Ford HT, Powles TJ, Ashley S, Glees JP, Torr M et al (1996) Factors affecting
acute skin toxicity in patients having breast irradiation after conservative surgery: a prospec-
tive study of treatment practice at the Royal Marsden Hospital. Clin Oncol (R Coll Radiol)
8:226–233
27. Emami B, Lyman J, Brow A, Coia L, Goitein M, Munzenrider JE et al (1991) Tolerance of
normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys 21:109–122
28. Wendt TG, Grabenbauer GG, Rodel CM et al (1998) Simultaneous radiochemotherapy ver-
sus radiotherapy alone in advanced head and neck cancer: a randomized multicenter study.
J Clin Oncol 16:1318–1132
29. Jost M, Kari C, Rodeck U (2000) The EGF receptor—an essential regulator of multiple epi-
dermal functions. Eur J Dermatol 10:505–510
30. Segaert S, Van Cutsem E (2005) Clinical signs, pathophysiology and management of skin
toxicity during therapy with epidermal growth factor receptor inhibitors. Ann Oncol
16:1425–1433
References 15

31. Pastore S, Mascia F, Mariani V, Girolomoni G (2008) The epidermal growth factor receptor
system in skin repair and inflammation. J Invest Dermatol 128:1365–1374
32. Bonner JA, Harari P, Giralt J, et al (2007) Duration of mucositis and dysphagia following
radiotherapy (± cetuximab) for locoregionally advanced head and neck cancer, p 97.
Multidisciplinary Head and Neck Cancer Symposium ASTRO, ASCO, and AHNS, January
18, Rancho Mirage, CA. Plenary Session 4
33. Safwat A, Bentzen SM, Turesson I, Hendry JH (2002) Deterministic rather than stochastic
factors explain most of the variation in the expression of skin telangiectasia after radiother-
apy. Int J Radiat Oncol Biol Phys 52:198–204
34. Porock D, Kristjanson L, Nikoletti S, Cameron F, Pedler P (1998) Predicting the severity of
radiation skin reactions in women with breast cancer. Oncol Nurs Forum 25:1019–1029
35. Twardella D, Popanda O, Helmbold I, Ebbeler R, Benner A, von Fournier D et al (2003)
Personal characteristics, therapy modalities and individual DNA repair capacity as predictive
factors of acute skin toxicity in an unselected cohort of breast cancer patients receiving radio-
therapy. Radiother Oncol 69:145–115
36. Ross JG, Hussey DH, Mayr NA, Davis CS (1993) Acute and late reactions to radiation ther-
apy in patients with collagen vascular diseases. Cancer 71:3744–3752
37. De Naeyer B, De Meerleer G, Braems S, Vakaet L, Huys J (1999) Collagen vascular diseases
and radiation therapy: a critical review. Int J Radiat Oncol Biol Phys 44:975–980
38. Morris MM, Powell SN (1997) Irradiation in the setting of collagen vascular disease: acute
and late complications. J Clin Oncol 15:2728–2735
39. Radiation dermatitis. DermNet NZ. Available from www.dermnetnz.org/reactions/radiation-­
dermatitis.htm
40. Ryan J, Bole C, Hickok J, Figueroa-Moseley C, Colman L, Khanna R, Pentland A, Morrow
G, Morrow G (2007) Post-treatment skin reactions reported by cancer patients differ by race,
not by treatment or expectations. Br J Cancer 97(1):14–21
41. Wells M, Macbride SH. Skin radiation reaction. Available from www.us.elsevierhealth.com
42. McCurdy D, Tai LQ, Frias S, Wang Z (1997) Delayed repair of DNA damage by ionizing
radiation in cells from patients with juvenile systemic lupus erythematosus and rheumatoid
arthritis. Radiat Res 147:48–54
43. Harris G, Cramp WA, Edwards JC, George AM, Sabovljev SA, Hart L et al (1985)
Radiosensitivity of peripheral blood lymphocytes in autoimmune disease. Int J Radiat Biol
Relat Stud Phys Chem Med 47:689–699
44. Swift M, Morrell D, Cromartie E, Chamberlin AR, Skolnick MH, Bishop DT (1986) The
incidence and gene frequency of ataxia-telangiectasia in the United States. Am J Hum Genet
39:573–583
45. Iannuzzi CM, Atencio DP, Green S, Stock RG, Rosenstein BS (2002) ATM mutations in
female breast cancer patients predict for an increase in radiation-induced late effects. Int
J Radiat Oncol Biol Phys 52:606–613
46. Camidge R, Price A (2001) Characterizing the phenomenon of radiation recall dermatitis.
Radiother Oncol 59:237–245
47. Bostrom A, Sjolin-Forsberg G, Wilking N, Bergh J (1999) Radiation recall—another call
with tamoxifen. Acta Oncol 38:955–959
48. Parry BR (1992) Radiation recall induced by tamoxifen. Lancet 340:49
49. Johns C (2012) Management of radiodermatitis. University of Pennsylvania School of
Nursing oncolink
50. Eichbaum M, Harms W, Bolz S, Schneeweiss A, Sohn C (2006) Generalized lichen ruber
planus—induced by radiotherapy of the breast? Onkologie 29:521–523. https://www.upto-
date.com/contents/radiation-dermatitis#H2540924292
51. Low GJ, Keeling JH (1990) Ionizing radiation-induced pemphigus. Case presentations and
literature review. Arch Dermatol 126:1319–1323
52. Badri T, Hammami H, Lachkham A, Benmously-Mlika R, Mokhtar I, Fenniche S (2011)
Radiotherapy-induced pemphigus vulgaris with autoantibodies targeting a 110 kDa epider-
mal antigen. Int J Dermatol 50(12):1475–1479
16 1 Radiation Dermatitis

53. Casamiquela KM, Cohen PR (2012) Radiation port dermatophytosis: tinea corporis occur-
ring at the site of irradiated skin. Dermatol Online J 18:5
54. Symptom management guidelines: radiation dermatitis.bc cancer agency. Available from
www.bccancerr.bc.ca
55. Chan R, Webster J, Chung B, Marquart L, Ahmed M, Garantziotis S (2014) Prevention and
treatment of acute radiation-induced skin reactions: a systematic review and meta-analysis of
randomized controlled trials. BMC Cancer 14:53
56. Bolderston A, Lloyd NS, Wong RK et al (2006) The prevention and management of acute
skin reactions related to radiation therapy: a systematic review and practice guideline.
Support Care Cancer 14:802–817
57. Kragballe K (1989) Topical corticosteroids: mechanisms of action. Acta Derm Venereol
Suppl (Stockh) 151:7–10. Discussion 47–52
58. Boström A, Lindman H, Swartling C, Berne B, Bergh J (2001) Potent corticosteroid cream
(mometasone furoate) significantly reduces acute radiation dermatitis: results from a double-­
blind, randomized study. Radiother Oncol 59:257–265
59. Schmuth M, Wimmer MA, Hofer S et al (2002) Topical corticosteroid therapy for acute
radiation dermatitis: a prospective, randomized, double-blind study. Br J Dermatol
146:983–991
60. Shukla PN, Gairola M, Mohanti BK, Rath GK (2006) Prophylactic beclomethasone spray to
the skin during postoperative radiotherapy of carcinoma breast: a prospective randomized
study. Indian J Cancer 43(4):180
61. Omidvari S, Saboori H, Mohammadianpanah M et al (2007) Topical betamethasone for pre-
vention of radiation dermatitis. Indian J Dermatol Venereol Leprol 73:209–215
62. Pignol JP, Olivotto I, Rakovitch E, Gardner S, Sixel K, Beckham W, Vu TT, Truong P,
Ackerman I, Paszat L (2008) A multicenter randomized trial of breast intensity-modulated
radiation therapy to reduce acute radiation dermatitis. J Clin Oncol 26(13):2085–2092
63. Freedman GM, Anderson PR, Li J, Eisenberg DF, Hanlon AL, Wang L, Nicolaou N (2006)
Intensity modulated radiation therapy (IMRT) decreases acute skin toxicity for women
receiving radiation for breast cancer. Am J Clin Oncol 29(1):66–70
64. Freedman G, Li T, Nicolaou N, Chen Y, Ma Ch C-M, Anderson P (2009) Breast IMRT
reduces time spent with acute dermatitis for women of all breast sizes during radiation. Int
J Radiat Oncol Biol Phys 74(3):689–694
65. Hawkins D, Abrahamse H (2007) Effect of multiple exposures of low-level laser therapy on
the cellular responses of wounded human skin fibroblasts. Photomed Laser Surg
24(6):705–714
66. Censabella S, Robijns J, Claes S, Bulens P, Mebis J (2014) Low level laser therapy for the
management of radiation dermatitis: preliminary results of a pilot study in breast cancer
patients. Ann Oncol 25(4):iv534
67. Censabella S, Claes S, Robijns J, Bulens P, Mebis J (2016) Photobiomodulation for the man-
agement of radiation dermatitis: the DERMIS trial, a pilot study of MLS(®) laser therapy in
breast cancer patients. Support Care Cancer 24(9):3925–3933
68. Dunst J, Semlin S, Pigorsch S, Müller AC, Reese T (2000) Intermittent use of amifostine
during postoperative radiochemotherapy and acute toxicity in rectal cancer patients.
Strahlenther Onkol 176:416–421
69. Koukourakis MI, Kyrias G, Kakolyris S et al (2000) Subcutaneous administration of amifos-
tine during fractionated radiotherapy: A randomized phase II study. J Clin Oncol
18:2226–2233
70. Halperin EC, Gaspar L, George S, Darr D, Pinnell S (1993 Jun 15) A double-blind, random-
ized, prospective trial to evaluate topical vitamin C solution for the prevention of radiation
dermatitis. CNS Cancer Consortium. Int J Radiat Oncol Biol Phys 26(3):413–416
71. Di Franco R, Calvanese M, Murino P, Manzo R, Guida C, Di Gennaro D, Anania C, Ravo
V (2012) Skin toxicity from external beam radiation therapy in breast cancer patients:
protective effects of resveratrol, lycopene, vitamin C and anthocyanin (Ixor®). Radiat
Oncol 7:12
References 17

72. Feight D, Baney T, Bruce S (2011) Evidence-based interventions for radiation dermatitis.
Clin J Oncol Nurs 15(5):481–492
73. Lin LC, Que J, Lin LK, Lin FC (2006) Zinc supplementation to improve mucositis and der-
matitis in patients after radiotherapy for head-and-neck cancers: a double-blind, randomized
study. Int J Radiat Oncol Biol Phys 65:745–750
74. Aquino-Parsons C, Lomas S, Smith K, Hayes J, Lew S, Bates AT, Macdonald AG (2010)
Phase III study of silver leaf nylon dressing vs standard care for reduction of inframammary
moist desquamation in patients undergoing adjuvant whole breast radiation therapy. J Med
Imag Rad Sci 41(4):215–221
75. Niazi TM, Vuong T, Azoulay L, Marijnen C, Bujko K, Nasr E et al (2012) Silver clear nylon
dressing is effective in preventing radiation-induced dermatitis in patients with lower gastro-
intestinal cancer: results from a phase III study. Int J Radiat Oncol Biol Phys 84:e305–e310
76. Vuong T, Franco E, Lehnert S, Lambert C, Portelance L, Nasr E et al (2004) Silver leaf nylon
dressing to prevent radiation dermatitis in patients undergoing chemotherapy and external
beam radiotherapy to the perineum. Int J Radiat Oncol Biol Phys 59(3):809–814
77. MASO65D (Xclair®) | ONS. Available from https://www.ons.org/intervention/
maso65d-xclair%C2%AE
78. Leonardi MC, Gariboldi S, Ivaldi GB et al (2008) A double-blind randomized, vehicle-­
controlled clinical study to evaluate the efficacy of MAS065D in limiting the effects of radia-
tion on the skin: interim analysis. Eur J Dermatol 18:317–321
79. Primavera G, Carrera M, Berardesca E, Pinnaró P, Messina M, Arcangeli G (2006) A double-­
blind, vehicle controlled clinical study to evaluate the efficacy of MAS065D (Xclair), a hyal-
uronic acid–based formulation, in the management of radiation-induced dermatitis. Cutan
Ocul Toxicol 25:165–171
80. Desser L, Holomanova D, Zavadova E, Pavelka K, Mohr T, Herbacek I (2001) Oral therapy
with proteolytic enzymes decreases excessive TGF-beta levels in human blood. Cancer
Chemother Pharmacol 47:S10–S15
81. Herst M (2014) Protecting the radiation-damaged skin from friction: a mini review. J Med
Radiat Sci 61(2):119–125
82. Pinnix C, Perkins GH, Strom EA, Tereffe W, Woodward W, JL O, Arriaga L, Munsell MF,
Kelly P, Hoffman KE, Smith BD (2012) Topical hyaluronic acid vs. standard of care for the
prevention of radiation dermatitis after adjuvant radiotherapy for breast cancer: single-blind
randomized phase III clinical trial. Int J Radiat Oncol Biol Phys 83(4):1089–1094
83. Liguori V, Guillemin C, Pesce GF et al (1997) Double-blind, randomized clinical study com-
paring hyaluronic acid cream to placebo in patients treated with radiotherapy. Radiother
Oncol 42:155–161
84. Abbas H, Bensadoun RJ (2012) Trolamine emulsion for the prevention of radiation dermatitis
in patients with squamous cell carcinoma of the head and neck. Support Care Cancer
20(1):185–190
85. Elliott EA, Wright JR, Swann RS, Nguyen-Tân F, Takita C, Bucci MK, Garden AS, Kim H,
Hug EB, Ryu J, Greenberg M, Saxton JP, Ang K, Berk L (2006) Phase III Trial of an emulsion
containing trolamine for the prevention of radiation dermatitis in patients with advanced
squamous cell carcinoma of the head and neck: results of Radiation Therapy Oncology Group
Trial 99-13. J Clin Oncol 24(13):2092–2097
86. Pommier P, Gomez F, Sunyach MP, D’Hombres A, Carrie C, Montbarbon X (2004) Phase III
randomized trial of Calendula officinalis compared with trolamine for the prevention of acute
dermatitis during irradiation for breast cancer. J Clin Oncol 22(8):1447–1453
87. Del Rosso JQ, Bikowski J (2008) Trolamine-containing topical emulsion: clinical applica-
tions in dermatology. Cutis 81(3):209–214
88. Wells M, Macmillan M, Raab G, MacBride S, Bell N, MacKinnon K et al (2004) Does aque-
ous or sucralfate cream affect the severity of erythematous radiation skin reactions? A ran-
domised controlled trial. Radiother Oncol 73:153–162
89. De Rauglaudre G, Courdi A, Delaby-Chagrin F, d’Hombres A, Hannoun-Levi JM, Moureau-­
Zabotto L, Richard-Tallet A, Rouah Y, Salem N, Thomas O, Nocera T, Mery S, Merial-Kieny C
18 1 Radiation Dermatitis

(2008) Tolerance of the association sucralfate/Cu-Zn salts in radiation dermatitis. Ann Dermatol
Venereol 1:11–15
90. Delaney G, Fisher R, Hook C, Barton M (1997) Sucralfate cream in the management of moist
desquamation during radiotherapy. Australas Radiol 41:270–275
91. Röper B, Kaisig D, Auer F, Mergen E, Molls M (2004) Thêta-Cream versus Bepanthol lotion
in breast cancer patients under radiotherapy. A new prophylactic agent in skin care?
Strahlenther Onkol 180(5):315–322
92. Wong RK, Bensadoun RJ, Boers-Doets CB et al (2013) Clinical practice guidelines for the
prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity
Study Group. Support Care Cancer 21(10):2933–2948
93. Løkkevik E, Skovlund E, Reitan JB, Hannisdal E, Tanum G (1996) Skin treatment with
bepanthen cream versus no cream during radiotherapy—a randomized controlled trial. Acta
Oncol 35(8):1021–1026
94. Censabella S, Claes S, Orlandini M, Braekers R, Thijs H, Bulens P (2014) Retrospective
study of radiotherapy-induced skin reactions in breast cancer patients: reduced incidence of
moist desquamation with a hydroactive colloid gel versus dexpanthenol. Eur J Oncol Nurs
18(5):499–504
95. Proksch E, Nissen HP (2002) Dexpanthenol enhances skin barrier repair and reduces inflam-
mation after sodium lauryl sulphate-induced irritation. J Dermatolog Treat 13(4):173–178
96. Fotouhi M, Samee F, Amoozegar Hashemi F, Hadad P, Meysami AP (2007) Topical calendula
and betamethasone valerate in the prevention of acute radiation dermatitis: a randomized
prospective trial. Tehran Univ Med J 65(3):23–29
97. Kodiyan J, Amber KT (2015) A review of the use of topical calendula in the prevention and
treatment of radiotherapy-induced skin reactions. Antioxidants 4:293–303
98. Aygenc E, Celikkanat S, Kaymakci M, Aksaray F, Ozdem C (2004) Prophylactic effect of
pentoxifylline on radiotherapy complications: a clinical study. Otolaryngol Head Neck Surg
130:351–356
99. Javan Chan R, Webster J, Chung B, Marquart L, Ahmed M, Garantziotis S (2014) Prevention
and treatment of acute radiation-induced skin reactions: a systematic review and meta-­
analysis of randomized controlled trials. BMC Cancer 14:53
100. Heggie S, Bryant GP, Tripcony L et al (2002) A phase III study on the efficacy of topical Aloe
vera gel on irradiated breast tissue. Cancer Nurs 25:442–451
101. Olsen DL, Raub W Jr, Bradley C et al (2001) The effect of Aloe vera gel/mild soap versus
mild soap alone in preventing skin reactions in patients undergoing radiation therapy. Oncol
Nurs Forum 28:543–547
102. Bennett C (2009) An investigation into the use of a non-metallic deodorant during radio-
therapy treatment: a randomised controlled trial. J Radiother Pract 8(1):3–9
103. Gee A, Churn M, Errington RD (2000) A randomised controlled trial to test a non-metallic
deodorant used during a course of radiotherapy. J Radiother Pract 1:205–212
104. Kong M, Shin SH, Lee EM, Yun EK (2014) The effect of laughter therapy on radiation der-
matitis in patients with breast cancer: a single-blind prospective pilot study. Onco Targets
Ther 7:2053–2059
105. Topical steroid. Available from https://en.wikipedia.org/wiki/Topical_steroid
106. Gray M (2004) Preventing and managing perineal dermatitis: a shared goal for wound and
continence care. J Wound Ostomy Continence Nurs 31(1):2–12
107. Gollins S, Gaffney C, Slade S, Swindell R (2008) RCT on gentian violet versus a hydrogel
dressing for radiotherapy-induced moist skin desquamation. J Wound Care 17(6):268–270,
272, 274–275
108. Mak SS, Zee CY, Molassiotis A, Chan SJ, Leung SF, Mo KF et al (2005) A comparison of
wound treatments in nasopharyngeal cancer patients receiving radiation therapy. Cancer Nurs
28(6):436–445
109. Macmillan MS, Wells M, MacBride S, Raab GM, Munro A, MacDougall H (2007)
Randomized comparison of dry dressings versus hydrogel in management of radiation-­
induced moist desquamation. Int J Radiat Oncol Biol Phys 68(3):864–872
References 19

110. Diggelmann KV, Zytkovicz AE, Tuaine JM, Bennett NC, Kelly LE, Herst PM (2010) Mepilex
Lite dressings for the management of radiation-induced erythema: a systematic inpatient con-
trolled clinical trial. Br J Radiol 83(995):971–978
111. Lacouture M, Anadkat M, Bensadoun R, Bryce J, Chan A, Epstein J, Eaby-Sandy B, Murphy
B (2011) Clinical practice guidelines for the prevention and treatment of EGFR inhibitor-­
associated dermatologic toxicities. Support Care Cancer 19(8):1079–1095
112. Kouvaris J, Kouloulias V, Plataniotis G, Kokakis J, Vlahos L (2001) Topical granulocyte-­
macrophage colony-stimulating factor for radiation dermatitis of the vulva. Br J Dermatol
144(3):646–647
113. Viswanath L, Bindhu J, Krishnamurthy B, Suresh K (2012) Granulocyte-colony stimulating
factor (G-CSF) accelerates healing of radiation induced moist desquamation of the skin. Klin
Onkol 25(3):199–205
114. Ma H, Zhang X, Bai M, Wang X (2007) Clinical effects of lianbai liquid in prevention and
treatment of dermal injury caused by radiotherapy. J Tradit Chin Med 27(3):193–196
115. Lee J, Lee SW, Hong JP, Shon MW, Ryu SH, Ahn SD (2016) Foam dressing with epidermal
growth factor for severe radiation dermatitis in head and neck cancer patients. Int Wound
J 13(3):390–393
Hair Loss
2

Whenever the skin is irradiated, hair loss may occur as a side effect. Because hair
loss of scalp induced by radiation of brain or head and neck cancer has a significant
importance due to its effect on appearance (Fig. 2.1), we focus on scalp hair loss in
this chapter.
Hair loss may be a temporary or permanent radiation side effect depending
on radiation dose [1]. Temporary hair loss is typically observed in all patients
undergoing whole-brain radiation therapy [2, 3]. Temporary hair loss with some
areas of permanent alopecia is one of the common complications (more than 10
out of 100), in patients with primary brain tumors treated with cranial radiation
therapy [4]. Permanent hair loss occurs in 50% of patients with the follicle dose
of 43.0 Gy [5].

Fig. 2.1 Hair loss at


posterior scalp after
radiation therapy for
sinonasal tumor that
exposed to exit dose of
antro-posterior radiation
beam

© Springer International Publishing AG 2017 21


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_2
22 2 Hair Loss

2.1 Mechanism

Radiation-induced hair loss is due to high susceptibility of anagen follicles to


radiation.
Hair follicles are located approximately 3–5 mm below the scalp [6]. Hair growth
has a cyclical process [7]. The three phases during the hair life cycle are anagen
(active phase), catagen (transitional phase), and telogen (resting phase). Most hairs
are normally in the anagen phase. During this phase, the bulb matrix cells are rap-
idly proliferating with high mitotic activity [8–10]. Any acute damage to actively
dividing matrix cells of anagen follicles leads to abrupt loss of hairs called anagen
effluvium. Chemotherapy and radiation therapy are common causes of anagen
effluvium [8, 9, 11, 12].
Ionizing radiation primarily targets DNA and leads to double-strand DNA breaks
[13, 14]. Radiation-induced DNA damage of hair follicular matrix cells causes apop-
tosis with increase in tumor-suppressor protein p53 levels in the matrix cells [15, 16].

2.2 Timing

Hair shedding may start as soon as 1–3 weeks after the first dose of radiation [17].
The hair follicle resumes normal cycling within a few weeks of treatment cessation
[9] and usually resolves within 2–3 months after completion of radiation therapy [5].
Regrowth of hair may be sparser with different thickness or texture after treatment.

2.2.1 Risk Factors

Radiation dose is the most significant factor-related radiation-induced alopecia.


Temporary alopecia can be detected after doses of about 2 Gy [18]. Permanent alo-
pecia has been reported in a range of 0–80% (median risk 5%) of patients with dose
of 36 Gy (2 Gy/fraction, 5 d/wk) and of 5–100% (median 15%) of patients with a
dose of 45 Gy. Permanent alopecia occurs with a 7 Gy single-fraction dose [5].
Lower-beam energies, use of multiple overlapping beams, or use of fixation mate-
rials with high thicknesses can increase the risk of radiation-induced alopecia [5].
Previous or concomitant chemotherapy (alkylating agents most commonly
reported) increase the risk of radiation-induced alopecia [5, 19].
Patients with personal history of alopecia may have a trend of developing perma-
nent alopecia. Patient’s age, family history of baldness, gender, tobacco use, and
diabetes have not reported to correlate with alopecia [5].

2.2.2 Symptoms

Radiation therapy will generally cause alopecia limited to the treated area but may
also occur in the adjacent area due to beam penumbra or at existing areas of the
radiation beam.
2.4 Prevention 23

Table 2.1 CTCAE hair loss scoring


Definition
Score 1 Hair loss of <50% of normal for that individual that is not obvious from distance but
only on close inspection; a different hairstyle may be required to cover the hair loss,
but it does not require a wig or hairpiece to camouflage
Score 2 Hair loss of ≥50% normal for that individual that is readily apparent to others; a wig
or hairpiece is necessary if the patient desires to completely camouflage the hair loss;
associated with psychosocial impact

After hair loss, scalp is sensitive to radiation damage and radiation dermatitis
may occur (see Chap. 1).

2.3 Scoring

Common Terminology Criteria for Adverse Events (CTCAE) v4.0 has defined two
scores for hair loss of scalp (Table 2.1) [20].

2.4 Prevention

It has been shown that patients affected by hair loss have a poorer quality of life,
lower self-esteem, and heightened self-consciousness [21, 22]. Novel strategies to
decrease radiation-induced alopecia have been proposed. New radiation delivery
techniques such as intensity-modulated radiation therapy (IMRT) or volumetrically
modulated arc therapy (VMAT) for whole-brain radiation therapy (WBRT) are
being evaluated to reduce the dose to the hair follicles [3, 23–25]. Primary results of
these techniques are promising, which warrant further investigation.
Tempol, formally 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl, is a
membrane-­permeable radical scavenger. Both the preclinical and clinical studies
showed that topical application of tempol may be effective at preventing radiation-­
induced alopecia [26–28]. Larger studies are needed to evaluate its efficacy and
safety in clinical practice.
Antioxidant compounds such as glutathione, lipoic acid, and antioxidant vitamins
A, C, and E could act as natural radioprotectors and reduce the toxicity of radiation
therapy. But possible tumor-protective effects of these nonselective free-­radical scav-
engers are matters of great concern. Topical application of these antioxidants has been
tested to reduce systemic absorption and subsequent tumor protection, but this does
not seem to be able to eliminate the tumor-protective effect. Additional studies need
to determine the safety of using these agents during radiation therapy [29].
Base on preclinical studies, systemic and topical application of prostaglandin E2,
systemic administration of vitamin D3, topically applied vasoconstrictors, subcuta-
neously applied keratinocyte growth factor, Panax ginseng administration, and caf-
feine may protect hair follicles from radiation toxicity [30–34]. Further clinical
trials should be conducted to prove the preventive effect of these agents on radiation-­
induced alopecia.
24 2 Hair Loss

2.5 Management

General recommendations during treatment may be helpful for the patient to mini-
mize scalp reaction including avoiding excessively hair combing, hair color and hair
styling products, hair dryers, using baby shampoo or other mild shampoo and hair
conditioner without any perfumes, and cutting hair to medium to short lengths [35].
Reconstructive surgery options can be considered in selective patients with post-
radiation therapy and permanent alopecia based on alopecia area, size, patient’s age,
and prognosis [36].
Botulinum toxin type A (BTXA) is used successfully to treat radiation-induced
alopecia in a case report, which warrants more studies to identify the mechanisms
and efficacy of BTXA [37].
There are no reports about other hair loss treatments such as minoxidil or low-­
level laser therapy in the setting of radiation-induced hair loss.

References
1. Ballonoff A. Complication of cranial irradiation. Available from http://www.uptodate.com/
contents/acute-complications-of-cranial-irradiation#H7
2. Gerrard GE, Prestwich RJ, Edwards A, Russon LJ, Richards F, Johnston CF, Kwok-Williams MC
(2003) Investigating the palliative efficacy of whole-brain radiotherapy for patients with multiple-
brain metastases and poor prognostic features. Clin Oncol (R Coll Radiol) 15(7):422–428
3. De Puysseleyr A, Van De Velde J, Speleers B, Vercauteren T, Goedgebeur A, Van Hoof T,
Boterberg T, De Neve W, De Wagter C, Ost P (2014) Hair-sparing whole brain radiotherapy
with volumetric arc therapy in patients treated for brain metastases: dosimetric and clinical
results of a phase II trial. Radiat Oncol 9:170
4. Side effects of brain irradiation—about cancer. Available from www.aboutcancer.com/brain_
side_effects.htm
5. Lawenda BD, Gagne HM, Gierga DP, Niemierko A, Wong WM, Tarbell NJ, Chen GT,
Hochberg FH, Loeffler JS (2004) Permanent alopecia after cranial irradiation: dose—response
relationship. Int J Radiat Oncol Biol Phys 60:879–887
6. de Viragh PA, Meuli M (1995) Human scalp hair follicle development from birth to adulthood:
statistical study with special regard to putative stem cells in the bulge and proliferating cells in
the matrix. Arch Dermatol Res 287:279–284
7. Philpott MP, Kealey T (2000) Cyclical changes in rat vibrissa follicles maintained in vitro.
J Invest Dermatol 115(6):1152–1155
8. Schwartz R. Anagen effluvium. Available from http://emedicine.medscape.com/
article/1073488-overview#a6
9. Kanwar AJ, Narang T (2013) Anagen effluvium. Indian J Dermatol Venereol Leprol
79(5):604–612
10. Schneider MR, Schmidt-Ullrich R, Paus R (2009) The hair follicle as a dynamic miniorgan.
Curr Biol 19(3):R132–R142
11. Ali SY, Singh G (2010) Radiation-induced Alopecia. Int J Trichology 2(2):118–119
12. Wen CS, Lin SM, Chen JC, Wang YH, Tseng SH (2003) Radiation-induced temporary alope-
cia after embolization of cerebral arteriovenous malformations. Clin Neurosurg 105:215–217
13. Hutchinson F (1966) The molecular basis for radiation effects on cells. Cancer Res
26:2045–2052
14. Fei P, El-Deiry WS (2003) P53 and radiation responses. Oncogene 22:5774–5783
15. Song S, Lambert P (1999) Different responses of epidermal and hair follicular cells to radia-
tion correlate with distinct patterns of p53 and p21 induction. Am J Pathol 155(4):1121–1127
16. Poeggeler B, Bodó E, Nadrowitz R, Dunst J, Paus R (2010) Simple assay for the study of
human hair follicle damage induced by ionizing irradiation. Exp Dermatol 19(8):e306–e309
References 25

17. Verma S, Srinivas C, Thomas M (2014) Radiation-induced temporary alopecia after emboliza-
tion of cerebral aneurysm. Indian J Dermatol 59(6):633
18. Potten CS, Burt PA, Roberts SA, Deshpande NA, Williams PC, Ramsden J (1996) Changes in
the cellularity of the cortex of human hairs as an indicator of radiation exposure. Radiat
Environ Biophys 35(2):121–125
19. Min C, Paganetti H, Winey B, Adams J, MacDonald S, Tarbell N, Yock T (2014) Evaluation
of permanent alopecia in pediatric medulloblastoma patients treated with proton radiation.
Radiat Oncol 9:220
20. National Cancer Institute (NCI), Common Terminology Criteria for Adverse Events (CTCAE)
Version 4.0.http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-­14_QuickReference_5x7.
pdf
21. Williamson D, Gonzalez M, Finlay AY (2001) The effect of hair loss on quality of life. J Eur
Acad Dermatol Venereol 15:137–139
22. Steinmann D, Paelecke-Habermann Y, Geinitz H, Aschoff R, Bayerl A, Bolling T, Bosch E,
Bruns F, Eichenseder-Seiss U, Gerstein J, Gharbi N, Hagg J, Hipp M, Kleff I, Müller A,
Schleicher U, Sehlen S, Theodorou M, Wypior HJ, Zehentmayr F, van Oorschot B, Vordermark
D (2012) Prospective evaluation of quality of life effects in patients undergoing palliative
radiotherapy for brain metastases. BMC Cancer 12:283
23. Mancini BR, Kim LH, Shaitelman SF, Yan D, Kestin LL, Grills IS (2010) Intensity modulated
or volumetric modulated radiation therapy (IMRT or VMAT) to reduce alopecia, xerostomia,
and otitis after whole brain radiation therapy for brain metastases: a planning analysis. Int
J Radiat Oncol Biol Phys 78:S840
24. Roberge D, Parker W, Niazi TM, Olivares M (2005) Treating the contents and not the con-
tainer: dosimetric study of hair-sparing whole brain intensity modulated radiation therapy.
Technol Cancer Res Treat 4:567–570
25. Weber DC, Caparrotti F, Laouiti M, Malek K (2011) Simultaneous in-field boost for patients
with 1 to 4 brain metastasis/es treated with volumetric modulated arc therapy: a prospective
study on quality-of-life. Radiat Oncol 6:79
26. Cuscela D, Coffin D, Lupton GP et al (1996) Protection from radiation-induced alopecia with
topical application of nitroxides: fractionated studies. Cancer J Sci Am 2:273–278
27. Goffman T, Cuscela D, Glass J et al (1992) Topical application of nitroxide protects radiation-­
induced alopecia in guinea pigs. Int J Radiat Oncol Biol Phys 22:803–806
28. Metz JM, Smith D, Mick R et al (2004) A phase I study of topical tempol for the prevention of
alopecia induced by whole brain radiotherapy. Clin Cancer Res 10:6411–6417
29. Citrin D, Cotrim AP, Hyodo F, Baum BJ, Krishna MC, Mitchell JB (2010) Radioprotectors and
mitigators of radiation-induced normal tissue injury. Oncologist 15:360–371
30. Baltalarli B, Bir F, Demirkan N, Abban G (2006) The preventive effect of vitamin D3 on
radiation-induced hair toxicity in a rat model. Life Sci 78:1646–1651
31. Hanson WR, Pelka AE, Nelson AK, Malkinson FD (1992) Subcutaneous or topical adminis-
tration of 16, 16 dimethyl prostaglandin E2 protects from radiation-induced alopecia in mice.
Int J Radiat Oncol Biol Phys 23:333–337
32. Hebbar SA, Mitra AK, George KC et al (2002) Caffeine ameliorates radiation-induced skin
reactions in mice but does not influence tumour radiation response. J Radiol Prot 22:63–69
33. Kim SH, Jeong KS, Ryu SY et al (1998) Panax ginseng prevents apoptosis in hair follicles and
accelerates recovery of hair medullary cells in irradiated mice. In Vivo 12:219–222
34. Booth C, Potten CS (2000) Keratinocyte growth factor increases hair follicle survival follow-
ing cytotoxic insult. J Invest Dermatol 114:667–673
35. Radiation therapy side effects: hair loss—Cleveland Clinic. Available from https://
my.clevelandclinic.org/health/articles/radiation-therapy-sideeffects-hair-loss
36. Rannan-Eliya YF, Rannan-Eliya S, Graham K, Pizer B, McDowell HP (2007) Surgical inter-
ventions for the treatment of radiation-induced alopecia in pediatric practice. Pediatr Blood
Cancer 49:731–736
37. Hyun MY, Kim BJ, Lee C, Kim JW (2014) Radiation-induced alopecia treated with Botulinum
toxin type a injection. Plast Reconstr Surg Glob Open 2(10):e226
Radiation Brain Injury
3

Cranial irradiation has an important role in the treatment of brain tumors either
with curative intent or for palliation. Cranial irradiation has a unique application
in prevention of distant metastasis to brain parenchyma, and prophylactic cranial
irradiation may be carried out in selected patients at high risk of neoplastic cranial
involvement. Brain is also an organ at risk during radiotherapy of tumors which
are located in base of skull and in some head and neck cancers. Cranial irradiation
in any condition can cause brain injuries that are classified into three groups based
on the timing of their occurrence after radiation exposure: acute (during radiation
or up to 6 weeks after radiation), early delayed or subacute (up to 6 months after
radiation), and late delayed (6 months or more after the completion of radiation).
However in more trials, brain injury simply is defined as acute (within 90 days of
the commencement of therapy) and late reactions (after 90 days of the commence-
ment of therapy) [1]. Reports indicate that patients that undergo standard fraction-
ated cranial irradiation and stereotactic radiosurgery may have acute brain
reactions in the range of 2–40% [1–12]. However, up to 50% of patients with high
doses per fraction (more than 3 Gy) to a large part of the brain may develop acute
encephalopathy [3, 13].
Early-delayed reactions may present with worsening of primary symptoms or
radiation somnolence syndrome (SS), which has been observed mainly after pro-
phylactic radiation therapy for leukemia in children, with a mean incidence of 50%
(in the range of 10–79% of these patients) [14–20].
It is important to differentiate brain tumor progression from radiation-induced
acute injury. Pseudoprogression is defined as radiologic abnormality in postirradia-
tion imaging because of radiation-induced injury that mimics tumor recurrence.
Pseudoprogression occurs a few weeks up to 3–6 months after radiation therapy
that is coincidental with early-delayed radiation reaction. The majority of patients
with pseudoprogression are asymptomatic, but it can present with worsening of
primary symptoms, transient cognitive decline, or somnolence syndrome [21].
Pseudoprogression incidence varies from 9% in patients treated with radiation

© Springer International Publishing AG 2017 27


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_3
28 3 Radiation Brain Injury

therapy alone to 32% in patients undergoing chemoradiotherapy [22, 23].


Somnolence syndrome may develop in adults as a subacute reaction [24, 25] but
have not been uniformly investigated.

3.1 Mechanism

Cranial irradiation causes vascular and cellular injuries. Endothelial cell damage,
vasodilation, increased in blood-brain barrier permeability, cerebral edema, and
consequent rise in intracranial pressure occur in early phase [3]. Oligodendrocytes
are extremely sensitive to radiation. Subacute reactions are thought to be a result of
oligodendrocyte depletion and transient demyelination [26, 27].

3.2 Timing

Brain edema could begin a few hours after the first treatment with a single dose of
2 Gy to a portion of the brain [28]. Symptoms of increased intracranial pressure or
worsening of primary neurologic symptoms can occur and progress with additional
fractions. With doses up to approximately 60 Gy, symptoms are usually mild and
transient [29, 30].
Subacute reaction (somnolence syndrome, cognitive dysfunction, transient neu-
rologic deficit) usually begins 4–8 weeks after treatment completion. The symp-
toms last from a few days to 6 weeks [14, 24, 31] and spontaneously subside over
few weeks to months [20]; however, somnolence syndrome can cause long-term
neurologic dysfunction [32].

3.3 Risk Factors

3.3.1 Treatment-Related Factors

Radiation therapy: Type of fractionation and fraction size, total dose [33], and field
size [34] are important in the severity of acute brain toxicity. There are more acute
cranial complications in the rapid course of radiation therapy with higher doses per
fraction [5, 35] or multiple daily fractionated radiation therapy [1]. Risk of SS may
be associated with the total dose of radiation [14] and fraction size [14, 18], but
these results need further studies to be better defined [19, 36].
Combined treatment: Combination of surgery [7, 11] or radiosurgery [10] with
fractionated radiation therapy seems to not significantly increase the acute compli-
cation, although surgery type (biopsy versus partial/total resection) may be a signifi-
cant variable to predict the occurrence of acute brain toxicities [1]. Chemotherapy
or radiation sensitizers during cranial irradiation are well tolerated without signifi-
cantly increasing in acute brain toxicity [37–42]. No significant difference has been
reported in the incidence of SS between patients that received radiation alone and
3.4 Symptoms 29

those that received irradiation and intrathecal methotrexate [15]. An obvious


increase in pseudoprogression rate occurs in patients that are treated with chemo-
therapy and radiation therapy versus radiation therapy alone.

3.3.2 Patient-Related Factor

Age over 50, functional status, neurological function, and mental status have not
been reported to be important in the occurrence of acute brain reactions [1].
Somnolence syndrome has not been related to patient factors such as age, initial
white blood count [14], and anatomical site or histological type of brain tumor [43].
Glioblastoma patients with promoter methylation of the repair enzyme
06-­methylguanine-DNA methyltransferase (MGMT) are at a higher risk of tumor
pseudoprogression [23].

3.4 Symptoms

In the acute phase, all presenting symptoms are related to increased intracranial
pressure including headache, nausea, vomiting, and mental status changes or an
exacerbation of the symptoms or signs caused by the lesion being treated [44].
These symptoms can be controlled with medication and are transient with conven-
tional fractionation (1.8–2 Gy/day given five times per week) to a total dose of
60 Gy to the whole brain or even higher doses to limited volumes [44]. However, in
very few patients with higher doses per fraction (more than 3 Gy) to a large area of
brain and in patients with significant pretreatment elevated intracranial pressure,
cerebral herniation may occur.
Imaging including computed tomography (CT) scan or MRI in acute radiation
brain injury is usually unremarkable and could differ from no clear changes to dif-
fuse brain swelling (Fig. 3.1) [45].
Somnolence is referred to as excessive drowsiness or sleep [15, 46]. The
somnolence syndrome is a collection of symptoms that consists of drowsiness,
lethargy, fatigue, anorexia, headache, dysphagia, dysarthria, nausea, and vomit-
ing and is sometimes associated with low-grade fever [36, 47–50]. Brain MRI
may demonstrate nonspecific white matter hyperintensity [51]. The cerebral spi-
nal fluid (CSF) analysis of these patients shows increased levels of protein and
pleocytosis [17], and electroencephalogram (EEG) shows diffuse generalized
slowing in electrocortical activity more than expected for all children that
receive cranial radiation [52].
Somnolence syndrome in adults may manifest with some different symptoms
from pediatric patients due to much greater dosage of radiation therapy applied in
adults [47]. A transient worsening in primary symptoms (e.g., epileptic seizure,
increased paralysis, etc.) can develop in addition to other classic symptoms of som-
nolence syndrome [47]. These symptoms may be severe enough to need supportive
treatment and steroid administration [24]. A transient cognitive dysfunction may
30 3 Radiation Brain Injury

Fig. 3.1 Increased edema in a patient that treated with radiation therapy for glioblastoma multi-
form. (a) Before starting of radiation therapy, (b) 6 weeks after completion of radiation therapy

occur during 6–8 weeks after the end of radiation therapy [53]. MRI findings in the
postradiation phase can vary from non-enhancing white matter hyperintensities on
T2-weighted imaging, indicative of edema, to an increased size of contrast-­
enhancing lesions within or near the irradiated tumor volume due to radiation-­
induced endothelial damage and blood-brain barrier disruption [22, 45].

3.5 Diagnosis

The diagnosis of acute brain reactions is based on clinical picture and response
assessment to steroid treatment.
Somnolence syndrome is characterized by excessive drowsiness or sleep accom-
panied by signs of elevated intracranial pressure such as headache, anorexia or nau-
sea, and vomiting. It is diagnosed based on a group of symptoms that starts
4–8 weeks after radiation therapy, especially in children.
In patients with clinical deterioration after brain radiation therapy, distin-
guishing between tumor progression and radiation reaction is challenging.
Imaging cannot help in this regard unless in new lesions appear outside the
radiation field. Symptoms related to treatment injury usually respond to escalat-
ing or starting steroid treatment. Patients closely follow-up and reimaging at
approximately 4-week intervals are a logical approach in these patients [30, 54].
Biopsy is an invasive action with confounding results and is not recommended
in this setting. The value of metabolic imaging like positron emission tomogra-
phy (PET) scan in this situation is unclear [55]. New functional MRI techniques
such as magnetic resonance spectroscopy or diffusion-weighted and perfusion
seem to be promising [56]. Further investigations are needed before they are
incorporated into widespread use.
3.7 Prevention and Management 31

Table 3.1 RTOG/EROTC radiation-induced brain injury grading [57]


Definition
Grade 1 Fully functional status (i.e., able to work) with minor neurologic findings, no
medication needed
Grade 2 Neurologic findings present sufficient to require home case/nursing assistance may
be required/medications including steroids/antiseizure agents may be required
Grade 3 Neurologic findings requiring hospitalization for initial management
Grade 4 Serious neurologic impairment which includes paralysis, coma, or seizures >3 per
week despite medication/hospitalization required

Table 3.2 Somnolence syndrome scale of Littman


Definition
None No evidence of change in behavior
Minimal Disturbance with some tiredness but activity not curtailed
Mild Decreased activity and increased tiredness, may have a low-grade pyrexia
Moderate Sleeping much of the day, decreased appetite, low-grade fever, most activities
curtailed
Severe Inactive, sleeping 18–20 h per day, low-grade fever, markedly decreased appetite,
and only taking oral fluids

3.6 Scoring

Different scoring systems have been defined for radiation-induced injury. Two of
them (RTOG/EORTC and Littman SS scale) are given in Tables 3.1 and 3.2.
Somnolence symptoms may be assessed by a patient-completed daily diary in
which main symptoms like drowsiness, sleep, fatigue, lethargy, mental concentra-
tion, or appetite are scored by patients with visual analogue scales consisting of
100 mm long with opposing sensations [58]. Littman et al. [19] provided an
observer-based scale (Table 3.2).

3.7 Prevention and Management

It has been recommended that pre-radiation corticosteroid administration should be


considered in patients with significant peritumoral brain edema and who have
symptomatic brain edema. Asymptomatic patients with minimal brain edema could
be spared from pretreatment corticosteroids [59, 60].
A short course of corticosteroids may be helpful in patients with acute and sub-
acute brain reaction to the irradiation.
Corticosteroids usually resolve the symptoms of somnolence syndrome; how-
ever, optimal steroid schedule for its management is still unknown [61]. The preven-
tive effect of steroids [31, 36, 43] and antidepressants [62] to reduce the incidence
of SS has been proposed. These results are needed to be tested in further studies
before conclusion.
32 3 Radiation Brain Injury

Dexamethasone is the drug of choice between steroids due to its long half-life,
low mineralocorticoid activity, and a relatively low effect on cognition [63].
Dexamethasone causes general improvement in patient condition within 4–6 h [64,
65], and neurological improvement occurred within 24–72 h in majority of patients
[66]; however, the pressure may not be consistently reduced until 2–4 days after
initial dosing [67].

3.7.1 Dexamethasone Prescription

Prophylactic:
Dexamethasone (oral or parenteral): 4–16 mg daily for the first 2 weeks of radiation
therapy [51, 59].
Therapeutic:
10 mg IV stat then 4 mg IM/IV every 6 h [51]

Corticosteroids have rapid and complete gastrointestinal absorption, and oral and
parenteral dosing is equivalent. Parenteral therapy should be converted to oral ther-
apy at the earliest appropriate opportunity [68].
The dose can be divided over 4 doses, but owing to its biologic half-life (about
36–54 h), dosing once or twice a day would be a rational practice [69].
Based on UptoDate “if dexamethasone dose of 16 mg is insufficient, the dose can
be increased up to 100 mg per day.”
A proton pomp inhibitor like omeprazole should be coadministered for gastric
protection with concomitant use of NSAIDs or bisphosphonates in the immediate
postoperative period, in those patients receiving unusually high doses of corticoste-
roids or previous history of gastrointestinal bleeding [70].
Some guidelines recommend Pneumocystis carinii prophylaxis with trimethoprim-­
sulfamethoxazole in patients with dexamethasone treatment during concomitant
chemoradiation therapy [63, 71].
In patients with diabetic mellitus, additional medications during steroid therapy
need to be increased based on blood glucose levels.
Higher doses of dexamethasone may be needed to control brain edema in patients
with anticonvulsants (e.g., phenytoin, carbamazepine and phenobarbital), due to
their ability to induce hepatic microsomes [70, 72]. In addition, serum phenytoin
concentration may be interfered with by steroids [73].
After maximal clinical improvement, dexamethasone dose is reduced to the low-
est effective dose and after the completion of radiation therapy is tapered over
2–4 weeks and discontinued [59, 74]. A gradual tapering of dexamethasone should
be considered because dexamethasone doses used in brain tumor patients can sup-
press the hypothalamic-pituitary-adrenocortical axis when it is given for more than
2 weeks. More importantly, gradual discontinuation of dexamethasone can prevent
rebound edema and recurrence of symptoms that may occur even with less than
14 days of treatment [68]. With less than 5–7 days of steroid use, treatment can usu-
ally be abruptly discontinued [75].
References 33

There are various steroid tapering guidelines in cancer patients. Guidelines for
use of steroids in cancer patients recommend that steroids be discontinued abruptly
if less than 4 mg per day of dexamethasone has been used for less than 3 weeks;
otherwise, a gradual tapering is indicated [76]. BC Cancer Agency Cancer
Management Guidelines for dexamethasone tapering recommends reducing by
4 mg every 5 days [71]. With any symptoms returning or worsening during tapering,
the dexamethasone should be return to the previous dose [77].
Because of the corticosteroid complications, other agents that can reduce vaso-
genic brain edema without corticosteroid side effects are under investigation.
Corticorelin acetate (a synthetic peptide formulation of the normal endogenous
human corticotropin-releasing factor) [78–81], vascular endothelial growth factor
(VEGF) receptor, tyrosine kinase inhibitors such as cediranib or monoclonal anti-
bodies (e.g., Bevacizumab [82, 83], and selective COX-2 inhibitors [84] are some of
these medications.

References
1. Lawrence YR, Wang M, Dicker AP, Andrews D, Curran WJ, Michalski JM et al (2011) Early
toxicity predicts long-term survival in high-grade glioma. Br J Cancer 104(9):1365–1371
2. Goetz P, Ebinu JO, Roberge D, Zadeh G (2012) Current standards in the management of cere-
bral metastases. Int J Surg Oncol 2012:493426
3. Young DF, Posner JB, Chu F, Nisce L (1974) Rapid-course radiation therapy of cerebral
metastases: results and complications. Cancer 34(4):1069–1076
4. Borgelt B, Gelber R, Larson M, Hendrickson F, Griffin T, Roth R (1981) Ultra-rapid high dose
irradiation schedules for the palliation of brain metastases: final results of the first two studies
by the radiation therapy oncology group. Int J Radiat Oncol Biol Phys 7(12):1633–1638
5. Harwood AR, Simpson WJ (1977) Radiation therapy of cerebral metastases: a randomized
prospective clinical trial. Int J Radiat Oncol Biol Phys 2(11):1091–1094
6. Rades D, Haatanen T, Schild SE, Dunst J (2007) Dose escalation beyond 30 grays in 10 frac-
tions for patients with multiple brain metastases. Cancer 110(6):1345–1350
7. Vecht CJ, Haaxma-Reiche H, Noordijk EM, Padberg GW, Voormolen JH, Hoekstra FH et al
(1993) Treatment of single brain metastasis: radiotherapy alone or combined with neurosur-
gery. Ann Neurol 33(6):583–590
8. Rades D, Bohlen G, Pluemer A, Veninga T, Hanssens P, Dunst J et al (2007) Stereotactic radio-
surgery alone versus resection plus whole-brain radiotherapy for 1 or 2 brain metastases in
recursive partitioning analysis class 1 and 2 patients. Cancer 109(12):2515–2521
9. O’Neill BP, Iturria NJ, Link MJ, Pollock BE, Ballman KV, O’Fallon JR (2003) A comparison
of surgical resection and stereotactic radiosurgery in the treatment of solitary brain metastases.
Int J Radiat Oncol Biol Phys 55(5):1169–1176
10. Andrews DW, Scott CB, Sperduto PW, Flanders AE, Gaspar LE, Schell MC et al (2004)
Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with
one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet
363(9422):1665–1672
11. Muacevic A, Wowra B, Siefert A, Tonn J-C, Steiger H-J, Kreth FW (2008) Microsurgery plus
whole brain irradiation versus gamma knife surgery alone for treatment of single metastases to
the brain: a randomized controlled multicentre phase III trial. J Neuro-Oncol 87(3):299–307
12. Malmström A, Grønberg BH, Marosi C, Stupp R, Frappaz D, Schultz H et al (2012)
Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in
patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet
Oncol 13(9):916–926
34 3 Radiation Brain Injury

13. Posner JB (1995) Neurologic complications of cancer. Oxford University Press, Oxford
14. Ch’ien LT, Aur RJ, Stagner S, Cavallo K, Wood A, Goff J et al (1980) Long-term neurological
implications of somnolence syndrome in children with acute lymphocytic leukemia. Ann
Neurol 8(3):273–277
15. Freeman J, Johnston P, Voke J (1973) Somnolence after prophylactic cranial irradiation in
children with acute lymphoblastic leukaemia. Br Med J 4(5891):523–525
16. Garwicz S, Aronson A, Elmqvist D, Landberg T (1975) Postirradiation syndrome and
EEG findings in children with acute lymphoblastic leukaemia. Acta Paediatr Scand
64(3):399–403
17. Siemes H, Rating D, Siegert M, Hanefeld F, Müller S, Gadner H et al (1980) Changes of CSF-­
protein pattern in children with acute lymphoblastic leukemia during prophylactic CNS ther-
apy (Berlin protocol). Med Pediatr Oncol 8(1):25–34
18. Parker D, Malpas J, Sandland R, Sheaff P, Freeman J, Paxton A (1978) Outlook following
“somnolence syndrome” after prophylactic cranial irradiation. Br Med J 1(6112):554
19. Littman P, Rosenstock J, Gale G, Krisch RE, Meadows A, Sather H et al (1984) The somno-
lence syndrome in leukemic children following reduced daily dose fractions of cranial radia-
tion. Int J Radiat Oncol Biol Phys 10(10):1851–1853
20. Rahman AA, Mannan M, Sadeque S (2008) Acute and long-term neurological complications
in children with acute lymphoblastic leukemia. Bangladesh Med Res Counc Bull
34(3):90–93
21. Parvez K, Parvez A, Zadeh G (2014) The diagnosis and treatment of pseudoprogression, radia-
tion necrosis and brain tumor recurrence. Int J Mol Sci 15(7):11832–11846
22. Brandsma D, Stalpers L, Taal W, Sminia P, van den Bent MJ (2008) Clinical features, mecha-
nisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol
9(5):453–461
23. Brandes AA, Franceschi E, Tosoni A, Blatt V, Pession A, Tallini G et al (2008) MGMT pro-
moter methylation status can predict the incidence and outcome of pseudoprogression after
concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol
26(13):2192–2197
24. Boldrey E, Sheline G (1966) Delayed transitory clinical manifestations after radiation treat-
ment of intracranial tumors. Acta Radiol Ther Phys Biol 5:5–10
25. Faithfull S, Brada M (1998) Somnolence syndrome in adults following cranial irradiation for
primary brain tumours. Clin Oncol 10(4):250–254
26. Armstrong C, Ruffer J, Corn B, DeVries K, Mollman J (1995) Biphasic patterns of memory
deficits following moderate-dose partial-brain irradiation: neuropsychologic outcome and pro-
posed mechanisms. J Clin Oncol 13(9):2263–2271
27. Rider W (1963) Radiation damage to the brain—a new syndrome. J Can Assoc Radiol
14:67–69
28. Kramer S, Lee KF (1974) Complications of radiation therapy: the central nervous system.
Semin Roentgenol 9:75–83
29. Liang BC (1999) Radiation-associated neurotoxicity. Neurol Clin 35:54–58
30. Walker AJ, Ruzevick J, Malayeri AA, Rigamonti D, Lim M, Redmond KJ et al (2014)
Postradiation imaging changes in the CNS: how can we differentiate between treatment effect
and disease progression? Future Oncol 10(7):1277–1297
31. Uzal D, Özyar E, Hayran M, Zorlu F, Atahan L, Yetkin S (1998) Reduced incidence of the
somnolence syndrome after prophylactic cranial irradiation in children with acute lymphoblas-
tic leukemia. Radiother Oncol 48(1):29–32
32. McTyre E, Scott J, Chinnaiyan P (2013) Whole brain radiotherapy for brain metastasis. Surg
Neurol Int 4(Suppl 4):S236
33. Kiebert G, Curran D, Aaronson N, Bolla M, Menten J, Rutten E et al (1998) Quality of life
after radiation therapy of cerebral low-grade gliomas of the adult: results of a randomised
phase III trial on dose response (EORTC trial 22844). Eur J Cancer 34(12):1902–1909
References 35

34. Garden AS, Maor MH, Yung WA, Bruner JM, Woo SY, Moser RP et al (1991) Outcome and
patterns of failure following limited-volume irradiation for malignant astrocytomas. Radiother
Oncol 20(2):99–110
35. Chao JH, Phillips R, Nickson JJ (1954) Roentgen-ray therapy of cerebral metastases. Cancer
7(4):682–689
36. Mandell LR, Walker RW, Steinherz P, Fuks Z (1989) Reduced incidence of the somnolence
syndrome in leukemic children with steroid coverage during prophylactic cranial radiation
therapy. Results of a pilot study. Cancer 63(10):1975–1978
37. Walker MD, Green SB, Byar DP, Alexander E Jr, Batzdorf U, Brooks WH et al (1980)
Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant
glioma after surgery. N Engl J Med 303(23):1323–1329
38. Bleehen N, Stenning SA (1991) Medical Research Council trial of two radiotherapy doses in
the treatment of grades 3 and 4 astrocytoma. The Medical Research Council brain tumour
working party. Br J Cancer 64(4):769
39. Taphoorn M, Stupp R, Coens C, Osoba D, Kortmann R, van den Bent M et al (2005) European
organisation for research and treatment of cancer brain tumour group; EORTC radiotherapy
group; national cancer institute of Canada clinical trials group. Health-related quality of life in
patients with glioblastoma: a randomised controlled trial. Lancet Oncol 6(12):937–944
40. Kocher PM, Eich H-T, Semrau R, Güner SA, Müller R-P (2005) Phase I/II trial of simultane-
ous whole-brain irradiation and dose-escalating topotecan for brain metastases. Strahlenther
Onkol 181(1):20–25
41. Abrey LE, Christodoulou C (2001) Temozolomide for treating brain metastases. Semin Oncol
28:34–42
42. Suh JH, Stea B, Nabid A, Kresl JJ, Fortin A, Mercier J-P et al (2006) Phase III study of
efaproxiral as an adjunct to whole-brain radiation therapy for brain metastases. J Clin Oncol
24(1):106–114
43. Powell C, Guerrero D, Sardell S, Cumins S, Wharram B, Traish D et al (2011) Somnolence
syndrome in patients receiving radical radiotherapy for primary brain tumours: a prospective
study. Radiother Oncol 100(1):131–136
44. Leibel SA, Sheline GE (1987) Radiation therapy for neoplasms of the brain. J Neurosurg
66(1):1–22
45. Sanghvi D (2015) Post-treatment imaging of high-grade gliomas. Indian J Radiol Imaging
25(2):102
46. Williams RL, Karacan IE, Moore CA (1998) Sleep disorders: diagnosis and treatment. Wiley,
New York
47. Faithfull S (1991) Patients’ experiences following cranial radiotherapy: a study of the somno-
lence syndrome. J Adv Nurs 16(8):939–946
48. Druckmann A (1929) Schlafsucht als Folge der Röntgenbestrahlung. Beitrag zur
Strahlenempfindlichkeit des Gehirns. Strahlentherapie 33(382–84):268
49. Vern TZ, Salvi S (2009) Somnolence syndrome and fever in pediatric patients with cranial
irradiation. J Pediatr Hematol Oncol 31(2):118–120
50. Kelsey CR, Marks LB (2006) Somnolence syndrome after focal radiation therapy to the pineal
region: case report and review of the literature. J Neuro-Oncol 78(2):153–156
51. Ballonoff A. (2016) Acute complications of cranial irradiation. Available from https://www.
uptodate.com/contents/acute-complications-of-cranial-irradiation. Accessed 10 Aug 2016
52. Pampiglione G (1974) Letter: somnolence in children with acute leukemia. Br Med
J 1(5899):158
53. Tallet AV, Azria D, Barlesi F, Spano J-P, Carpentier AF, Gonçalves A et al (2012) Neurocognitive
function impairment after whole brain radiotherapy for brain metastases: actual assessment.
Radiat Oncol 7(1):1
54. Halperin EC, Brady LW, Wazer DE, Perez CA (2013) Perez & Brady’s principles and practice
of radiation oncology. Lippincott Williams & Wilkins, Philadelphia, PA
36 3 Radiation Brain Injury

55. Taal W, Brandsma D, de Bruin HG, Bromberg JE, Swaak-Kragten AT, Sillevis Smitt PA et al
(2008) Incidence of early pseudo-progression in a cohort of malignant glioma patients treated
with chemoirradiation with temozolomide. Cancer 113(2):405–410
56. Zeng Q-S, Li C-F, Liu H, Zhen J-H, Feng D-C (2007) Distinction between recurrent glioma
and radiation injury using magnetic resonance spectroscopy in combination with diffusion-­
weighted imaging. Int J Radiat Oncol Biol Phys 68(1):151–158
57. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
58. Faithfull S (1992) The diary method for nursing research: a study of somnolence syndrome.
Eur J Cancer Care 1(2):13–18
59. Ryken TC, McDermott M, Robinson PD, Ammirati M, Andrews DW, Asher AL et al (2010)
The role of steroids in the management of brain metastases: a systematic review and evidence-­
based clinical practice guideline. J Neuro-Oncol 96(1):103–114
60. Hempen C, Weiss E, Hess CF (2002) Dexamethasone treatment in patients with brain metas-
tases and primary brain tumors: do the benefits outweigh the side-effects? Support Care Cancer
10(4):322–328
61. Woodford K (2007) Somnolence syndrome after cranial irradiation: a literature review. The
Radiographer 54(3):30
62. Proctor S, Kernahan J, Taylor P (1981) Depression as component of post-cranial irradiation
somnolence syndrome. Lancet 317(8231):1215–1216
63. Roth P, Wick W, Weller M (2010) Steroids in neurooncology: actions, indications, side-effects.
Curr Opin Neurol 23(6):597–602
64. Alberti E, Hartmann A, Schütz H-J, Schreckenberger F (1978) The effect of large doses of
dexamethasone on the cerebrospinal fluid pressure in patients with supratentorial tumors.
J Neurol 217(3):173–181
65. Jarden J, Dhawan V, Moeller J, Strother S, Rottenberg D (1989) The time course of steroid
action on blood-to-brain and blood-to-tumor transport of 82Rb: a positron emission tomo-
graphic study. Ann Neurol 25(3):239–245
66. Soffietti R, Cornu P, Delattre J, Grant R, Graus F, Grisold W et al (2006) EFNS guidelines on
diagnosis and treatment of brain metastases: report of an EFNS task force. Eur J Neurol
13(7):674–681
67. Miller JD, Leech P (1975) Effects of mannitol and steroid therapy on intracranial volume-­
pressure relationships in patients. J Neurosurg 42(3):274–281
68. Nahaczewski AE, Fowler SB, Hariharan S (2004) Dexamethasone therapy in patients with
brain tumors-a focus on tapering. J Neurosurg Nurs 36(6):340–343
69. Vecht CJ, Hovestadt A, Verbiest H, Van Vliet J, Van Putten W (1994) Dose-effect relationship
of dexamethasone on Karnofsky performance in metastatic brain tumors a randomized study
of doses of 4, 8, and 16 mg per day. Neurology 44(4):675
70. Dietrich J, Rao K, Pastorino S, Kesari S (2011) Corticosteroids in brain cancer patients: ben-
efits and pitfalls. Expert Rev Clin Pharmacol 4(2):233–242
71. BCCA protocol summary for dexamethasone as treatment for cerebral edema or CNS swell-
ing. http://www.bccancer.bc.ca/chemotherapy-protocolssite/Documents/Supportive%20Care/
SCDEXA_Protocol_1May09.pdf
72. Ruegg S (2002) Dexamethasone/phenytoin interactions: neurooncological concerns. Swiss
Med Wkly 132(29/30):425–426
73. Lawson LA, Blouin RA, Smith RB, Rapp RP, Young AB (1981) Phenytoin-dexamethasone
interaction: a previously unreported observation. Surg Neurol 16(1):23–24
74. Weissman DE, Janjan NA, Erickson B, Wilson FJ, Greenberg M, Ritch PS et al (1991) Twice-­
daily tapering dexamethasone treatment during cranial radiation for newly diagnosed brain
metastases. J Neuro-Oncol 11(3):235–239
75. Szabo GC, Winkler SR (1995) Withdrawal of glucocorticoid therapy in neurosurgical patients.
Surg Neurol 44(5):498
References 37

76. Guidelines for use of steroids in cancer patients. Available from ttps://www.researchgate.net/
fi l e . P o s t F i l e L o a d e r. h t m l ? i d = 5 6 9 a 6 3 f 3 5 f 7 f 7 1 a 5 d a 8 b 4 5 8 7 & a s s e t Key = A S % 3
A318546062577664%401452958707609
77. Kostaras X, Cusano F, Kline G, Roa W, Easaw J (2014) Use of dexamethasone in patients with
high-grade glioma: a clinical practice guideline. Curr Oncol 21(3):e493
78. Recht L, Mechtler LL, Wong ET, O’Connor PC, Rodda BE (2013) Steroid-sparing effect of
corticorelin acetate in peritumoral cerebral edema is associated with improvement in steroid-­
induced myopathy. J Clin Oncol 31(9):1182–1187
79. Tjuvajev J, Uehara H, Desai R, Beattie B, Matei C, Zhou Y et al (1996) Corticotropin-releasing
factor decreases vasogenic brain edema. Cancer Res 56(6):1352–1360
80. Villalona-Calero M, Eckardt J, Burris H, Kraynak M, Fields-Jones S, Bazan C et al (1998) A
phase I trial of human corticotropin-releasing factor (hCRF) in patients with peritumoral brain
edema. Ann Oncol 9(1):71–77
81. Moliterno JA, Henry E, Pannullo SC (2009) Corticorelin acetate injections for the treatment of
peritumoral brain edema. Expert Opin Investig Drugs 18(9):1413–1419
82. Vredenburgh JJ, Cloughesy T, Samant M, Prados M, Wen PY, Mikkelsen T et al (2010)
Corticosteroid use in patients with glioblastoma at first or second relapse treated with bevaci-
zumab in the BRAIN study. Oncologist 15(12):1329–1334
83. Batchelor TT, Sorensen AG, di Tomaso E, Zhang W-T, Duda DG, Cohen KS et al (2007)
AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and
alleviates edema in glioblastoma patients. Cancer Cell 11(1):83–95
84. Portnow J, Suleman S, Grossman SA, Eller S, Carson K (2002) A cyclooxygenase-2 (COX-2)
inhibitor compared with dexamethasone in a survival study of rats with intracerebral 9L glio-
sarcomas. Neuro-Oncology 4(1):22–25
Radiation Orbital Toxicity
4

The orbit contains the globe and connective tissues including extraocular muscles,
orbital fat, eyelids, eyelashes, lacrimal gland, and lacrimal draining system. Different
parts of the globe (e.g., the lens, iris, conjunctiva, sclera, retina, ciliary body, cornea,
and optic nerve) have different sensitivities to irradiation. Some structures like the
optic nerve respond to radiation in a chronic manner (late-responding tissue), and
some like the conjunctiva and eyelids are acute-responding tissues. Some parts of
the mentioned structures are very sensitive, and some of them like the lens and
sclera are very resistant to radiation. The orbital area may be irradiated as the treat-
ment target or as an organ at risk in the vicinity of the target. Radiation-induced
acute side effects in this area require prompt attention to prevent long-term issues
like ophthalmitis, permanent dry eyes, and impaired visual acuity. In this chapter,
we will focus on more sensitive acute-responding tissues.
Most of the acute side effects of orbital radiation therapy have a mild severity,
and there is often no need for treatment break [1–10]. These acute side effects (e.g.,
dry eyes, excessive tearing, conjunctivitis, transient periorbital erythema, and
edema) have been reported in 30–50% of patients based on volume of the area
within the radiation field [2, 3, 10–13]. Based on animal studies, the acute side
effects of orbital radiation therapy in decreasing frequency include conjunctivitis,
blepharitis, keratoconjunctivitis sicca, keratitis, and ulcerative keratitis [14].
The orbit is a small area that compared with its size, consists of a collection of
many different structures as mentioned above. Each orbital structure has varying
sensitivity to radiation effects and different signs and symptoms will develop at dif-
ferent times during radiation therapy. Here, the acute radiation effects will be
reviewed for different orbital structures separately in each section.

© Springer International Publishing AG 2017 39


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_4
40 4 Radiation Orbital Toxicity

4.1 Blepharitis and Eyelid Dermatitis

The eyelid is a thin fold of skin and its response to radiation is more than skin at
other sites. It also contains hair follicles in its free edge that are sensitive to radia-
tion. Radiation blepharitis (eyelid edge inflammation), as a radiation dermatitis,
begins with erythema followed by dry and moist desquamation (and rarely necro-
sis). Erythema takes approximately 2–4 weeks to develop after radiation exposure;
it is usually transient and disappears rapidly. Moist desquamation of skin can
develop after 5–6 weeks of radiation therapy (50–60 Gy in 1.8–2.2 Gy daily frac-
tions). Healing is typically slow and may take up to 6–12 weeks [15, 16].
The following guidelines will provide comfort and healing of the periocular skin
radiation-induced dermatitis and blepharitis [17]:

• Washing the eyelids with lukewarm water and mild soap and gently drying at
least once daily.
• Applying an eyelid moisturizer, black tea soaks (soak tea bags in hot water, allow
them to cool, lie down, and put them over the eyes).
• Avoiding extremes of heat and cold, skin irritants, and any eyelid rubbing or
scratching friction.
• More severe dermatitis in eyelid skin may need a radiation break and should be
treated like a skin burn using silver sulfadiazine ointment or similar medications.

4.2 Eyelash Loss

Loss of the eyelashes may occur (Fig. 4.1) with doses more than 10–20 Gy (1.5–2 Gy
per fraction) [16, 18]. It may be temporary and regrowth after approximately
2 months [15] or may be permanent at radiation doses of more than 30 Gy [16].
Eyelash loss can result in irritation of the conjunctiva and cornea due to loss of
the protective blink reflex. These complications are discussed below.

4.3 Conjunctivitis

Signs and symptoms of radiation-induced conjunctivitis include conjunctival injec-


tion, watery discharge, and discomfort that occur 1–3 weeks after the start of radia-
tion treatment. Acute conjunctivitis is common with doses ≥30 Gy [11, 19].
Secondary infectious conjunctivitis (mostly bacterial or rarely viral) may also occur
[20] (Fig. 4.1). A thicker, purulent, yellow-green discharge should raise suspicion
for bacterial conjunctivitis [21].
Radiation conjunctivitis is a clinical diagnosis based on patient signs and symp-
toms including a red eye, discharge, and normal vision as well as exclusion of other
diagnoses in patients with a history of irradiation to or near the orbit [21].
4.4 Xerophthalmia 41

Fig. 4.1 Conjunctivitis


and eyelash loss at the end
of radiation therapy for
advanced maxillary sinus
undifferentiated carcinoma

The risk of conjunctival toxicity can be reduced by positioning the orbit out of
the radiation field or reducing dose buildup on the conjunctival surface by higher
beam energy or treating patients with the eye open during radiation exposure if eyes
are irradiated from the anterior aspect of the orbit [20].
It is recommended that artificial tears be used 4–8 times daily to relieve the irrita-
tion caused by conjunctivitis [20].
Secondary infectious conjunctivitis should be managed as a primary form. Viral
conjunctivitis is self-limited and topical antihistamine/decongestants may be needed
for symptom relief. The efficacy of antiviral agents is not clear [22, 23]. Bacterial
conjunctivitis is treated with a broad-spectrum topical antibiotic (trimethoprim with
polymyxin B eye drops or erythromycin ophthalmic ointment). Gram stain or cul-
tures generally are not carried out, and empiric antibiotics are usually prescribed
[17, 24, 25].

4.4 Xerophthalmia

Tears are produced by the lacrimal functional unit consisting of the lacrimal glands,
ocular surface (cornea and conjunctiva), eyelids, meibomian glands, and the inter-
connecting innervation [26, 27].
The tear film consists of three layers: mucous (inner layer), aqueous (middle
layer), and lipid (outer layer).
The mucous layer is produced by conjunctival goblet cells and epithelial
cells of the cornea and conjunctiva. The lacrimal glands consist of the main
lacrimal gland and the accessory Krause and Wolfring lacrimal glands, which
are located around the upper border of the tarsal plate and the conjunctival for-
nix, respectively. The aqueous component is secreted by these glands. The lipid
component is secreted by the meibomian glands (at the rim of the eyelids inside
the tarsal plate) and Zeis glands (anterior to the Meibomian gland, at distal eye-
lid margin) [28].
42 4 Radiation Orbital Toxicity

Radiation therapy leads to damage to the meibomian glands [29] and cause lac-
rimal gland acinar cell apoptosis and gland atrophy [30] or both [27]. Tear film
aqueous and lipid deficiency develop due to dysfunction of lacrimal glands and
Meibomian gland, respectively [20, 31].
With low-dose radiation therapy (about 24–25 Gy) of lymphoid lesions of the
orbit and ocular adnexa, early mild radiation-induced xerophthalmia and chemosis
have been noted in up to 50% of patients [12]. Irradiation of lacrimal glands to doses
more than 30–40 Gy can lead to dry eye syndrome; the incidence increases dramati-
cally with doses ≥50 Gy and doses greater than 60 Gy may cause permanent tear
loss [20, 32, 33].
Diagnosis is based on characteristic symptoms and clinical appearance. Early
effects are conjunctival inflammation, chemosis (swelling of the conjunctiva), and
tear film instability with a resultant dry eye sensation [34]. Patients develop a red,
painful, scratchy eye (foreign-body sensation), and photophobia. Severe problems
may produce corneal desiccation, ulceration with bacterial infection, neovascular-
ization, opacification, and ultimately perforation [35].
Xerophthalmia can be avoided by positioning lacrimal glands out of the radiation
field, shielding them, or modifying dose distributions by using intensity-modulated
radiation therapy (IMRT) [36, 37].
Supplemental lubrication or artificial tears are the mainstay of treatment for
xerophthalmia; however, they act only as a replacement therapy without any effect
on tear secretion. There are various artificial tear products with different formula-
tions with no evidence suggesting a specific single brand being superior [38].
Preservative-free eye drops contain fewer additives and are generally recommended
in the setting of extreme and long-term use of teardrop including severe dry eyes or
multiple teardrop application daily due to lower effects of these products on the
cornea and conjunctival epithelium [39].
In addition to artificial teardrops, there are other forms of supplemental lubrica-
tions like gels or ointments. Artificial tear ointments and gels offer longer-lasting
relief but may blur vision [38].
There is no proven difference in efficacy between different topical dry eye treat-
ments, including artificial tears, ointments, or gels [40].

Artificial Tear Prescription:


Solution: Instill 1–2 drops into eye(s) as necessary to relieve symptoms [39].
Ointment: Pull down lower lid of affected eye and apply 0.25 mL of ointment to the
inside of the eyelid [39].

Stimulation of tear production has been seen by using topical sodium hyaluronate,
cyclosporine, and tacrolimus, which increase the aqueous component of the tear layer
and goblet cell density while also decreasing the inflammation process [41, 42]. These
agents need to be studied in radiation-induced xerophthalmia. Oral pilocarpine could
palliate dry eye and dry mouth symptoms in Sjögren’s syndrome [43, 44] and has also
been used in the management and prevention of radiation-­induced xerostomia (see
Chap. 7). But currently, there is no evidence for its application in radiation-induced
xerophthalmia.
4.5 Corneal Toxicity 43

Topical anti-inflammatory agents (corticosteroids or nonsteroidal anti-­


inflammatory drugs (NSAIDs)) are beneficial for selective patients with severe dry
eyes [45], but there isn’t sufficient evidence for their efficacy in radiation-induced
xerophthalmia.
Autologous serum eye drops (with essential tear components for epithelial pro-
liferation, similar properties of tears, and no additive preservatives) [46], punctal
occlusion (mechanical blocking of the draining system) [47], and dry eye mois-
ture chamber goggles (a chamber for protecting the dry eyes from the environ-
ment) [20], all have been suggested for severe dry eye treatment with some
limitations in clinical use including need for frequent blood tests and potential
risk of infection transmission in autologous serum eye drops, insufficient evi-
dence of efficacy in punctal occlusion, and cosmetic concerns in wearing the
moisture chamber goggles.

4.5 Corneal Toxicity

Acute corneal toxicity of radiation therapy can be secondary to radiation xeroph-


thalmia or a primary effect of irradiation on the corneal surface epithelium, corneal
stroma, and endothelium [20].
Secondary keratopathy due to tear film dysfunction is the common form of acute
corneal toxicity and is presented by superficial punctate epithelial erosions or, in the
case of severe dry eyes, corneal scarring [34].
Direct radiation injury can induce punctuate corneal erosions and corneal edema
at doses of 40–50 Gy in 4–5 weeks and corneal ulceration at doses more than 60 Gy
(with conventional fractionated radiation) and with 20 Gy delivered in a single dose
[48].
Corneal damage is diagnosed clinically by signs and symptoms and eye exami-
nation including slit lamp exam. Patients usually present with photophobia, foreign-­
body sensation, tearing, pain, haloes, decreased vision, and red eye. In the penlight
examination, conjunctival injection may be present. In slit lamp microscope exami-
nation, a corneal opacity or infiltration with corneal ulcers could be seen. The cor-
nea may have a hazy appearance with corneal edema. Punctate epithelial erosions
and corneal ulcers stain positively with fluorescein.
A corneal shield, which is placed between the lids and globe, could minimize
corneal radiation dose and prevent toxicity if the treatment target is superficial to the
cornea. These internal eye shields are used in radiation treatment planning of skin
cancer near the eye and can give excellent protection from lower-energy X-rays and
electron beams with energies ≥6 MeV [49, 50].

4.5.1 Treatment

4.5.1.1 Topical Lubricants (e.g., Ophthalmic Ointment)


Patient should be instructed to pull down the lower lid of the affected eye and apply
a small amount of ointment once daily or more if needed [51, 52].
44 4 Radiation Orbital Toxicity

Some practitioners prescribe lubricant antibiotic ointment (e.g., erythromycin


0.5% ophthalmic ointment, polymyxin B/trimethoprim (Polytrim) ophthalmic solu-
tion, and sulfacetamide 10% ophthalmic ointment) [53].

4.5.1.2 Analgesics
Pain is common with corneal damage and is alleviated with oral nonsteroidal anti-­
inflammatory drugs (NSAIDs) and acetaminophen or narcotic analgesics (in severe
cases). Topical NSAIDs (e.g., diclofenac 0.1%, ketorolac 0.4%), are administered
by some practitioners in selective patients that request immediate pain relief or that
cannot tolerate oral analgesics, but these topical agent have not been approved for
this indication [54].
Patching and topical cycloplegics and topical anesthetics have not shown signifi-
cant benefit for uncomplicated corneal abrasions [53].

4.5.1.3 Ophthalmologist Consult


Early ophthalmologist consultation is recommended and in the setting of significant
vision loss, corneal infiltration or ulcer, vision worsening, or no improvement of
symptoms with initial management, referring a patient to an ophthalmologist is nec-
essary [53].

4.6 Iris Toxicity

Acute complications of radiation therapy in the anterior chamber are rarely reported,
but transient early iritis can develop after single doses ≥10 Gy [20]. Iritis presents
with pain, photophobia, red eye, and blurred vision and is distinguished from other
causes of red eye by slit lamp examination (presence of leukocytes in anterior cham-
ber) [55]. There is little published evidence about management of radiation iritis.
Ophthalmologic consultation should be considered early in these patients.

References
1. Prabhu RS, Liebman L, Wojno T, Hayek B, Hall WA, Crocker I (2012) Clinical outcomes of
radiotherapy as initial local therapy for graves’ ophthalmopathy and predictors of the need for
post-radiotherapy decompressive surgery. Radiat Oncol 7:95
2. Mallick S, Das S, Benson R, Roshan V, Bhasker S (2015) Outcome of primary orbital lym-
phoma treated with induction chemotherapy followed by conformal radiotherapy. J Egypt Natl
Canc Inst 27(3):113–117
3. Zhou P, Ng AK, Silver B, Li S, Hua L, Mauch PM (2005) Radiation therapy for orbital lym-
phoma. Int J Radiat Oncol Biol Phys 63(3):866–871
4. Bessell EM, Henk JM, Whitelocke RA, Wright JE (1987) Ocular morbidity after radiotherapy
of orbital and conjunctival lymphoma. Eye (Lond) 1:90–96
5. Bhatia S, Paulino AC, Buatti JM, Mayr NA, Wen BC (2002) Curative radiotherapy for primary
orbital lymphoma. Int J Radiat Oncol Biol Phys 54(3):818–823
6. Smitt MC, Donaldson SS (1993) Radiotherapy is successful treatment for orbital lymphoma.
Int J Radiat Oncol Biol Phys 26(1):59–66
References 45

7. Bolek TW, Moyses HM, Marcus RB Jr, Gorden L 3rd, Maiese RL, Almasri NM, Mendenhall
NP (1999) Radiotherapy in the management of orbital lymphoma. Int J Radiat Oncol Biol
Phys 44(1):31–36
8. Kennerdell JS, Johnson BL, Deutsch M (1979) Radiation treatment of orbital lymphoid hyper-
plasia. Ophthalmology 86(5):942–947
9. Lanciano R, Fowble B, Sergott RC, Atlas S, Savino PJ, Bosley TM, Rubenstein J (1990) The
results of radiotherapy for orbital pseudotumor. Int J Radiat Oncol Biol Phys 18(2):407–411
10. De Cicco L, Cella L, Liuzzi R, Solla R, Farella A, Punzo G, Tranfa F, Strianese D, Conson M,
Bonavolontà G, Salvatore M, Pacelli R (2009) Radiation therapy in primary orbital lymphoma:
a single institution retrospective analysis. Radiat Oncol 4:60
11. Stafford SL, Kozelsky TF, Garrity JA, Kurtin PJ, Leavitt JA, Martenson JA, Habermann TM
(2001) Orbital lymphoma: radiotherapy outcome and complications. Radiother Oncol
59(2):139–144
12. Kennerdell JS, Flores NE, Hartsock RJ (1999) Low-dose radiotherapy for lymphoid lesions of
the orbit and ocular adnexa. Ophthal Plast Reconstr Surg 15(2):129–133
13. Fasola CE, Jones JC, Huang DD, Le QT, Hoppe RT, Donaldson SS (2013) Low-dose radiation
therapy (2 Gy × 2) in the treatment of orbital lymphoma. Int J Radiat Oncol Biol Phys
86(5):930–935
14. Roberts SM, Lavach JD, Severin GA, Withrow SJ, Gillette EL (1987) Ophthalmic complica-
tions following megavoltage irradiation of the nasal and paranasal cavities in dogs. J Am Vet
Med Assoc 190:43–47
15. Effects of radiation on the eyes. Available from www.cancerresearchuk.org
16. Finger PT (2009) Radiation therapy for orbital tumors: concepts, current use, and ophthalmic
radiation side effects. Surv Ophthalmol 54(5):545–568
17. Yeung K. Bacterial conjunctivitis treatment & management. Available from. http://emedicine.
medscape.com/article/1191730-treatment? pa=Yzv32%2FmiVFacR2DSX4AcojySCmfFtq6u
xIU3d3YOxY%2FbHHZ3T4scerQ1G2lqyNIc8SIvl8zjYv73GUyW5rsbWA%3D%3D
18. Gordon KB, Char DH, Sagerman RH (1995) Late effects of radiation on the eye and ocular
adnexa. Int J Radiat Oncol Biol Phys 31:1123–1139
19. Radiation effects on eye & periorbital tissues. Available from http://isocentre.wikidot.com/
headneck:radiation-effects-on-eye-periorbitaltissues
20. Jeganathan VS, Wirth A, MacManus MP (2011) Ocular risks from orbital and periorbital radi-
ation therapy: a critical review. Int J Radiat Oncol Biol Phys 79:650–659
21. Jacobs DS. Conjunctivitis. Available from www.uptodate.com
22. Skevaki CL, Galani IE, Pararas MV, Giannopoulou KP, Tsakris A (2011) Treatment of viral
conjunctivitis with antiviral drugs. Drugs 71(3):331–347
23. Scott I. Viral conjunctivitis treatment & management. Available from emedicine.medscape.com
24. Yeung K. Bacterial conjunctivitis medication. Available from emedicine.medscape.com
25. Bacterial conjunctivitis. Available from emedicine.medscape.com
26. Stern ME, Beuerman RW, Fox RI, Gao J, Mircheff AK, Pflugfelder SC (1998) The pathology of
dry eye: the interaction between the ocular surface and lacrimal glands. Cornea 17(6):584–589
27. Stern ME, Gao J, Siemasko KF, Beuerman RW, Pflugfelder SC (2004) The role of the lacrimal
functional unit in the pathophysiology of dry eye. Exp Eye Res 78(3):409–416
28. Dry eye disease: pathophysiology, classification, and diagnosis—See more at http://www.
ajmc.com/journals/supplement/2008/2008-04-vol14-n3suppl/apr08-3141ps079-s087/P-­-
1#sthash.cUFHmWYv.dpuf
29. Karp LA, Streeten BW, Cogan DG (1979) Radiation-induced atrophy of the meibomian gland.
Arch Ophthalmol 97:303–305
30. Stephens LC, Schultheiss TE, Peters LJ, Ang KK, Gray KN (1988) Acute radiation injury of
ocular adnexa. Arch Ophthalmol 106(3):389–391
31. Pinard CL, Mutsaers AJ, Mayer MN, Woods JP (2012) Retrospective study and review of
ocular radiation side effects following external-beam cobalt-60 radiation therapy in 37 dogs
and 12 cats. Can Vet J 53(12):1301–1307
46 4 Radiation Orbital Toxicity

32. Brady LW (1996) Ocular complications of high-dose radiotherapy. Oncology (Williston Park)
10(7):981–982, 984
33. Jereb B, Lee H, Jakobiec FA, Kutcher J (1984) Radiation therapy of conjunctival and orbital
lymphoid tumors. Int J Radiat Oncol Biol Phys 10(7):1013–1019
34. Durkin S, Roos D, Higgs B, Casson R, Selva D (2007) Ophthalmic and adnexal complications
of radiotherapy. Acta Ophthalmol Scand 85(3):240–250
35. Parsons JT, Bova FJ, Million RR, Fitzgerald CR (1996) Response of the normal eye to high
dose radiotherapy. Oncology 10(6):837–852
36. Metcalfe P, Chapman A, Arnold A, Arnold B, Tangboonduangjit P, Capp A, Fox C (2004)
Intensity-modulated radiation therapy: not a dry eye in the house. Australas Radiol 48(1):35–44
37. Claus F, Boterberg T, Ost P, De Neve W (2002) Short term toxicity profile for 32 sinonasal
cancer patients treated with IMRT. Can we avoid dry eye syndrome? Radiother Oncol
64(2):205–208
38. Artificial tears—Mayo Clinic. Available from www.mayoclinic.org/diseases-conditions
39. Artificial tears—Medscape reference. Available from reference.medscape.com
40. Doughty MJ, Glavin S (2009 Nov 1) Efficacy of different dry eye treatments with artificial
tears or ocular lubricants: a systematic review. Ophthalmic Physiol Opt 29(6):573–583
41. Berdoulay A, English RV, Nadelstein B (2005) Effect of topical 0.02% tacrolimus aqueous
suspension on tear production in dogs with keratoconjunctivitis sicca. Vet Ophthalmol
8(4):225–232
42. Kaswan RL, Salisbury MA, Ward DA (1989) Spontaneous canine keratoconjunctivitis sicca.
A useful model for human keratoconjunctivitis sicca: treatment with cyclosporine eye drops.
Arch Ophthalmol 107(8):1210–1216
43. Papas AS, Sherrer YS, Charney M, Golden HE, Medsger TA Jr, Walsh BT, Trivedi M, Goldlust
B, Gallagher SC (2004) Successful treatment of dry mouth and dry eye symptoms in Sjögren’s
syndrome patients with oral pilocarpine: a randomized, placebo-controlled, dose-adjustment
study. J Clin Rheumatol 10(4):169–177
44. Vivino FB, Al-Hashimi I, Khan Z, LeVeque FG, Salisbury PL 3rd, Tran-Johnson TK,
Muscoplat CC, Trivedi M, Goldlust B, Gallagher SC (1999) Pilocarpine tablets for the treat-
ment of dry mouth and dry eye symptoms in patients with Sjögren syndrome: a randomized,
placebo-controlled, fixed-dose, multicenter trial. P92-01 study group. Arch Intern Med
159(2):174–181
45. Colligris B, Alkozi HA, Pintor J (2014) Recent developments on dry eye disease treatment
compounds. Saudi J Ophthalmol 28(1):19–30
46. Quinto GG, Campos M, Behrens A (2008) Autologous serum for ocular surface diseases. Arq
Bras Oftalmol 71(6 Suppl):47–54
47. Ervin AM, Wojciechowski R, Schein O (2010) Punctal occlusion for dry eye syndrome.
Cochrane Database Syst Rev 9:CD006775. doi:10.1002/14651858.CD006775.pub2
48. Barabino S, Raghavan A, Loeffler J, Dana R (2005) Radiotherapy-induced ocular surface dis-
ease. Cornea 24:909–914
49. Weaver RD, Gerbi BJ, Dusenbery KE (1998) Evaluation of eye shields made of tungsten and
aluminum in high-energy electron beams. Int J Radiat Oncol Biol Phys 41:233–237
50. Shiu AS, Tung SS, Gastorf RJ et al (1996) Dosimetric evaluation of lead and tungsten eye
shields in electron beam treatment. Int J Radiat Oncol Biol Phys 35:599–604
51. Sodium chloride hypertonic, ophthalmic. Available from reference.medscape.com
52. Ocular lubricant ointments. Available from reference.medscape.com/
53. Wipperman JL, Dorsch JN (2013) Evaluation and management of corneal abrasions. Am Fam
Physician 87(2):114–120
54. Weaver CS, Terrell KM (2003) Evidence-based emergency medicine. Update: do ophthalmic
nonsteroidal anti-inflammatory drugs reduce the pain associated with simple corneal abrasion
without delaying healing? Ann Emerg Med 41(1):134–140
55. Rosenbaum J. Uveitis: etiology, clinical manifestation, and diagnosis. Available from www.
uptodate.com
Ear Toxicity
5

The acute ear complications of radiation therapy include external, middle, and inner
ear injuries. The acute external ear complications include otitis externa or skin reac-
tions involving the preauricular region, the auricle, and the external auditory canal
that occur in about 28% of head and neck cancer patients treated with radiation
therapy [1]. The acute middle ear complications that include mastoiditis, Eustachian
tube dysfunction, consequential otitis media, and transient conductive hearing loss
occur in 40–45% of head and neck cancer patients during or after radiation therapy
[2, 3]. The acute inner ear complications include sensorineural hearing loss (SNHL)
and tinnitus [4]. SNHL may occur early after treatment in up to 50% of patients with
head and neck tumors treated with radiation therapy [5–7].

5.1 Mechanism

The external ear is a tube covered by skin that conducts sound waves to the middle
ear. Radiation effects on the external ear mimics the skin effects of radiation and for
mechanism, timing, and symptoms (see Chap. 1).
The mechanism of radiation-induced otitis media is due to swelling of the mucosa
following the radiation and subsequent obstruction of the Eustachian tube. The
Eustachian tube obstruction results in resorption of the air oxygen and nitrogen from the
middle ear, which results in negative pressure and tympanic retraction leading to con-
ductive hearing loss. With further reduction of the negative pressure in the middle ear
cavity, fluid transudation takes place resulting in serous otitis media [4, 8].
Stria vascularis in the inner ear is responsible for endolymph production and
absorption. Stria vascularis injury after radiation exposure leads to endolymphatic
hydrops and temporarily increased intralabyrinthine pressure, which could lead to
ear fullness, hearing loss, tinnitus, dizziness, and vertigo [9].
Hearing loss may be conductive, sensorineural, or mixed type. Early hearing loss
during radiation therapy is usually conductive due to Eustachian tube dysfunction
and radiation-induced otitis media. The incidence of SNHL increases with time, and

© Springer International Publishing AG 2017 47


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_5
48 5 Ear Toxicity

sensorineural or mixed-type hearing loss occurs either near the end of or shortly
after the completion of radiation therapy. The sensory component of early hearing
loss is usually related to stria vascularis degeneration and disturbances in normal
perilymph and endolymph physiology [9, 10].

5.2 Timing

Acute otitis media usually occurs within a few weeks of radiation therapy. It is usually
transient [1] and resolves within a few weeks after completion of the treatment [4].
SNHL may occur near the end of treatment or after the completion of radiation
therapy and increase with time. SNHL may be transient or permanent. Transient
SNHL could recover within 6–12 months; however, it may last over 12 months in a
few cases [5].
Patients with no severe SNHL (less than 30 dB drop from baseline) and no post-
irradiation serous otitis media have a good chance for recovery of sensorineural
hearing within 6 months to 1 year. On the other hand, if severe SNHL develops or
the SNHL persists beyond 1 year, it is likely permanent [5].

5.3 Risk Factors

No specific risk factor has been defined for radiation-induced ear toxicities. As with
other organs, the maximum dose and ear irradiated volume could affect the preva-
lence and severity of toxicities. Background skin disease such as lupus erythema-
tous and other autoimmune disease may cause more toxicity for the external ear.
Previous middle ear disease such as otitis media or mastoiditis may also increase
middle ear radiation-induced toxicities. Concomitant administration of chemother-
apy may also increase the risk and severity of otitis in external ear (Fig. 5.1).
Platinum-based regimens are used as induction or concurrent chemotherapy
in locally advanced head and neck cancers and can potentially affect inner ear toxic-
ity so the risk of SNHL may increase.

Fig. 5.1 External otitis in


a patient treated with
cisplatin-based
chemoradiation of head
and neck cancer
5.5 Scoring 49

Fig. 5.2 Otitis media after


chemoradiation for parotid
tumors

5.4 Symptoms and Diagnosis

Symptoms of otitis media are conductive hearing loss, ear pain, fullness, and tinnitus in
the affected ear [4]. As a result of radiation therapy, the tympanic membrane may become
dull, retracted, bulged, and congested or may remain normal (Fig. 5.2) [3]. An inner ear
injury can manifest with tinnitus, dizziness, vertigo, and high-­frequency SNHL [5].
Diagnosis of these complications is based on patient symptoms and ear examina-
tion including otoscopic assessment. Hearing loss may be diagnosed by simple tests
like general assessment of each ear with words spoken at various volumes or a tun-
ing fork or by more thorough audiometry testing. A pure tone audiometer test offers
an audiogram based on a person’s ability to hear sounds with different loudness and
pitches and characterizes the type and degree of hearing loss.

5.5 Scoring

No distinct grading system has been published for external, middle, and inner ear
radiation-induced toxicities separately. Radiation Therapy Oncology Group (RTOG)
and the European Organization for Research and Treatment of Cancer (EORTC)
have defined the ear toxicity grading system, which considers all three parts as an
organ (see Table 5.1) [11].

Table 5.1 RTOG/EORTC ear toxicity scoring


Definition
Grade 1 Mild external otitis with erythema, pruritus, secondary to dry desquamation not
requiring medication. Audiogram unchanged from baseline
Grade 2 Moderate external otitis requiring topical medication/serous otitis media/hypoacusis
on testing only
Grade 3 Severe external otitis with discharge or moist desquamation/symptomatic hypoacusis/
tinnitus not drug related
Grade 4 Deafness
50 5 Ear Toxicity

5.6 Management

All patients that are undergoing radiation therapy to a portion of the temporal bone
or any ear compartment should have pretreatment audiometric evaluation (pure tone
audiometry, otoacoustic emission audiometry, tympanometry) [12]. Patients should
be assessed again after the end of treatment or sooner, if the clinical situation war-
rants [13, 14], and then audiometric evaluation should be repeated based on patient
symptoms at each visit.
All patients should be informed to maintain aural hygiene. Any traumatic actions
to the ear canal like the use of a curette for cerumen removal should be avoided. The
routine use of agents such as mineral oil or carbamide peroxide supplemented, as
needed, with gentle irrigation is recommended to prevent cerumen impaction [13].
External otitis is a skin reaction to radiation exposure that commonly occurs dur-
ing radiation therapy and resolves after the completion of treatment [9]. External
otitis symptoms can be alleviated with over-the-counter analgesics and topical ear
drops in our experience; however, there is no study evaluating the use of different
topical medications in treatment of this complication during radiation therapy.
Corticosteroids (betamethasone or dexamethasone otic solutions) decrease
inflammation and relieve ear pain [15]. Corticosteroids could be used 2–4 drops in
the affected ear canal every 2 or 3 h. Patients should tilt the head to the affected ear,
instill the drops, and keep this position for about 5 min [15].
Acetic acid drop changes the pH of the ear canal [16] and has antibacterial and
antifungal properties [17]. The patient should be instructed to use acetic acid 4–6
drops every 2–3 h into the external auditory canal [17].
The combination of the hydrocortisone and acetic acid is available in the form of
otic solution that can be used in patients with external otitis [16].
Perforated tympanic membrane is a contraindication to the use of any medication
in the external ear canal.
Otitis media is generally treated symptomatically with analgesics and antipyretics.
Antipyrine and benzocaine otic solution is a combination solution containing
antipyrine, benzocaine, oxyquinoline sulfate, and anhydrous glycerin that is applied
as a local anesthetic for otitis media [18]. It can be used every 1–2 h as needed for
pain relief with a sufficient amount of solution to fill the affected ear canal [19].
Aqueous lidocaine 2% can provide rapid pain relief in acute otitis media.
Aqueous lidocaine is not available in a bottle with a dropper. Injectable lidocaine
2% can be used with a dropper [20].
Oral analgesics (e.g., NSAIDs, acetaminophen) can reduce acute otitis media
pain much slower than topical medication [21]. Oral analgesics may be used in
combination with topical agents such as lidocaine and benzocaine [18].
Middle ear ventilation tube insertion may be needed in the setting of long-lasting
radiation-induced secretory otitis media (3–6 months) [22].
References 51

References
1. Bhandare N, Antonelli PJ, Morris CG, Malayapa RS, Mendenhall WM (2007) Ototoxicity
after radiotherapy for head and neck tumors. Int J Radiat Oncol Biol Phys 67(2):469–479
2. Jereczek-Fossa BA, Zarowski A, Milani F, Orecchia R (2003) Radiotherapy-induced ear toxic-
ity. Cancer Treat Rev 29(5):417–430
3. Upadhya I, Jariwala N, Datar J (2011) Ototoxic effects of irradiation. Indian J Otolaryngol
Head Neck Surg 63(2):151–154
4. Borsanyi SJ, Blanchard CL (1962) Ionizing radiation and the ear. JAMA 181(11):958–961
5. Kwong DL, Wei WI, Sham JS, Ho W, Yuen P, Chua DT et al (1996) Sensorineural hearing loss
in patients treated for nasopharyngeal carcinoma: a prospective study of the effect of radiation
and cisplatin treatment. Int J Radiat Oncol Biol Phys 36(2):281–289
6. Malgonde MS, Nagpure P, Kumar M (2015) Audiometric patterns in ototoxicity after radio-
therapy and chemotherapy in patients of head and neck cancers. Indian J Palliat Care 21(2):164
7. Yilmaz YF, Aytas FI, Akdogan O, Sari K, Savas ZG, Titiz A et al (2008) Sensorineural hearing
loss after radiotherapy for head and neck tumors: a prospective study of the effect of radiation.
Otol Neurotol 29(4):461–463
8. Bhide S, Harrington K, Nutting C (2007) Otological toxicity after postoperative radiotherapy
for parotid tumours. Clin Oncol 19(1):77–82
9. Linskey ME, Johnstone PA (2003) Radiation tolerance of normal temporal bone structures:
implications for gamma knife stereotactic radiosurgery. Int J Radiat Oncol Biol Phys
57(1):196–200
10. Singh J, Jaiwardhan G, Yadav S, Gulia J, Bhisnoi S (2014) Effect of radiotherapy on hearing
thresholds in patients of head and neck malignancies. Int J Otorhinolaryngol 16(1)1–6. https://
print.ispub.com/api/0/ispub-article/14729
11. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
12. Saluja M, Thakur J, Azad R, Sharma D, Mohindroo N, Seam R et al (2014) Radiotherapy
induced hearing loss in head neck cancers: screening with DPOAE. Head Neck Oncol 6(3):3
13. O’Neill JV, Katz AH, Skolnik EM (1979) Otologic complications of radiation therapy.
Otolaryngol Head Neck Surg 87(3):359–363
14. Raaijmakers E, Engelen AM (2002) Is sensorineural hearing loss a possible side effect of
nasopharyngeal and parotid irradiation? A systematic review of the literature. Radiother Oncol
65(1):1–7
15. Betamethasone otic [Internet]. Available from www.drugs.com
16. Otitis externa medication [Internet]. Available from emedicine.medscape.com
17. Acetic acid [Internet]. Available from www.drugs.com
18. 19-Antipyrine and benzocaine ear drops [Internet]. Available from www.drugs.com
19. Antipyrine and benzocaine otic medical facts from drugs [Internet]. Available from ­https://
www.drugs.com
20. Prasad S, Ewigman B (2008) Use anesthetic drops to relieve acute otitis media pain. J Family
Pract 57(6):370
21. Bertin L, Pons G, d’Athis P, Duhamel J, Maudelonde C, Lasfargues G et al (1996) A random-
ized, double-blind, multicentre controlled trial of ibuprofen versus acetaminophen and placebo
for symptoms of acute otitis media in children. Fundam Clin Pharmacol 10(4):387–392
22. Anteunis LJ, Wanders SL, Hendriks JJ, Langendijk JA, Manni JJ, de Jong JMA (1994)
Prospective longitudinal study on radiation-induced hearing loss. Am J Surg 168(5):408–411
Oral Mucositis
6

Almost no patients that are undergoing radiation therapy of head and neck cancers
can run away from radiation-induced oral mucositis, especially if the oral cavity is
included in the treatment target. Trotti et al. have confirmed the high incidence of
induced oral mucositis in a systematic review with rates of 97% during conventional
radiation therapy, 100% during altered fractionation radiation therapy, and 89%
during chemoradiation therapy [1]. In a retrospective study of 204 head and neck
cancer patients that received radiation therapy with or without chemotherapy, oral
mucositis occurred in 91% of patients; the rates of mucositis grades 1, 2, 3, and 4
were 4%, 21%, 60%, and 6%, respectively [2].
Another study of 450 head and neck cancer patients that received radiation ther-
apy with or without chemotherapy found that the majority of patients (83%) devel-
oped oral mucositis (mild in 19%, moderate in 35%, and severe in 28% of patients).
Oral mucositis has significant pain, dysgeusia, and odynophagia that can result
in dehydration, malnutrition, and reduced quality of life scores.
Oral mucositis also can reduce radiation therapy tolerance and consequently
affect treatment results. Unplanned breaks/delays in radiation therapy were reported
in 2.4%, 15.8%, and 46.8% of patients with mild, moderate, and severe oral muco-
sitis, respectively [3].

6.1 Mechanism

The oral mucosa is covered by stratified squamous epithelium. The basal layer of
mucosal epithelium has columnar cells with rapid division properties to maintain a
constant epithelial population as cells are shed from the surface [4]. The lamina
propria underlies the epithelium, which consists of fibroblasts and connective tis-
sue, capillaries, inflammatory cells (macrophages), and extracellular matrix [4, 5].
Radiation-induced oral mucositis is not a simple epithelial process and results from

© Springer International Publishing AG 2017 53


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_6
54 6 Oral Mucositis

complex pathways embracing all different cellular and tissue compartments of the
mucosa. It has been proposed that endothelial and connective tissue damage pre-
cedes epithelial changes in irradiated oral mucosa [5].
Radiation directly damages cellular DNA of rapidly dividing cells of the oral
basal epithelium and cells in the underlying tissue [6]. Radiation also generates oxi-
dative stress and reactive oxygen species (ROS) that lead to further tissue damage by
activating a number of transcription factors such as nuclear factor-κB (NF-­κB) in the
epithelium, endothelium, macrophages, and mesenchymal cells. Subsequently, the
upregulation of genes and the production of pro-inflammatory cytokines including
TNF-α, IL-1β, and IL-6 occur, leading to an injury to the connective tissue, endothe-
lium, and apoptosis of cells within the basal epithelium. Pro-­inflammatory cytokines
also activate molecular pathways that amplify production of TNF-α, IL-1, and IL-6
and resulted in further tissue damage (positive feedback loop).
Until this stage, the clinical appearance of mucosa seems to be normal, and only
the thinning of the mucosa and edema related to acute vascular response may
develop and cause early signs and symptoms of mucositis (mild erythema and burn-
ing sensation). But as radiation therapy continues, all events from the prior phase
result in further injuries and loss of the basal epithelial stem cells with continuing
cell loss from the mucosal surface, reducing the cellular population of the mucous
membrane. Atrophic changes and breakdown of mucosa occur and patients experi-
ence obvious symptoms of mucositis [6–12].
The ulcerative phase of mucositis is accompanied by significant inflammatory
cell infiltration. Beside the intrinsic factors, this phase may be affected by extrinsic
factors that have reciprocal relation together. Damaged epithelium of the ulcerative
mucositis is susceptible to bacterial colonization. Following the colonization with a
mixed microbial flora, including mostly gram-negative bacteria, bacterial cell wall
products penetrate the injured mucosa and increasingly stimulate pro-inflammatory
cytokine release, amplifying the severity of mucositis and tissue injury [6]. When
radiation therapy is completed, tissue repair begins with renewal of epithelial prolif-
eration and differentiation and reestablishment of the local microbial flora [13].

6.2 Timing

In conventional fractioned radiotherapy (2 Gy per fraction daily), mucosal erythema


with mild discomfort occurs within 1–2 weeks of treatment at doses of approxi-
mately 10–20 Gy [13]. Patchy pseudomembranous formation can develop after
2 weeks (20 Gy), and ulcerative radiation-induced mucositis often arises at doses of
more than 30 Gy and peaks during the fourth to fifth week of therapy. This timing
of symptom presentation is varied based on treatment schedule. In accelerated radi-
ation therapy, mucositis reaches its peak within 3 weeks [11, 14]. The symptoms
usually become more severe with treatment and remain for about 2–3 weeks after
the end of therapy at its peak [2, 15]. Healing then begins and may take weeks to
months (generally 3–6 weeks) to resolve [16]. However, chronic open wounds rec-
ognized as soft-tissue necrosis may occur in a few cases due to excessive depletion
of mucosal stem cells depending on their recovery [17].
6.3 Risk Factors 55

6.3 Risk Factors

Several factors have been identified for the development of more severe oral muco-
sitis during radiation therapy. These factors could be related to treatment, tumor,
and/or patient characteristics.

6.3.1 Tumor Site

Primary tumors of the oral cavity, oropharynx, or nasopharynx [16] increase the risk
of oral mucositis due to including higher volumes of the oral mucosa and salivary
gland in irradiation fields.
In the treatment planning of oral cavity, oropharyngeal, and nasopharyngeal can-
cer, a significant surface of oral mucosa includes in radiation field that increase the
rate of oral mucositis. In the oral cavity, some areas, including the lateral borders and
ventral surface of the tongue as well as the soft palate and floor of the mouth, have
increased susceptibility to developing oral mucositis due to higher cell turnover
rates. Instead, buccal mucosa has less sensitivity to radiation-induced mucositis [13].
Salivary gland radiation-induced injury leads to changes in quality (low production of
glycoproteins and an increased acidity of saliva) and quantity of saliva (see Chap. 7) that
inhibit the protective effects of saliva and increases the risk of developing mucositis [18].

6.3.2 Concomitant Systemic Therapy

When chemotherapy is combined with radiation therapy, both normal tissue and
tumor response alter due to their additive effects. Both radiation therapy and anti-
neoplastic agents disrupt normal cell division of the oral mucosa and result in
increased oral mucositis incidence [13, 19, 20]. Dose, type, and schedule of sys-
temic therapy all affect severity and frequency of mucositis in the setting of concur-
rent chemoradiotherapy (weekly versus every 3 weeks; cisplatin or paclitaxel are
associated with higher rates of oral mucositis) [21, 22].
There is a paucity of data about oral mucositis rates when cetuximab is used concur-
rently with radiation therapy. Some proposed no exacerbation of the common mucosi-
tis associated with radiation therapy [23, 24], and others found an increase of oral
mucositis compared to radiation alone or conventional cisplatin with radiation therapy
[25–27]. Future clinical trials are needed to more precisely evaluate the incidence and
severity of mucositis, the time of occurrence, and the impact on quality of life and
treatment interruption of patients treated with cetuximab and radiation therapy.

6.3.3 Radiation Dose

Higher total radiation dose to the oral cavity and higher dose per fraction are signifi-
cantly correlated to the grade of acute mucositis. It has been reported that patients
with cancer of the larynx, hypopharynx, oral cavity, nasopharynx, or oropharynx
56 6 Oral Mucositis

that are treated with radiation therapy with cumulative radiation dosage more than
50 Gy are at an increased risk for oral mucositis [3, 16].
With the introduction of high conformal radiation therapy, the correlation
between the dose to the oral cavity and the severity of acute mucositis has been
evaluated more precisely.
In a study of patients undergoing intensity modulated radiation therapy (IMRT)
for head and neck cancer, it was demonstrated that a cumulative point dose of 39 Gy
resulted in mucositis for 3 weeks or longer; mild severity and short duration of
mucositis were found at cumulative point doses less than 32 Gy [28]. Another study
found that the percentage of oral cavity volume receiving doses higher than 15, 30,
40, 45, and 50 Gy significantly correlated with acute mucositis grade [29].

6.3.4 Radiation Fractionation Schedule

Altered fractionation radiation schedules, such as concomitant boost or


hyperfractionation seem to be associated with higher incidence of severe muco-
sitis [15, 30–34].

6.3.5 Patient-Related Factors

The incidence of mucositis is related to various patient variables including younger


patients, smoking, alcohol consumption, metallic dental restorations, preexisting
periodontal disease, low body mass index, poor functional status, low leukocyte
count, advanced disease and stage, a prior history of severe mucositis, and various
comorbid conditions [3, 35–37].
It has been suggested that with increasing number of patient-related factors, the
risk, duration, and severity of mucositis increases (see below).
Some argue that younger cancer patients have a higher rate of mucosal turnover
that increases their susceptibility to mucositis [38].
The use of dental guards is proposed in the areas of metallic restoration because
it reduces back scatter exacerbated radiation doses to adjacent mucosa caused by
metallic restoration materials [13].
Genetic polymorphisms may be a susceptibility factor for radiation-induced
mucositis (XRCC1 polymorphisms, NBN polymorphisms) [39, 40]. It has been seen
that cytokine phenotype may be correlated to the risk of radiation-induced injury.
During radiation therapy, an elevation in serum levels of cytokines IL-6, TNF-α, and
IL-1β and low levels of IL-8 seem to correlate with mucositis severity [35, 41–43].
A weak correlation between high pretreatment epidermal growth factor (EGF) lev-
els and decreased severity of mucositis has been seen that is suggestive of the protective
EGF effect for oral mucosa damage [44]. Further study is needed to clarify cytokines
and growth factors role in predicting, preventing and treating oral mucositis.
Suresh et al. evaluated a collection of patient-related risk factors and proposed a
comprehensive tool to predict the probable incidence and severity of mucositis in
head and neck cancer patients receiving chemoradiotherapy. In this suggested
6.4 Symptoms 57

system, patients are scored based on age > 40, erythrocyte sedimentation rate (ESR)
>3-times the upper limit, albumin <3.0 g/dL, white blood count (WBC) less than
3000/μL, Eastern cooperative oncology group (ECOG) performance status (PS) of
more than 2, stage III or higher disease, use of tobacco, and presence of any comor-
bid conditions. One point is given to each of these parameters. Scores of 3 or less
and 6 or above predicted the differences in the incidence of mucositis [45].

6.4 Symptoms

The early signs of radiation mucositis are red appearance of the oral mucosa due to
dilation of capillaries in the endothelial layer and reduced epithelial thickness. A
whitish appearance may be seen due to transient hyperkeratinization prior to ery-
thema. Patients are mostly asymptomatic or complain of a mild burning sensation
or intolerance to spices or hot food in this early phase. Erythema (Fig. 6.1) is fol-
lowed by fibrinous pseudomembranous formation, followed by erosion and ulcer-
ation (Fig. 6.2). Under the pseudomembranes, the epithelial surfaces are denuded,
and hemorrhage occurs easily. Pseudomembranous ulcerative lesions are very

Fig. 6.1 Grade 1 of oral


mucositis at second week
(14 Gy) of radiation
therapy

Fig. 6.2 Grade 2–3 of oral


mucositis at fifth week
(48 Gy) of radiation
therapy
58 6 Oral Mucositis

painful, and patients complain of severe pain and difficulty in chewing that inter-
feres with their oral intake or speaking, eventually leading to weight loss.
Oral pain follows a similar pattern of objective clinical findings of oral mucositis
but it may begin sooner and reach its peak earlier (between weeks 2 and 4) [2].
Correlation between patient-reported and clinical manifestations of mucositis is low
in early parts of treatment [46].
Bacterial infection (usually gram-negative) or viral infections (such as herpes
simplex virus (HSV)) and fungal infections (usually candidiasis) can sometimes be
superimposed on oral mucositis [13, 37]. Infectious mucosal lesions often extend
beyond the field of radiation. An infected oral mucosa usually manifests with a deep
ulceration with a yellow-white necrotic center and raised borders. Fungal mucositis
presents with white removable fungal plaques [13]; however, erythematous forms
may occur and complicate the exact diagnosis [47].

6.5 Scoring

Patients undergoing radiation therapy to the oral cavity should be seen at least once
a week. At each visit, patient symptoms and their oral intake should be assessed;
their oral mucosa should also be examined.
A variety of assessment scales exist for measurement of oral mucositis. Three of
the most commonly utilized scales (Table 6.1) for radiation-induced mucositis are
toxicity criteria of the Radiation Therapy Oncology Group (RTOG), the European
Organization for Research and Treatment of Cancer (EORTC) [48], the National
Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) [49],
and the World Health Organization (WHO) oral toxicity scale [50].

Table 6.1 RTOG, WHO, and CTCAE mucositis grading


RTOG WHO CTCAE
Grade 1 Irritation/may experience mild Mild Asymptomatic or mild
pain not requiring analgesic Soreness ± erythema symptoms; intervention
not indicated
Grade 2 Patchy mucositis that may Moderate Moderate pain; not
produce an inflammatory Erythema, ulcers; interfering with oral
serosanguinous discharge/may patient can swallow intake; modified diet
experience moderate pain solid food indicated
requiring analgesia
Grade 3 Confluent fibrinous mucositis/ Severe Severe pain; interfering
may include severe pain requiring Ulcers with with oral intake
narcotic extensive erythema;
patient cannot
swallow food
Grade 4 Ulceration, hemorrhage, or Life-threatening
necrosis consequences; urgent
intervention indicate
Grade 5 Death
6.6 Prevention 59

6.6 Prevention

Maintenance of good oral hygiene is critical to preventing oral mucositis. All


patients should be informed of proper oral hygiene before starting treatment:

–– Cleaning teeth with toothpaste and a soft toothbrush after each meal and at bed-
time [51], using a mild-tasting toothpaste or a solution of 1 teaspoon of salt
added to 4 cups (1 quart) of water (if not tolerating any toothpaste) [19].
–– Dental flossing once daily.
–– Dental screening at least several weeks before the beginning of therapy extrac-
tions and major surgeries should be planned 10–21 days and 4–6 weeks before
beginning radiation therapy, respectively [52].
–– Rinsing the mouth two to four times a day (1 tablespoon (15 mL) of oral rinse,
swish in oral cavity for 30 s, then spit out).
Recommended oral rinses [51]:
• Water
• Sodium chloride 0.9% for irrigation
• Salt solution by adding a little salt (1/4 to 1/2 a teaspoon) to a cup of warm
water
–– Be aware that alcohol-based mouth rinses should be avoided.
–– Moisten lips with a moisturizing cream.
–– Adequate oral fluid intake (daily fluid intake of 8–12 cups, 2–3 L).
–– Avoiding alcohol, tobacco, caffeine, fluid or foods with high sugar, highly acidic
fluids and foods (e.g., lemon glycerin swabs, vitamin C lozenges), hot and spicy
foods, and crunchy foods.

6.6.1 Prophylactic Interventions

Benzydamine is a nonsteroidal anti-inflammatory drug with local analgesic, anes-


thetic, and antimicrobial properties that inhibits pro-inflammatory cytokines includ-
ing IL1 and TNF-α. It has been shown to reduce the severity and frequency of
mucositis in patients with head and neck cancer undergoing radiation therapy.
Benzydamine oral rinse appears to be effective in the prevention of radiation-­
induced oral mucositis in head and neck cancer patients [53–58]. Benzydamine
(15 mL of oral rinse 0.15%) is used as a mouth rinse. It should be held or rinse
within the mouth for at least 30 s, followed by expulsion from the mouth and try not
to swallow, 3–4 times per day. It is recommended to begin the day prior to radiation
therapy and continue during treatment.
Benzydamine is well tolerated. If patients complain of numbness or irritation or
a burning sensation as result of benzydamine usage, diluting the liquid with an equal
amount of warm water helps alleviate this issue. Other side effects are nausea and
60 6 Oral Mucositis

vomiting, xerostomia, headache, cough, and pharyngeal irritation. It can be absorbed


through the oral mucosa and should be used with caution in patients with renal
impairment.

6.6.2 Prophylactic Intervention Under Evaluation

Intensity-modulated radiation therapy (IMRT) allows sparing a greater area of the


oral mucosa from higher doses of radiation. However, an obvious superiority of
IMRT to conventional three-dimensional conformal radiotherapy (3D–CRT) and
conventional two-dimensional radiotherapy (2DRT) in the prevention of the acute
oral mucositis is not established [59, 60].
Low-level laser therapy seems to be effective in the prophylaxis of radiation-­
induced oral mucositis [61–66]. As a noninvasive measurement, it can decrease the
incidence and severity of oral mucositis [67]. Laser therapy modulates three main
biological effects: an analgesic effect with increase in endorphin release and pain
signal inhibition, an anti-inflammatory and wound healing properties with energy
production at the mitochondrial level, and an increase in vascularity and regenera-
tion of injured tissues [2, 63, 68]. It refers to local application of a high-density
monochromatic narrow-band light source with the output power range from 5 to
500 milliwatts and a wave-length between 600 and 1000 nanometers [69]. It is a
simple procedure that is administered immediately after radiation therapy and takes
only about 6–8 min to administer. All is done extraorally unless intraoral lesions
develop [47].
Glutamine is a nonessential amino acid in the body. Glutathione, a byproduct of
glutamine metabolism, protects normal tissues against oxidative injury [70, 71].
Glutamine depletion may develop in cancer patients due to cachexia and normal
tissue damage from radiation or chemotherapy [72].
It has been found that oral glutamine administered daily during radiation therapy
can reduce the severity and frequency of oral mucositis in head and neck cancer
patients. These results warrant further investigation in future trials [72–77].
Zinc renders multiple essential functions in the body such as cell proliferation,
wound healing, free radical protection, immunity, and anti-inflammatory effects. It
has been shown that oral zinc sulfate can prevent and cause delay in development of
oral mucositis [78].
Payayor (Clinacanthus nutans) is a traditional herbal medicine originating from
Thailand with anti-inflammatory and analgesic properties. There are some studies
that support payayor as a prophylactic intervention for radiation-induced oral muco-
sitis [79, 80]. However, no guideline for use exists currently due to insufficient
evidence.
Calendula officinalis, commonly known as Marigold, possesses some anti-­
inflammatory and antioxidant properties. It has been suggested that Calendula offi-
cinalis can be effective in decreasing oral mucositis severity [81, 82].
Natural honey has been recently shown to be effective in reducing radiation-­
induced mucositis. Honey has immunomodulatory properties and antibacterial
6.6 Prevention 61

activity and can accelerate wound healing. Honey is an easily available agent that
warrants further trials to validate its effect [83–85].
Royal jelly is a secretion of hypopharyngeal and mandibular glands of worker
bees with antioxidative, antibiotic, and anti-inflammatory action. Royal jelly could
improve the signs and symptoms of oral mucositis and shorten healing time [86].
Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been reported
to regulate proliferation and maturation of leukocytes, macrophages, and dendritic
cells. GM-CSF can improve wound healing by enhancing fibroblasts and keratino-
cyte growth [87].
Subcutaneous GM-CSF has been shown to decrease pain and severity of oral
mucositis [88, 89]. However, topical administration of GM-CSF to treat radiation-­
induced oral mucositis has mixed results [87, 90, 91]. Saarilahti et al. studied topical
use of GM-CSF and reported promising effects in prophylaxis of radiation-induced
oral mucositis [92]. There are also conflicting data about effectiveness of subcutane-
ous GM-CSF in radiation-induced mucositis prophylaxis [93, 94].
Keratinocyte growth factor (KGF) is an epithelial cell growth factor expressed by
fibroblasts and endothelial cells and has an important role in wound healing by
increasing proliferation and maintaining integrity of epithelial cells and enhancing
neovascularization and collagen deposition [95].
Primary results show that recombinant human keratinocyte growth factor
Palifermin resulted in reduction of radiation mucositis with no stimulation of the
proliferation of tumor cell lines [96–98], which needs further evaluation.
Oral recombinant human epidermal growth factor (rhEGF) has been shown to
improve wound repair by stimulating epithelialization. rhEGF appears to be effec-
tive for the treatment of radiation-induced mucositis [99, 100]. Another growth fac-
tor with promising results in preclinical study is velafermin (recombinant human
fibroblast growth factor-20, rhFGF-20) [101]. Further studies are needed to deter-
mine these agents’ efficacy and safety for prevention and treatment of radiation-­
induced mucositis.
Amifostine is a thiol compound that selectively protects normal tissue from radia-
tion effects [102]. Amifostine decreases the frequency and severity of xerostomia. The
salivary preservation by amifostine may offer a protective effect against oral mucositis
[17]. However, the amifostine studies for the prevention of oral mucositis offer insuf-
ficient evidence to support its use for this purpose. Additional investigation is needed
to clarify the role of amifostine as an intervention for oral mucositis prevention [103].
Fluconazole (100 mg/daily) in head and neck cancer patients receiving radiation
therapy can lead to decreased candida carriage and incidence of severe mucositis
[5, 104–107]. Patients that are receiving head and neck radiation therapy are at
increased risk of developing oral candida infection (17–29%) and colonization
(93%) [108].
PTA is a multi-agent lozenge containing a mixture of polymyxin E, tobramycin,
and amphotericin B and has a broad spectrum of antibacterial and antifungal effects.
It has been used to prevent radiation-induced mucositis with inconclusive results.
Results of the topical antibiotic approach in prevention of oral mucositis are insuf-
ficient and need to be further studied [109–114].
62 6 Oral Mucositis

Chlorhexidine antimicrobial oral rinse is not recommended for the prevention of


radiation-induced oral mucositis. It has no clinical benefit for the reduction or pre-
vention of radiation-induced oral mucositis [18]. Chlorhexidine oral rinse is not
well tolerated and may cause a degree of discomfort (e.g., taste alteration, burning
sensation and teeth staining) without any clinical benefit [115].
Sucralfate is an oral ulcer coating complex of sucrose-sulfate-aluminum salt.
Clinical trials found no significant advantage for sucralfate in the prevention or
treatment of radiation-induced mucositis [116–120].
Prostaglandins are known to be cytoprotective. Misoprostol is a synthetic prosta-
glandin E1 analogue. Misoprostol mouthwash has been studied to prevent oral
mucositis in head and neck cancer patients, and the data does not demonstrate a
prevention benefit [121, 122]. There are limited studies for prostaglandin E2 appli-
cation in the radiation-induced mucositis prevention and treatment that do not draw
any conclusions.
Steroids have several effects on different systems of the human body and have
been studied for radiation-induced oral mucositis prevention. Currently, there is
insufficient evidence to support using systemic steroids for reducing the frequency
or severity of oral mucositis [47].
Early morning radiation delivery has been shown to marginally reduce the sever-
ity of mucositis because of circadian variations in cell cycle proteins. The most
radiosensitive phase of the cell cycle (G2-M) occurs in the late afternoon and eve-
ning in human oral mucosa [123, 124]. Currently, there is no recommendation for
this observation in clinical practice [125].
Pilocarpine has not shown any preventive effect on oral mucositis during radia-
tion therapy in head and neck cancer patients [125].
Supplemental antioxidants during radiation therapy have been proposed to pro-
tect the normal tissue and reduce side effects caused by radiation therapy. However,
these antioxidants may act unselectively and protect cancer cells against the damag-
ing effects of reactive oxygen species induced by radiation. Antioxidant supplement
safety during radiation therapy is particularly controversial. The American Cancer
Society and most other national nutrition guidelines recommend that patients with
active cancer treatments limit the usage of supplements to obvious deficiency of
essential agents [126].
RK-0202 (RxKinetix), an oral rinse, comprises the potent antioxidant
N-acetylcysteine in a polymer matrix. RK-0202 has demonstrated primarily posi-
tive results in reducing the incidence of severe mucositis in patients treated with
radiation therapy for head and neck cancer [127].
Vasoconstrictor agents such as phenylephrine with transient vasoconstriction,
tissue hypoxia, and suppression of mucosal cell death during irradiation would pre-
vent radiation-induced oral mucositis based on preclinical studies [128]. Further
clinical trials should be conducted to prove the preventive effect of these agents on
radiation-induced mucositis.
Caphosol (Cytogen Corp) is an electrolyte solution with high ionic content
(Ca2+ and PO43− ions) that help tissue repair by diffusing ions into the inter­cellular
6.7 Management 63

spaces in the epithelium, thus permeating the mucosal lesion and modulate the
inflammatory process. Recent studies did not find significant reduction in the inci-
dence or duration of severe oral mucositis in patients receiving head and neck radia-
tion therapy [129, 130].
Oxygen nebulization that uses high-flow oxygen could improve local mucosal
oxygen content leading to angiogenesis, anti-inflammatory effect, and improved
wound healing. It has been studied in patients with nasopharyngeal cancer to pre-
vent radiation-induced mucositis with promising results. Future studies are required
to better determine effectiveness [131].
Several herbal mouth rinses like Korean red ginseng [132], manuka (Leptospermum
scoparium) and kanuka (Kunzea ericoides) [133], or chamomile [134] have been
studied with a positive effect on the development of radiation-­induced mucositis.
Future investigation is needed to confirm the efficacy and safety of these products.
Persian traditional medicines have different compounds with various local and
systemic effects on the mucosal surface and can theoretically be used to reduce
mucositis [135]. One of the combinations (Malva sylvestris L and Alcea digitata
(Boiss) Alef), which is effective in reducing xerostomia [136], has also been evalu-
ated in a small randomized trial in our patients and primary results are promising
(not published yet).
The mammalian target of rapamycin (mTOR) inhibition plays a role in the pro-
tection of normal oral epithelial cells from radiation-induced epithelial stem cell
depletion. mTOR inhibition with rapamycin may have a potential effect on the pre-
vention of radiation-induced mucositis [137]. Clinical studies need to evaluate
rapamycin efficacy in this setting.
Mothers against decapentaplegic homolog 7 (SMAD7) is a protein encoded by the
SMAD7 gene and has an antagonistic effect on transforming growth factor beta (TGF-
β) and NF-κB signaling. Its prophylactic and therapeutic effects on radiation-­induced
oral mucositis as a well as its safety need to be determined in future studies [138].
Wobe-Mugos is a combination of proteolytic enzymes comprised of papain,
trypsin, and chymotrypsin that has an anti-inflammatory effect. It seems not to be
efficient in preventing radiation-induced oral mucositis [139].

6.7 Management

Principles of management consist of patient assessment, oral care, management of


oral pain, treatment of secondary infection, and consideration of nutritional
support.

6.7.1 Patient Assessment

All patients treated with radiation therapy should be seen as least once weekly,
and the oral mucosa should be examined at each visit. In the patients with oral
64 6 Oral Mucositis

mucositis, baseline grading of oral mucositis and the patient’s general status
should be determined. After initial assessment, decision-making about treatment
protocol can be provided. Most cases can be managed in an outpatient setting
except in the setting of grade 4 mucositis, fever (more than 38.3 °C), or severe
neutropenia. Patients with inadequate fluid intake may require oral supplementa-
tion or IV hydration. Complete blood count is proposed in patients that have
severe mucositis, fever, or at risk of developing neutropenia.

6.7.2 Mouth Care

Mouth care includes all measurements noted for maintaining good oral hygiene (see
above), with intensity and frequency modification based on mucositis grading [140].

6.7.3 Management of Oral Pain

Pain management is the most important aspect of symptom control. Systemic anal-
gesia may be prescribed in addition to topical agents in moderate to severe pain.
Pain control can encourage patients to eat and drink more and wash the mouth more
efficiently, resulting in improved medication effects.

6.7.3.1 Benzydamine Mouth Wash


Benzydamine can reduce the severity of oral mucositis and associated pain in
radiation-­induced oral mucositis [51, 141].
Benzydamine (15 mL of oral rinse 0. 15%) is used as a mouth rinse. It should be
held in the mouth for at least 30 s, followed by expulsion (should not be swallowed),
up to every 1–2 h.
Benzydamine is well tolerated. If patients complain of numbness or irritation or
a burning sensation, dilution with an equal amount of warm water may reduce these
symptoms. Other side effects are nausea and vomiting, xerostomia, headache,
cough, and pharyngeal irritation. It can be absorbed through the oral mucosa and
should be used with caution in patients with renal impairment.

6.7.3.2 Doxepin
Doxepin is a tricyclic antidepressant. Topical application is prescribed for pruritus
and neuropathic pain [142]. Topical doxepin rinse has been shown to be an adequate
analgesia for oral mucositis pain up to 4 h after application. Patients usually have
good compliance; however, mild burning or stinging, unpleasant taste, and drowsi-
ness could develop as common adverse effects [143].
Usage: oral rinse at a dosage of 25 mg (10 mg/mL × 2.5) diluted in 5 mL of water
for 1 min 3–6 times per day.
6.7 Management 65

6.7.3.3 Magic Mouthwash


Magic mouthwash usually contains at least three of these basic ingredients [144]:

–– An antibiotic
–– An antihistamine or local anesthetic
–– An antifungal
–– A corticosteroid
–– An antacid

Magic mouthwash is used every 4–6 h, maintained in the mouth for 1–2 min
before being either spit out or swallowed (in pharyngeal or esophageal involve-
ment). It’s recommended not to eat or drink for at least 30 min after using magic
mouthwash [144].
There are various formulations for magic mouthwash with no standard mixture.
Some of the more common formulations are defined here [145]:

• 80 mL viscous lidocaine 2%, 80 mL Mylanta, 80 mL diphenhydramine (12.5 mg


per 5 mL elixir), 80 mL nystatin 100,000 U suspension, 80 mL prednisolone
(15 mg per 5 mL solution), and 80 mL distilled water
• 1 part viscous lidocaine 2%, 1 part Maalox (do not substitute Kaopectate), and 1
part diphenhydramine (12.5 mg per 5 mL elixir)
• 30 mL viscous lidocaine 2%, 60 mL Maalox (do not substitute Kaopectate),
30 mL diphenhydramine (12.5 mg per 5 mL elixir), and 40 mL Carafate (1 gm
per 10 mL)
• 100 mL dexamethasone (0.5 mg per 5 mL elixir), 60 mL nystatin 100,000 U
suspension, 100 mL diphenhydramine (12.5 mg per 5 mL elixir), and 3 capsules
tetracycline 500 mg

Different contributors of magic mouthwash are added for different purposes:

Diphenhydramine provides local analgesia but can exacerbate xerostomia.


Lidocaine also provides local analgesia.
Corticosteroids reduce inflammatory responses.
Nystatin is added as an antifungal agent.
Tetracycline as an antibiotic (inhibits bacterial protein synthesis).
Antacid (magnesium hydroxide/aluminum hydroxide) component to adequately
coat the oral mucosa [54].

Side effects of magic mouthwash may include problems with taste, a burning or
tingling sensation in the mouth, drowsiness, constipation, diarrhea, and nausea.
Lidocaine can cause a gag reflex impairment and increase the risk of aspiration,
mouth numbness, and tongue biting.
66 6 Oral Mucositis

6.7.3.4 Gelclair
As an oral gel, Gelclair consists mainly of polyvinylpyrrolidone and sodium hyal-
uronate. It is indicated for the management of oral mucositis-related pain due to
establishing a barrier over the oral mucosa [142].
Usage: Oral gel, available in 15 mL single-use packet, is poured into a glass.
Then add up to two tablespoons or 40 mL of water, stir the mixture well and use
immediately. Rinse for at least 1 min and spit 3 times a day or as needed. Do not eat
or drink for at least 1 h following treatment; this may provide relief for up to 7 h.

6.7.3.5 CAM2028
CAM2028 is a bioadhesive barrier-forming lipid solution that can also act as a lipid-­
based drug carrier system for local and extended delivery of benzydamine. After
application, lipid components spread in the oral cavity and form a barrier that pro-
tects the sore and inflamed mucosa. Recently, it has been shown that CAM2028,
with benzydamine or without benzydamine, provides significant pain reduction for
patients suffering from oral mucositis. Its effect begins within 5 min of application
and is maintained for up to 8 h [146, 147].
Usage: Patients are instructed to spray a 0.15 mL-metered dose of the liquid into
the oral cavity 1–3 times and to allow 5 min for the bioadhesive barrier to form
before eating or drinking [146].

6.7.3.6 MF 5232


MF 5232 (Mucotrol) is a concentrated oral polyherbal gel wafer formulation with
local analgesic, antioxidant and immunomodulatory activity, and wound-healing
properties [148]. It has received FDA approval for pain relief associated with oral
lesions [149]. The efficacy of MF 5232 therapy in radiation-induced mucositis man-
agement has been reported [148]. Further studies are required to confirm these posi-
tive results.

6.7.3.7 Acetylcysteine
Acetylcysteine oral rinse, as a mucolytic agent to reduce the viscosity of mucous
secretions, is being studied to improve saliva thickness and painful mouth sores in
patients with head and neck cancer undergoing radiation therapy [150].

6.7.3.8 Opiates
Low-dose opiates may be used in patients with inadequate pain relief with nonnar-
cotic topical agents. Oral, transdermal, or parenteral opiates may be used. Elixir
should not be used because it contains alcohol, which exacerbates mucositis.
Constipation prophylactic laxatives should be considered in patients taking opi-
ates [47].
Fentanyl has various forms that all can be used in patients with oral mucositis
depending on patient preference. Transdermal fentanyl patch can deliver a steady-­
state dose through the skin and provide pain relief to cancer patients with less con-
stipation and sedation than morphine [151]. The level of drug increase over 12–24 h
and reach steady state in about 72 h. Oral transmucosal fentanyl is a sweetened
6.7 Management 67

matrix of fentanyl that is dissolved in the mouth. It is effective for breakthrough


pain and could be particularly useful in patients with mucositis. It has been demon-
strated that oral transmucosal fentanyl citrate may provide more efficacious treat-
ment options than morphine sulfate in treating breakthrough cancer pain [152, 153].
Fentanyl buccal tablet allows rapid absorption through the oral mucosa and pro-
duces a greater early systemic level than oral transmucosal fentanyl citrate. Fentanyl
buccal tablet can be absorbed with a similar profile in patients with or without mild
oral mucositis [154].
Mouthwashes with a morphine-containing solution can effectively decrease pain
in oral mucositis associated with radiation therapy compared to magic mouthwash.
More studies are required in this regard [155, 156].
Sublingual methadone has been proposed as an alternative of standard treat-
ment for mucositis-related pain. Additional studies need to better illuminate the
potential benefits of this formulation of methadone in radiation-induced mucosi-
tis [153].
Usage:
Morphine (opioid-naive patients): SC/IM; 5–10 mg q4hr PRN/IV; 2.5–5 mg
q3-4 hr. PRN/oral; 15–30 mg PO q4hr PRN; 10–20 mg PR q4hr.
Fentanyl transdermal patch (12.5 mcg/hr., 25 mcg/hr., 50 mcg/hr., 75 mcg/hr.,
100 mcg/hr): 25–100 mcg/hr., reapplied q72hr (or every 48 hr. administration may
be needed in a few cases).
Initial patch is selected based on patient need for daily PO morphine dose, then
decrease by 25–50%.
Oxycodone: 10 mg PO q12hr initially; increase by 25–50% gradually every
1–2 days, q12hr dosing interval should be maintained.
Methadone: 2.5 mg PO q8-12 hr.; increase slowly, then every 3–5 days.

6.7.3.9 Corticosteroids
Corticosteroids are useful [51] and are usually administered as a component of a
magic solution. Hydrocortisone tablet (2.5 mg buccal tablets) can be used orally.
Patients are instructed to dissolve the tablet slowly in mouth in contact with the
ulcer three times daily.

6.7.3.10 Gabapentin
Gabapentin is an antiepileptic drug. It is an effective agent in the treatment of neu-
ropathic pain. It also has an application for pain relief that is related to radiation-­
induced mucositis that seems to be successful [157].
Gabapentin begins at a dose of 600 mg and can gradually increase to 1800–
3600 mg/day.

6.7.3.11 Other Analgesic Agents


Carmellose sodium paste [52]
Lidocaine 1% gel [52]
Choline salicylate dental gel [52]
Kaolin-pectin [158]
68 6 Oral Mucositis

Codeine/acetaminophen (1–2 tablets dissolved in a little water) as a mouthwash


up to four times daily [52]
Aspirin 300 mg (1–2 tablets dissolved in a little water) as a mouthwash up to four
times daily [52].

6.7.3.12 Sucralfate
Sucralfate mouthwash is not recommended to be used to treat oral mucositis in
patients receiving radiation therapy for head and neck cancer.

6.7.3.13 Phenytoin Mouthwash


Phenytoin mouthwash was compared with normal saline in patients with radiation-­
induced mucositis in a pilot study. Phenytoin mouthwash (1% solution) was more
effective for pain control without significant efficacy on mucositis severity [159].

6.7.3.14 Low-Level Laser Therapy


Low-level laser therapy is a promising treatment therapy of oral mucositis that has
been shown to reduce mucositis pain significantly [47, 160].

6.7.4 Antifungals

As was previously mentioned in Sect. 7.4, patients undergoing head and neck radia-
tion therapy are at risk of developing candidiasis [108]. Fungal mucositis presents
with white removable fungal plaques [13]; however, erythematous form may occur
and complicate exact diagnosis [47]. The diagnosis is usually based on clinical sus-
picious and can be readily confirmed through identification of yeast forms on gram
stain or KOH preparation of the scrapings [161].
Patients with early, mild presentation are treated with topical agents including nystatin
or clotrimazole. Patients with moderate to severe candidiasis or recurrent disease are
treated with more potent systemic agents including fluconazole and itraconazole [161].
Clotrimazole troches (10 mg oral troches): Dissolve 1 troche in the oral cavity 5
times a day for 14 days [161] (not tolerated in patients with xerostomia).
Nystatin oral suspension (100,000 U/mL): rinse mouth with 5 mL (400,000–
6,000,000 units) four times a day. Swish in mouth and retain for 2 min, and then
swallow or expectorate [161]. Continue treatment for at least 48 h after perioral
symptoms have disappeared.
Fluconazole: 200 mg PO loading dose then 100–200 mg daily for 1–2 weeks [162]
Itraconazole oral solution: 200 mg daily for 1–2 weeks [162]

6.7.5 Antivirals

The incidence of herpes simplex virus-1 (HSV-1) infection has been estimated to be
29% in patients with mucositis during head and neck cancer radiation therapy,
which is isolated from smears taken from patients with ulcerative mucositis [162].
The lesions present primarily as mucosal vesicles, which rupture spontaneously and
References 69

form ulcerative lesions. The early vesicular stage may be so rapid that it is not seen
in immunocompromised patients [163].
Patients with oral mucositis that experience a prolonged course of mucositis with
extreme pain or vesicular lesions, secondary HSV infection should be considered.
The gold standard diagnostic test for HSV-related oral mucositis is isolation of HSV
tissue culture (results usually take 2–7 days). A more rapid diagnostic test for muco-
cutaneous lesions uses staining of skin scrapings.
Treatment with oral or intravenous acyclovir or oral valacyclovir decreases the
duration of viral shedding and improves the severity of oral mucositis.

Oral acyclovir: 400 mg three times a day for 7–10 days (five times a day in immu-
nocompromised patients) [163]
Intravenous acyclovir (If oral intolerance develops): 5 mg/kg three times a day
[163, 164]
Oral valacyclovir: 1 g three times a day for 7–10 days [164]

6.7.6 Feeding Tube/Nutritional Support

In patients with severe symptoms of radiation mucositis, oral nutritional is compro-


mised and can lead to weight loss and nutritional depletion. Nutritional support and
feeding tubes should be considered in these patients [3, 165].
For short-term access, nasogastric and nasojejunal tubes may be used; for longer-­
term access, gastrostomy, gastrojejunostomy, and jejunostomy can be placed.
Percutaneous endoscopic gastrostomy is the method of choice unless in the setting
of esophagitis, gastroparesis, aspiration pneumonia, or limited gastric volume that
jejunostomy tube may be placed instead [166].
In patients without a functioning gastrointestinal tract due to obstruction, intrac-
table vomiting, diarrhea, poor GI motility, short bowel syndrome or severe pancre-
atitis, and total parenteral nutrition may be appropriate [166].

References
1. Trotti A, Bellm LA, Epstein JB, Frame D, Fuchs HJ, Gwede CK et al (2003) Mucositis inci-
dence, severity and associated outcomes in patients with head and neck cancer receiving
radiotherapy with or without chemotherapy: a systematic literature review. Radiother Oncol
66(3):253–262
2. Elting LS, Keefe DM, Sonis ST, Garden AS, Spijkervet F, Barasch A et al (2008) Patient-­
reported measurements of oral mucositis in head and neck cancer patients treated with radio-
therapy with or without chemotherapy. Cancer 113(10):2704–2713
3. Vera-Llonch M, Oster G, Hagiwara M, Sonis S (2006) Oral mucositis in patients undergoing
radiation treatment for head and neck carcinoma. Cancer 106(2):329–336
4. Squier CA, Kremer MJ (2000) Biology of oral mucosa and esophagus. J Natl Cancer Inst
Monogr 29:7–15
5. Sonis ST (2004) The pathobiology of mucositis. Nat Rev Cancer 4(4):277–284
6. Kumar PS, Balan A, Sankar A, Bose T (2009) Radiation induced oral mucositis. Indian
J Palliat Care 15(2):95
70 6 Oral Mucositis

7. Logan RM, Gibson RJ, Sonis ST, Keefe DM (2007) Nuclear factor-κB (NF-κB) and cycloox-
ygenase-­2 (COX-2) expression in the oral mucosa following cancer chemotherapy. Oral
Oncol 43(4):395–401
8. Gibson R, Bowen J, Cummins A, Logan R, Healey T, Keefe D (2004) Ultrastructural changes
occur early within the oral mucosa following cancer chemotherapy [abstract A-373]. Supp
Care Cancer 12(6):389
9. Denham JW, Hauer-Jensen M (2002) The radiotherapeutic injury—a complex ‘wound’.
Radiother Oncol 63(2):129–145
10. Sonis ST, Elting LS, Keefe D, Peterson DE, Schubert M, Hauer-Jensen M et al (2004)
Perspectives on cancer therapy-induced mucosal injury. Cancer 100(S9):1995–2025
11. Baker DG (1982) The radiobiological basis for tissue reactions in the oral cavity following
therapeutic x-irradiation: a review. Arch Otolaryngol 108(1):21–24
12. Biswal BM (2008) Current trends in the management of oral mucositis related to cancer treat-
ment. Malays J Med Sci 15(3):4
13. Shih A, Miaskowski C, Dodd MJ, Stotts NA, MacPhail L (2003) Mechanisms for radiation-­
induced oral mucositis and the consequences. Cancer Nurs 26(3):222–229
14. Köstler WJ, Hejna M, Wenzel C, Zielinski CC (2001) Oral mucositis complicating chemo-
therapy and/or radiotherapy: options for prevention and treatment. CA Cancer J Clin
51(5):290–315
15. Hilderley L (1997) Principles of teletherapy. In: Hassey-Dow K, Bucholtz J (eds) Nursing
care in radiation oncology, 2nd edn. Saunders, Philadelphia, PA, pp 6–20
16. Lalla RV, Sonis ST, Peterson DE (2008) Management of oral mucositis in patients who have
cancer. Dent Clin N Am 52(1):61–77
17. Rosenthal DI, Trotti A (2009) Strategies for managing radiation-induced mucositis in head
and neck cancer. Semin Radiat Oncol 19:29–34
18. Peterson D, Bensadoun R-J, Roila F, Group EGW (2011) Management of oral and gastroin-
testinal mucositis: ESMO clinical practice guidelines. Ann Oncol 22(suppl 6):vi78–vi84
19. Supportive P, Board PCE. Oral complications of chemotherapy and head/neck radiation
(PDQ®). 2016.
20. Seiwert TY, Salama JK, Vokes EE (2007) The chemoradiation paradigm in head and neck
cancer. Nat Clin Pract Oncol 4(3):156–171
21. Hoffmann W, Belka C, Schmidberger H, Budach W, Bochtler H, Hess CF et al (1997)
Radiotherapy and concomitant weekly 1-hour infusion of paclitaxel in the treatment of
head and neck cancer—results from a Phase I trial. Int J Rad Oncol Biol Phys 38(4):
691–696
22. Tsan D-L, Lin C-Y, Kang C-J, Huang S-F, Fan K-H, Liao C-T et al (2012) The comparison
between weekly and three-weekly cisplatin delivered concurrently with radiotherapy for
patients with postoperative high-risk squamous cell carcinoma of the oral cavity. Radiat
Oncol 7(1):1
23. Musio D, De Felice F, Bulzonetti N, Tombolini V (2013) Cetuximab and oral mucositis: is it
different from oral mucositis caused by other drugs? Otolaryngology 2013.
doi:10.4172/2161-119X.1000147
24. Mehra R, Cohen RB, Burtness BA (2008) The role of cetuximab for the treatment of squa-
mous cell carcinoma of the head and neck. Clin Adv Hematol Oncol 6(10):742
25. Walsh L, Gillham C, Dunne M, Fraser I, Hollywood D, Armstrong J et al (2011) Toxicity of
cetuximab versus cisplatin concurrent with radiotherapy in locally advanced head and neck
squamous cell cancer (LAHNSCC). Radiother Oncol 98(1):38–41
26. Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB et al (2006) Radiotherapy
plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med
354(6):567–578
27. Pryor DI, Porceddu SV, Burmeister BH, Guminski A, Thomson DB, Shepherdson K et al
(2009) Enhanced toxicity with concurrent cetuximab and radiotherapy in head and neck can-
cer. Radiother Oncol 90(2):172–176
References 71

28. Narayan S, Lehmann J, Coleman MA, Vaughan A, Yang CC, Enepekides D et al (2008)
Prospective evaluation to establish a dose response for clinical oral mucositis in patients under-
going head-and-neck conformal radiotherapy. Int J Rad Oncol Biol Phys 72(3):756–762
29. Shogan J, Bhatnagar A, Heron D, Smith R, Andrede R, Huq M et al (2005) Dosimetric cor-
relation of oral cavity dose with acute mucositis in patients treated with intensity modulated
radiation therapy (IMRT) and chemotherapy. Int J Radiat Oncol Biol Phys 63:S74–SS5
30. Budach W, Hehr T, Budach V, Belka C, Dietz K (2006) A meta-analysis of hyperfractionated
and accelerated radiotherapy and combined chemotherapy and radiotherapy regimens in unre-
sected locally advanced squamous cell carcinoma of the head and neck. BMC Cancer 6:28
31. Russo G, Haddad R, Posner M, Machtay M (2008) Radiation treatment breaks and ulcerative
mucositis in head and neck cancer. Oncologist 13(8):886–898
32. Fu KK et al (1993) A phase III randomized study to compare twice daily hyperfractionation,
accelerated hyperfractionation with concomitant boost to standard fractionation radiotherapy
for squamous cell carcinomas of the head and neck. Data on file, Radiation Treatment
Oncology Group RTOG (90–03). Philadelphia
33. Horiot J-C, Le Fur R, N’guyen T, Chenal C, Schraub S, Alfonsi S et al (1992) Hyperfractionation
versus conventional fractionation in oropharyngeal carcinoma: final analysis of a randomized
trial of the EORTC cooperative group of radiotherapy. Radiother Oncol 25(4):231–241
34. Mariani S (2001) Management of radiation-induced toxicity in patients with head and neck
cancer. Medscape Hematology-Oncology eJournal 4(2)
35. Meirovitz A, Kuten M, Billan S, Abdah-Bortnyak R, Sharon A, Peretz T et al (2010)
Cytokines levels, severity of acute mucositis and the need of PEG tube installation during
chemo-radiation for head and neck cancer-a prospective pilot study. Radiat Oncol 5(1):1
36. Naidu MUR, Ramana GV, Rani PU, Suman A, Roy P (2004) Chemotherapy-induced and/or
radiation therapy-induced oral mucositis-complicating the treatment of cancer. Neoplasia
6(5):423–431
37. Sonis ST (2013) Oral mucositis in head and neck cancer: risk, biology, and management. Am
Soc Clin Oncol Educ Book doi:10.1200/EdBook_AM.2013.33.e236
38. Miyamoto M (2007) Reducing the incidence and severity of oral mucositis—can it be done?
US Oncological Disease 1(2):18–21
39. Ren J-H, Dai X-F, Yan G-L, Jin M, Liu C-W, Yang K-Y et al (2014) Acute oral mucositis in
nasopharyngeal carcinoma patients treated with radiotherapy: association with genetic poly-
morphism in DNA DSB repair genes. Int J Radiat Biol 90(3):256–261
40. Venkatesh GH, Manjunath VB, Mumbrekar KD, Negi H, Fernandes DJ, Sharan K et al
(2014) Polymorphisms in radio-responsive genes and its association with acute toxicity
among head and neck cancer patients. PLoS One 9(3):e89079
41. Xanthinaki A, Nicolatou-Galitis O, Athanassiadou P, Gonidi M, Kouloulias V, Sotiropoulou-­
Lontou A, Pissakas G, Kyprianou K, Kouvaris J, Patsouris E (2008) Apoptotic and inflamma-
tion markers in oral mucositis in head and neck cancer patients receiving radiotherapy:
preliminary report. Supp Care Cancer 16:1025–1033
42. Akmansu M, Unsal D, Bora H, Elbeg S (2005) Influence of locoregional radiation treatment
on tumor necrosis factor-alpha and interleukin-6 in the serum of patients with head and neck
cancer. Cytokine 31:41–45
43. Haddad R, Sonis S, Posner M, Wirth L, Costello R, Braschayko P et al (2009) Randomized
phase 2 study of concomitant chemoradiotherapy using weekly carboplatin/paclitaxel with or
without daily subcutaneous amifostine in patients with locally advanced head and neck can-
cer. Cancer 115(19):4514–4523
44. Epstein JB, Gorsky M, Guglietta A, Le N, Sonis ST (2000) The correlation between epider-
mal growth factor levels in saliva and the severity of oral mucositis during oropharyngeal
radiation therapy. Cancer 89(11):2258–2265
45. Suresh A, Varma PP, Sinha S, Deepika S, Raman R, Srinivasan M et al (2010) Risk-scoring
system for predicting mucositis in patients of head and neck cancer receiving concurrent
chemoradiotherapy [rssm-hn]. J Cancer Res Ther 6(4):448
72 6 Oral Mucositis

46. Gussgard AM, Jokstad A, Hope AJ, Wood R, Tenenbaum H (2015) Radiation induced muco-
sitis in patients with head and neck cancer–should the signs or the symptoms be measured?
J Can Dent Assoc 81:f11
47. Soto EF-DJ, DeVita BA (2015) Hellman, and Rosenberg’s cancer: principles & practice of
oncology, 10th edn. Lippincott Williams & Wilkins, Philadelphia
48. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
49. NCI N (2009) DHHS common terminology criteria for adverse events v4. 0. National Cancer
Institute, Bethesda, MD
50. Organization WH. WHO handbook for reporting results of cancer treatment. 1979.
51. Taylor S (2014) Guidelines for prevention and management of oral mucositis. West Sussex
and Hampshire Cancer Network NHS, Surrey
52. Devi S, Singh N (2014) Dental care during and after radiotherapy in head and neck cancer.
Nat J Maxillofac Surg 5(2):117
53. Epstein JB, Silverman S, Paggiarino DA, Crockett S, Schubert MM, Senzer NN et al (2001)
Benzydamine HCl for prophylaxis of radiation-induced oral mucositis. Cancer 92(4):875–885
54. Lalla RV, Schubert MM, Bensadoun R-J, Keefe D (2006) Anti-inflammatory agents in the
management of alimentary mucositis. Supp Care Cancer 14(6):558–565
55. Kazemian A, Kamian S, Aghili M, Hashemi F, Haddad P (2009) Benzydamine for prophy-
laxis of radiation-induced oral mucositis in head and neck cancers: a double-blind placebo-­
controlled randomized clinical trial. Eur J Cancer Care 18(2):174–178
56. Sheibani KM, Mafi AR, Moghaddam S, Taslimi F, Amiran A, Ameri A (2015) Efficacy of
benzydamine oral rinse in prevention and management of radiation-induced oral mucositis: a
double-blind placebo-controlled randomized clinical trial. Asia Pac J Clin Oncol 11(1):22–27
57. Kin-Fong CK, Ka TYJ (2005) A pilot study of chlorhexidine and benzydamine oral rinses for
the prevention and treatment of irradiation mucositis in patients with head and neck cancer.
Cancer Nurs 29(5):423–430
58. Hanna EY (2002) Prevention of radiation-induced oral mucositis with benzydamine. Curr
Oncol Rep 4(1):65–67
59. Kouloulias V, Thalassinou S, Platoni K, Zygogianni A, Kouvaris J, Antypas C et al (2013)
The treatment outcome and radiation-induced toxicity for patients with head and neck carci-
noma in the IMRT era: a systematic review with dosimetric and clinical parameters. Biomed
Res Int 2013:401261
60. Khuntia D, Harris J, Bentzen S, Kies M, Meyers J, Foote R et al (2008) Increased oral muco-
sitis after IMRT versus non-IMRT when combined with cetuximab and cisplatin or docetaxel
for head and neck cancer: preliminary results of RTOG 0234. Int J Radiat Oncol Biol Phys
72(1):S33
61. Antunes HS, Herchenhorn D, Small IA, Araújo CM, Viégas CMP, Cabral E et al (2013) Phase
III trial of low-level laser therapy to prevent oral mucositis in head and neck cancer patients
treated with concurrent chemoradiation. Radiother Oncol 109(2):297–302
62. Genot M-T, Klastersky J (2005) Low-level laser for prevention and therapy of oral mucositis
induced by chemotherapy or radiotherapy. Curr Opin Oncol 17(3):236–240
63. Jadaud E, Bensadoun R (2012) Low-level laser therapy: a standard of supportive care for
cancer therapy-induced oral mucositis in head and neck cancer patients? Laser Ther 21(4):297
64. Antunes H, Herchenhorn D, Araujo C, Cabral E, Ferreira EdS, Small I, et al (2011) Phase III
trial of low-level laser therapy to prevent induced oral mucositis in head and neck cancer
patients submitted to concurrent chemoradiation. ASCO Annual Meeting Proceedings
65. Figueiredo ALP, Lins L, Cattony AC, Falcão AFP (2013) Laser therapy in oral mucositis
control: a meta-analysis. Rev Assoc Med Bras 59(5):467–474
66. Lopes CO, Mas JRI, Zângaro RA (2006) Low level laser therapy in the prevention of
radiotherapy-­induced xerostomia and oral mucositis. Radiol Bras 39(2):131–136
67. Arbabi-Kalati F, Arbabi-Kalati F, Moridi T (2013) Evaluation of the effect of low level laser
on prevention of chemotherapy-induced mucositis. Acta Med Iran 51(3):157
References 73

68. Tezel A, Kara C, Balkaya V, Orbak R (2009) An evaluation of different treatments for recur-
rent aphthous stomatitis and patient perceptions: Nd: YAG laser versus medication. Photomed
Laser Surg 27(1):101–106
69. Barrett S (2009) A skeptical look at low level laser therapy. Quackwatch Retrieved 2010–07,23
70. Rouse K, Nwokedi E, Woodliff JE, Epstein J, Klimberg VS (1995) Glutamine enhances
selectivity of chemotherapy through changes in glutathione metabolism. Ann Surg 221(4):420
71. Savarese DM, Savy G, Vahdat L, Wischmeyer PE, Corey B (2003) Prevention of chemo-
therapy and radiation toxicity with glutamine. Cancer Treat Rev 29(6):501–513
72. Chattopadhyay S, Saha A, Azam M, Mukherjee A, Sur PK (2014) Role of oral glutamine in
alleviation and prevention of radiation-induced oral mucositis: a prospective randomized
study. South Asian J Cancer 3(1):8
73. Silverman S Jr (2007) Diagnosis and management of oral mucositis. J Support Oncol 5(2
Suppl 1):13–21
74. Tsujimoto T, Yamamoto Y, Wasa M, Takenaka Y, Nakahara S, Takagi T et al (2015) L-glutamine
decreases the severity of mucositis induced by chemoradiotherapy in patients with locally
advanced head and neck cancer: a double-blind, randomized, placebo-controlled trial. Oncol
Rep 33(1):33–39
75. Vidal-Casariego A, Calleja-Fernández A, Ballesteros-Pomar MD, Cano-Rodríguez I (2013)
Efficacy of glutamine in the prevention of oral mucositis and acute radiation-induced esopha-
gitis: a retrospective study. Nutr Cancer 65(3):424–429
76. Huang E-Y, Leung SW, Wang C-J, Chen H-C, Sun L-M, Fang F-M et al (2000) Oral gluta-
mine to alleviate radiation-induced oral mucositis: a pilot randomized trial. Int J Radiat Oncol
Biol Phys 46(3):535–539
77. Brian D, Lawenda MD (2013) Use glutamine to reduce the severity of mucositis and neu-
ropathy (during chemotherapy or radiation therapy). Integrat Oncol Essent 25
78. Moslemi D, Babaee N, Damavandi M, Pourghasem M, Moghadamnia A (2014) Oral zinc
sulphate and prevention of radiation-induced oropharyngealmucositis in patients with head
and neck cancers: a double blind, randomized controlled clinical trial. Int J Radiat Res
12(3):235–241
79. Putwatana P, Sanmanowong P, Oonprasertpong L, Junda T, Pitiporn S, Narkwong L (2009)
Relief of radiation-induced oral mucositis in head and neck cancer. Cancer Nurs 32(1):
82–87
80. Putwatana P (ed) (2007) Comparison of glycerine payayor and benzydamine in prevention
and relief of oral mucositis among patients with head and neck cancer receiving radiotherapy.
The 18th International Nursing Research Congress focusing on evidence-based practice
81. Babaee N, Moslemi D, Khalilpour M, Vejdani F, Moghadamnia Y, Bijani A et al (2013)
Antioxidant capacity of Calendula officinalis flowers extract and prevention of radiation
induced oropharyngeal mucositis in patients with head and neck cancers: a randomized con-
trolled clinical study. DARU J Pharmaceut Sci 21(1):1
82. Babaee N, Moslemi D, Khalilpour M, Vejdani F, Bijani A, Moghadamnia AA, et al (2013)
Investigation of the effect of Calendula officinalis extract on preventing radiotherapy-induced
oral mucositis. Reports of Radiotherapy and Oncology 1(1)
83. Maiti P, Ray A, Mitra T, Jana U, Bhattacharya J, Ganguly S (2012) The effect of honey on
mucositis induced by chemoradiation in head and neck cancer. J Indian Med Assoc
110(7):453–456
84. Richards D (2012) Evidence to support the use of honey for prevention of oral mucositis in
cancer patients is limited. Evid Based Dent 13(3):74
85. Hawley P, Hovan A, McGahan CE, Saunders D (2014) A randomized placebo-controlled trial
of manuka honey for radiation-induced oral mucositis. Supp Care Cancer 22(3):751–761
86. Erdem Ö, Güngörmüs Z (2014) The effect of royal jelly on oral mucositis in patients under-
going radiotherapy and chemotherapy. Holist Nurs Pract 28(4):242–246
87. Raoul J-L, Cadre B, Le Prisé E, Boucher E (2003) Local injections of granulocyte-­macrophage
colony-stimulating factor (Gm-CSF) for the treatment of radiation-induced mucosa ulcers.
Radiother Oncol 68(3):303–304
74 6 Oral Mucositis

88. Masucci G, Broman P, Kelly C, Lindahl S, Malmberg L, Reizenstein J et al (2005) Therapeutic


efficacy by recombinant human granulocyte/monocyte-colony stimulating factor on mucosi-
tis occurring in patients with oral and oropharynx tumors treated with curative radiotherapy.
Med Oncol 22(3):247–256
89. Wagner W, Alfrink M, Haus U, Matt J (1998) Treatment of irradiation-induced mucositis
with growth factors (rhGM-CSF) in patients with head and neck cancer. Anticancer Res
19(1B):799–803
90. Nicolatou O, Sotiropoulou-Lontou A, Skarlatos J, Kyprianou K, Kolitsi G, Dardoufas K
(1998) A pilot study of the effect of granulocyte–macrophage colony–stimulating factor on
oral mucositis in head and neck cancer patients during x-radiation therapy: a preliminary
report. Int J Radiat Oncol Biol Phys 42(3):551–556
91. Rovirosa A, Ferre J, Biete A (1998) Granulocyte macrophage–colony–stimulating factor
mouthwashes heal oral ulcers during head and neck radiotherapy. Int J Radiat Oncol Biol
Phys 41(4):747–754
92. Saarilahti K, Kajanti M, Joensuu T, Kouri M, Joensuu H (2002) Comparison of granulocyte-­
macrophage colony-stimulating factor and sucralfate mouthwashes in the prevention of
radiation-­induced mucositis: a double-blind prospective randomized phase III study. Int
J Radiat Oncol Biol Phys 54(2):479–485
93. McAleese J, Bishop K, A’Hern R, Henk J (2014) Randomized phase II study of GM-CSF to
reduce mucositis caused by accelerated radiotherapy of laryngeal cancer. Br J Radiol
74:608–613
94. Hoffman KE, Pugh SL, James JL, Scarantino C, Movsas B, Valicenti RK et al (2014) The
impact of concurrent granulocyte–macrophage colony-stimulating factor on quality of life in
head and neck cancer patients: results of the randomized, placebo-controlled radiation ther-
apy oncology group 9901 trial. Qual Life Res 23(6):1841–1858
95. Barasch A, Epstein J, Tilashalski K (2009) Palifermin for management of treatment-induced
oral mucositis in cancer patients. Biologics 3:111–116
96. Brizel D, Herman T, Goffinet D, Sailer S, Agarwala S, Schwartz G et al (2001) A phase I/II
trial of escalating doses of recombinant human keratinocyte growth factor (rHuKGF) in head
& neck cancer (HNC) patients receiving radiotherapy (RT) with concurrent chemotherapy
(CCT). Int J Radiat Oncol Biol Phys 51(3):40
97. Brizel DM, Murphy BA, Rosenthal DI, Pandya KJ, Glück S, Brizel HE et al (2008) Phase II
study of palifermin and concurrent chemoradiation in head and neck squamous cell carci-
noma. J Clin Oncol 26(15):2489–2496
98. Ning S, Shui C, Khan WB, Benson W, Lacey DL, Knox SJ (1998) Effects of keratinocyte
growth factor on the proliferation and radiation survival of human squamous cell carcinoma
cell lines in vitro and in vivo. Int J Radiat Oncol Biol Phys 40(1):177–187
99. Hong J, LEE SW, Song S, Ahn S, Shin S, Choi E et al (2009) Recombinant human epidermal
growth factor treatment of radiation-induced severe oral mucositis in patients with head and
neck malignancies. Eur J Cancer Care 18(6):636–641
100. Lee S-w, Jung KI, Kim YW, Jung HD, Kim HS, Hong JP (2007) Effect of epidermal growth
factor against radiotherapy-induced oral mucositis in rats. Int J Radiat Oncol Biol Phys
67(4):1172–1178
101. Ara G, Watkins BA, Zhong H, Hawthorne TR, Karkaria CE, Sonis ST et al (2008) Velafermin
(rhFGF-20) reduces the severity and duration of hamster cheek pouch mucositis induced by
fractionated radiation. Int J Radiat Biol 84(5):401–412
102. Kouvaris JR, Kouloulias VE, Vlahos LJ (2007) Amifostine: the first selective-target and
broad-spectrum radioprotector. Oncologist 12(6):738–747
103. Nicolatou-Galitis O, Sarri T, Bowen J, Di Palma M, Kouloulias VE, Niscola P et al (2013)
Systematic review of amifostine for the management of oral mucositis in cancer patients.
Supp Care Cancer 21(1):357–364
104. Nicolatou-Galitis O, Velegraki A, Sotiropoulou-Lontou A, Dardoufas K, Kouloulias V,
Kyprianou K et al (2006) Effect of fluconazole antifungal prophylaxis on oral mucositis in
head and neck cancer patients receiving radiotherapy. Supp Care Cancer 14(1):44–51
References 75

105. Mücke R, Kaben U, Libera T, Knauerhase H, Ziegler P, Hamann D et al (1996) Use of fluco-
nazole as antimycotic prophylaxis in radiotherapy of patients with head and neck tumors.
Mycoses 40:53–55
106. Gava A, Ferrarese F, Tonetto V, Coghetto F, Marazzato G, Zorat P (1996) Can the prophylac-
tic treatment of mycotic mucositis improve the time of performing radiotherapy in head and
neck tumors? Radiol Med 91(4):452–455
107. Koc M, Aktas E (2003) Prophylactic treatment of mycotic mucositis in radiotherapy of
patients with head and neck cancers. Jpn J Clin Oncol 33(2):57–60
108. Jham BC, da Silva Freire AR (2006) Oral complications of radiotherapy in the head and neck.
Rev Bras Otorrinolaringol 72(5):704–708
109. Sharma A, Rath G, Chaudhary S, Thakar A, Mohanti BK, Bahadur S (2012) Lactobacillus
brevis CD2 lozenges reduce radiation-and chemotherapy-induced mucositis in patients with
head and neck cancer: a randomized double-blind placebo-controlled study. Eur J Cancer
48(6):875–881
110. Satheeshkumar P, Chamba MS, Balan A, Sreelatha K, Bhatathiri V, Bose T (2010)
Effectiveness of triclosan in the management of radiation-induced oral mucositis: a random-
ized clinical trial. J Cancer Res Ther 6(4):466
111. Symonds R, McIlroy P, Khorrami J, Paul J, Pyper E, Alcock S et al (1996) The reduction of
radiation mucositis by selective decontamination antibiotic pastilles: a placebo-controlled
double-blind trial. Br J Cancer 74(2):312
112. Spijkervet FK, Van Saene HK, Van Saene JJ, Panders AK, Vermey A, Mehta DM et al (1991)
Effect of selective elimination of the oral flora on mucositis in irradiated head and neck can-
cer patients. J Surg Oncol 46(3):167–173
113. Okuno SH, Foote RL, Loprinzi CL, Gulavita S, Sloan JA, Earle J et al (1997) A randomized
trial of a nonabsorbable antibiotic lozenge given to alleviate radiation-induced mucositis.
Cancer 79(11):2193–2199
114. Oguchi M, Shikama N, Sasaki S, Gomi K, Katsuyama Y, Ohta S et al (1998) Mucosa-adhesive
water-soluble polymer film for treatment of acute radiation-induced oral mucositis. Int
J Radiat Oncol Biol Phys 40(5):1033–1037
115. Foote RL, Loprinzi CL, Frank AR, O’Fallon JR, Gulavita S, Tewfik HH et al (1994)
Randomized trial of a chlorhexidine mouthwash for alleviation of radiation-induced mucosi-
tis. J Clin Oncol 12(12):2630–2633
116. Makkonen TA, Boström P, Vilja P, Joensuu H (1994) Sucralfate mouth washing in the preven-
tion of radiation-induced mucositis: a placebo-controlled double-blind randomized study. Int
J Radiat Oncol Biol Phys 30(1):177–182
117. Franzén L, Henriksson R, Littbrand B, Zackrisson B (1995) Effects of sucralfate on mucositis
during and following radiotherapy of malignancies in the head and neck region: a double-­
blind placebo-controlled study. Acta Oncol 34(2):219–223
118. Lievens Y, Haustermans K, Van den Weyngaert D, Van den Bogaert W, Scalliet P, Hutse­
baut L et al (1998) Does sucralfate reduce the acute side-effects in head and neck cancer
treated with radiotherapy? A double-blind randomized trial. Radiother Oncol 47(2):
149–153
119. Carter DL, Hebert ME, Smink K, Leopold KA, Clough RL, Brizel DM (1999) Double blind
randomized trial of sucralfate vs placebo during radical radiotherapy for head and neck can-
cers. Head Neck 21(8):760–766
120. Etiz D, Erkal H, Serin M, Küçük B, Heparı A, Elhan A et al (2000) Clinical and histopatho-
logical evaluation of sucralfate in prevention of oral mucositis induced by radiation therapy
in patients with head and neck malignancies. Oral Oncol 36(1):116–120
121. Nicolatou-Galitis O, Sarri T, Bowen J, Di Palma M, Kouloulias VE, Niscola P et al (2013)
Systematic review of anti-inflammatory agents for the management of oral mucositis in can-
cer patients. Supp Care Cancer 21(11):3179–3189
122. Veness M, Foroudi F, Gebski V, Timms I, Sathiyaseelan Y, Cakir B et al (2006) Use of topical
misoprostol to reduce radiation-induced mucositis: results of a randomized, double-blind,
placebo-controlled trial. Australas Radiol 50(5):468–474
76 6 Oral Mucositis

123. Goyal M, Shukla P, Gupta D, Bisht SS, Dhawan A, Gupta S et al (2009) Oral mucositis in
morning vs. evening irradiated patients: a randomised prospective study. Int J Radiat Biol
85(6):504–509
124. Bjarnason GA, MacKenzie RG, Nabid A, Hodson ID, El-Sayed S, Grimard L et al (2009)
Comparison of toxicity associated with early morning versus late afternoon radiotherapy in
patients with head-and-neck cancer: a prospective randomized trial of the National Cancer
Institute of Canada clinical trials group (HN3). Int J Radiat Oncol Biol Phys 73(1):166–172
125. Jensen SB, Jarvis V, Zadik Y, Barasch A, Ariyawardana A, Hovan A et al (2013) Systematic
review of miscellaneous agents for the management of oral mucositis in cancer patients. Supp
Care Cancer 21(11):3223–3232
126. Cassileth B, Yarett I (2012) Antioxidant supplementation in patients with cancer: is it safe and
effective? The ASCO Post 3:15
127. Chambers M, Welsh D, Scrimger R, Zehn W, Epstein J, Troha J, et al (eds) (2006) RK-0202
for radiation-induced oral mucositis. ASCO Annual Meeting Proceedings
128. Soref CM, Fahl WE (2014) A new topical vasoconstrictor–based strategy for prevention of
oral mucositis. Oral Surg Oral Med Oral Pathol Oral Radiol 117(4):454–461
129. Wong KH, Kuciejewska A, Sharabiani MT, Ng-Cheng-Hin B, Hoy S, Hurley T et al (2017)
A randomised controlled trial of Caphosol mouthwash in management of radiation-induced
mucositis in head and neck cancer. Radiother Oncol 122:207–211
130. Rao NG, Trotti A, Kim J, Schell MJ, Zhao X, Amdur RJ et al (2014) Phase II multicenter trial
of Caphosol for the reduction of mucositis in patients receiving radiation therapy for head and
neck cancer. Oral Oncol 50(8):765–769
131. Xu J, Yan R, Zhuo P-Y, Li R-R, Ge H-X, Lu W-F (2014) Effectiveness of oxygen nebulization
at preventing radiotherapy-induced mucositis in patients with nasopharyngeal cancer. Int
J Nurs Sci 1(2):176–179
132. Chang JW, Choi JW, Lee BH, Park JK, Shin YS, Oh Y-T et al (2014) Protective effects of
Korean red ginseng on radiation-induced oral mucositis in a preclinical rat model. Nutr
Cancer 66(3):400–407
133. Maddocks-Jennings W, Wilkinson JM, Cavanagh HM, Shillington D (2009) Evaluating the
effects of the essential oils Leptospermum scoparium (manuka) and Kunzea ericoides
(kanuka) on radiotherapy induced mucositis: a randomized, placebo controlled feasibility
study. Eur J Oncol Nurs 13(2):87–93
134. Talaipour A, Hadad P, Sahba S, Sakhdari S (2000) Chamomile mouth rinse effects on muco-
sitis reduction after radiotherapy. J Dent Med 13(1):57–62
135. Ameri A, Heydarirad G, Mahdavi Jafari J, Ghobadi A, Rezaeizadeh H, Choopani R (2015)
Medicinal plants contain mucilage used in traditional Persian medicine (TPM). Pharm Biol
53(4):615–623
136. Ameri A, Heydarirad G, Rezaeizadeh H, Choopani R, Ghobadi A, Gachkar L (2016)
Evaluation of efficacy of an herbal compound on dry mouth in patients with head and neck
cancers a randomized clinical trial. J Evid Based Complementary Altern Med 21:30–33
137. Iglesias-Bartolome R, Patel V, Cotrim A, Leelahavanichkul K, Molinolo AA, Mitchell JB
et al (2012) mTOR inhibition prevents epithelial stem cell senescence and protects from
radiation-induced mucositis. Cell Stem Cell 11(3):401–414
138. Han G, Bian L, Li F, Cotrim A, Wang D, Lu J et al (2013) Preventive and therapeutic effects
of Smad7 on radiation-induced oral mucositis. Nat Med 19(4):421–428
139. Dörr W, Herrmann T (2007) Efficacy of Wobe-Mugos® E for reduction of oral mucositis
after radiotherapy. Strahlenther Onkol 183(3):121–127
140. ORAL MUCOSITIS - BC Cancer Agency [Internet] (2013) Available from www.bccancer.
bc.ca/nursing
141. Roopashri G, Jayanthi K, Guruprasad R (2011) Efficacy of benzydamine hydrochloride,
chlorhexidine, and povidone iodine in the treatment of oral mucositis among patients under-
going radiotherapy in head and neck malignancies: a drug trail. Contemp Clin Dent
2(1):8–12
References 77

142. Berger AM, Shuster JL, Von Roenn JH (2013) Principles and practice of palliative care and
supportive oncology. Lippincott Williams & Wilkins, Philadelphia, PA
143. Leenstra JL, Miller RC, Qin R, Martenson JA, Dornfeld KJ, Bearden JD et al (2014) Doxepin
rinse versus placebo in the treatment of acute oral mucositis pain in patients receiving head
and neck radiotherapy with or without chemotherapy: a phase III, randomized, double-blind
trial (NCCTG-N09C6 [alliance]). J Clin Oncol 32(15):1571–1577
144. Moynihan TJ I have mouth sores from receiving chemotherapy. I’ve heard that something
called “magic mouthwash” might help. What is it?
145. Healing E, No O. (2009) Pharmacist’s letter/prescriber’s letter
146. Hadjieva T, Cavallin-Ståhl E, Linden M, Tiberg F (2014) Treatment of oral mucositis pain
following radiation therapy for head-and-neck cancer using a bioadhesive barrier-forming
lipid solution. Supp Care Cancer 22(6):1557–1562
147. Tiberg F C-S, Linden L, Thuresson K, Hadjieva T (2009) Treatment of oral mucositis pain by a
bioadhesive barrier-forming lipid solution. MASCC International Symposium; Rome, Italy, June
148. Naidu MURG, Ratnam SV, Sudhavani T, Naidu KJ, Roy P, Suresh P, Rani PU, Mohan IK
(2005) A randomised, double-blind, parallel, placebo-controlled study to evaluate the effi-
cacy of MF 5232 (Mucotrol), a concentrated oral gel wafer, in the treatment of oral mucositis.
Drugs R D 6(5):291–298
149. FDA approvals: mucotrol, clindesse, multihance, and others [Internet]. (2016) Available from
www.mdscape.com
150. Acetylcysteine rinse in reducing saliva thickness and mucositis in patients with head and
neck cancer undergoing radiation therapy [Internet]. Available from Clinical Trials.gov
151. Ahmedzai S, Brooks D, Group T-FCT (1997) Transdermal fentanyl versussustained-release
oral morphine in cancer pain: preference, efficacy, and quality of life. J Pain Symptom Manag
13(5):254–261
152. Coluzzi PH, Schwartzberg L, Conroy JD, Charapata S, Gay M, Busch MA et al (2001)
Breakthrough cancer pain: a randomized trial comparing oral transmucosal fentanyl citrate
(OTFC®) and morphine sulfate immediate release (MSIR®). Pain 91(1):123–130
153. Gupta A, Duckles B, Giordano J (2009) Use of sublingual methadone for treating pain of
chemotherapy-induced oral mucositis. J Opioid Manag 6(1):67–69
154. Darwish M, Kirby M, Robertson P, Tracewell W, Jiang JG (2007) Absorption of fentanyl
from fentanyl buccal tablet in cancer patients with or without oral mucositis. Clin Drug Invest
27(9):605–611
155. Sarvizadeh M, Hemati S, Meidani M, Ashouri M, Roayaei M, Shahsanai A (2015) Morphine
mouthwash for the management of oral mucositis in patients with head and neck cancer. Adv
Biomed Res 4:44
156. Vayne-Bossert P, Escher M, de Vautibault CG, Dulguerov P, Allal A, Desmeules J et al (2010)
Effect of topical morphine (mouthwash) on oral pain due to chemotherapy-and/or
radiotherapy-­induced mucositis: a randomized double-blinded study. J Palliat Med
13(2):125–128
157. Bar Ad V, Weinstein G, Dutta PR, Dosoretz A, Chalian A, Both S et al (2010) Gabapentin for
the treatment of pain syndrome related to radiation-induced mucositis in patients with head
and neck cancer treated with concurrent chemoradiotherapy. Cancer 116(17):4206–4213
158. Barker G, Loftus L, Cuddy P, Barker B (1991) The effects of sucralfate suspension and
diphenhydramine syrup plus kaolin-pectin on radiotherapy-induced mucositis. Oral Surg
Oral Med Oral Pathol 71(3):288–293
159. Baharvand M, Hamian M, Moosavizadeh M, Mortazavi A, Ameri A (2015) Phenytoin
mouthwash to treat cancer therapy-induced oral mucositis: a pilot study. Indian J Cancer
52(1):81
160. Lowry F (2015) Laser treatment halts oral mucositis in its tracks. Oncology Nursing Society
(ONS) 40th Annual Congress 2015
161. Treatment of oropharyngeal and esophageal candidiasis [Internet]. Available from www.
uptodate.com
78 6 Oral Mucositis

162. Nicolatou-Galitis O, Athanassiadou P, Kouloulias V, Sotiropoulou-Lontou A, Dardoufas K,


Polychronopoulou A et al (2006) Herpes simplex virus-1 (HSV-1) infection in radiation-­
induced oral mucositis. Supp Care Cancer 14(7):753–762
163. Fitzmorris KL, Greene JN, Sandin RL, Field T (2000) Recognition and management of her-
pes simplex mucositis in neutropenic patients. Inf Med 17(6):413–416
164. Treatment of herpes simplex virus type 1 infection in immune competent patients. Uptodate
2015 [Internet]. Available from www.uptodate.com
165. Elting LS, Cooksley CD, Chambers MS, Garden AS (2007) Risk, outcomes, and costs of
radiation-induced oral mucositis among patients with head-and-neck malignancies. Int
J Radiat Oncol Biol Phys 68(4):1110–1120
166. Deng G, Cassileth BR (2013) Supportive care and quality of life. Perez and Brady’s princi-
ples and practice of radiation oncology. 6th edn. Philadelphia, PA, Lippincott Williams &
Wilkins, Section V, Chapter 97
Xerostomia
7

Dry mouth (xerostomia) is one of the most common complications of radiation


therapy in head and neck cancers due to the high radiosensitivity of the salivary
glands.
Xerostomia is a common debilitating side effect of radiation therapy for head
and neck cancers that affects chewing, eating, tasting, swallowing, tooth decay, and
speaking. Xerostomia has a negative impact on quality of life in cancer patients.
It has been estimated that the incidence of acute xerostomia induced by radiation
therapy of head and neck cancers is 60–90% depending on tumor site, irradiation
technique, and time of evaluation; it is observed in 30% of patients with advanced
cancer in need of starting a palliative care program [1, 2].
In regard to timing of radiation therapy, the incidence of xerostomia is 6%, 93%,
and 83% before, during, and 1–3 months after treatment, respectively [3]. In regard
to type of radiation therapy, xerostomia occurs in 81%, and 71% of patients under-
went conventional head and neck radiation therapy during and 1–3 months after
treatment, respectively. New radiation techniques such as intensity-modulated radi-
ation therapy (IMRT) may spare salivary glands more than conventional therapies
and decrease xerostomia rate and severity [3].
Moderate to severe xerostomia occurs in 60.2% of patients with nasopharyngeal
carcinoma and in 32.9% of patients with other sites of head and neck cancer
3 months after treatment with intensity-modulated radiation therapy [4].

7.1 Mechanism

In order to understand the mechanism of xerostomia induction in radiation therapy,


salivary gland physiology needs to be understood.
The terms of major and minor salivary glands refer to their anatomic size. There
is a difference in the quality of content and quantity of production during different
time points through the day, which may affect the clinical symptoms related to each
salivary gland dysfunction.

© Springer International Publishing AG 2017 79


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_7
80 7 Xerostomia

Normal salivary flow is highly variable and is usually 0.25 mL/min (1–1.5 L per
day). A salivary flow of less than 0.12–0.16 mL/min is considered to be abnormal [5].
The parotid glands have only serous-secreting acinar cells and exclusively pro-
duce serous watery secretion. Up to 50% of stimulated saliva and approximately
20% of unstimulated saliva secretion volume comes from parotid glands
secretion.
The submandibular glands have both serous and mucous acinar cell types
(10% mucous cells and 90% serous cells) and produce a mixed serous and mucous
saliva. They contribute 65% of unstimulated saliva secretion and 35% of stimulated
saliva secretion.
The sublingual glands mainly have mucous acinar cells and produce predomi-
nantly thick, viscous saliva. They contribute less than 10% of unstimulated saliva
volume.
The minor glands, which are distributed throughout the upper aerodigestive
mucosa (e.g., labial, buccal, lingual, and palatinal mucosa), are mixed glands largely
comprised of mucous acinar cells. The minor glands produce less than 10% of the
total volume of saliva [6].
During the day, major saliva production in stimulated and unstimulated condi-
tions is related to parotid and submandibular glands, respectively [7].These major
salivary gland dysfunctions are major causes of xerostomia induced by radiation
therapy.
Salivary glands are highly radiosensitive. Serous acinar cells of the salivary
glands are well differentiated and have a slow mitotic rate and turnover, but they
behave like acutely responding tissues to radiation and undergo interphase cell
death by apoptosis [8, 9]. Mucous acinar cells of salivary glands have a lower
radiosensitivity than serous acinar cells, and they have a trend to retain their
function for some time later [10]. The parotid glands seem to lose more function
than do the other salivary glands, resulting in a decrease in watery saliva and
accumulation of sticky mucus. However, the difference in radiosensitivity
between the parotid and submandibular/sublingual salivary glands is still contro-
versial [11–13].
It has been found that saliva production reduces during the first days of radiation
therapy. Membrane damage of saliva-producing cells confounding with receptor-­
mediated signaling pathways of water excretion and functional loss is responsible
for the early hyposalivation. Cell death does not occur in the acute phase but is a late
event [14].
Saliva is a complex fluid, mostly composed of water (99%) and a minority of
various nonorganic and organic substances such as enzymes, hormones, antibodies,
antimicrobial constituents, and growth factors. Saliva quality changes during radia-
tion therapy, including increased viscosity, decreased transparency, yellow/brown
discoloration, declined production of glycoproteins (e.g., immunoglobulin A),
decreased salivary pH (from 6.8 and 7.0 to 5.5), altered salivary electrolyte levels
(increases in the concentrations of sodium, chloride, calcium, and magnesium with
slight potassium level change), and shift in certain intraoral microbial populations
7.3 Risk Factors 81

(increase in Streptococcus mutans and species of Lactobacillus, Candida (primarily


Candida albicans), and Staphylococcus, with parallel decreases in Streptococcus
sanguinis and species of Neisseria and Fusobacterium) [13, 15–18].

7.2 Timing

Radiation-induced xerostomia usually initiates early during radiation therapy for


head and neck cancers containing salivary glands within the radiation fields. When
the radiation dose reaches 10–20 Gy with 2 Gy per day, which corresponds to the
first to second week of treatment, a 50–60% decrease in salivary flow occurs, and
the patient may begin to experience mild to moderate dryness of the mouth, which
may progressively worsen over the course of treatment. After 7 weeks of radiation
therapy, salivary flow decreases to approximately 20% and continues to further drop
for more than 6 months after completion of treatment [11, 19, 20]. Some recovery
is possible until 12–18 months after radiation therapy depending on the dose
received by the salivary glands and the volume of the gland tissue included in the
irradiation volume; there is also a compensatory hypertrophy of the non-irradiated
salivary gland tissue. Xerostomia may rarely recover a few years after radiation
therapy [19, 20].
Radiation-induced salivary gland damage may be reversible or permanent based
on the radiation dose. With a dose less than 60 Gy, changes in the salivary glands,
including edema and inflammation, are reversible. Although, for acceptable func-
tion, the radiation doses to salivary glands should be more lower than 60 Gy. When
the dose exceeds 60 Gy, changes may be permanent, with fibrosis and glandular
degeneration [13].

7.3 Risk Factors

The occurrence and severity of radiation therapy-induced xerostomia are dependent on


radiation-related factors, mostly salivary gland radiation dose and volume within the
radiation field [21]. Altered fractionation schedules’ impact on the incidence of xero-
stomia is not clearly defined. It seems that accelerated and hyperfractionated radiation
therapy both increase acute side effects of treatment including xerostomia [22].
Concurrent chemotherapy is associated with a significant risk of developing
acute and late xerostomia [21]. However it has been shown in some studies that
concomitant chemotherapy and intensity-modulated radiation therapy do not
increase the incidence of acute or late xerostomia relative to intensity-modulated
radiation therapy alone [23]. Concomitant cetuximab (a monoclonal antibody
against EGFR) doesn’t increase the xerostomia rate when it’s prescribed concur-
rently with radiation therapy [24].
Patient age is also a contributing factor. With increasing age, the vulnerability of
salivary glands to radiation injury and development of xerostomia increases [7]. It
82 7 Xerostomia

should be noted that increasing age does not cause hyposalivation by itself [25]. The
higher incidence of xerostomia in elderly patients is mainly due to their comorbidi-
ties and use of medications with the potential to develop xerostomia.
No significant association between the risk of developing xerostomia and ethnic-
ity, marital, or socioeconomic status has been observed [21].
It has been proposed that the progression of xerostomia can be improved by mix-
ing clinical and dose-volume factors. The mean dose given to the contralateral and
ipsilateral parotid glands is the most significant predictors in multivariable normal
tissue complication probability models for xerostomia. Age, financial status,
T stage, and educational level are proposed clinical predictive factors for radiation-­
induced xerostomia. However some of these clinical datasets such as financial status
and education may affect the patient-reported xerostomia, many of which need to be
investigated before they are incorporated into the models [4].
The volumes of parotid and submandibular glands are decreased due to radiation
therapy. The parotid gland volume reduces about 30% during radiotherapy. The
lateral regions of the irradiated parotid glands move inward. The irradiated subman-
dibular glands also shrink and move upward. Parotid shrinkage during treatment is
accompanied by a decrease in tissue density consistent with a relative increase in fat
over glandular tissue [26, 27].
Parotid gland density and volume variations during radiation therapy may pos-
sibly predict acute xerostomia. It has been found that a higher score of acute xero-
stomia is predicted by higher density and volume variations in the first 2 weeks of
treatment. Further studies are necessary to definitively assess the potential of early
density/volume changes in identifying more sensitive patients at higher risk of
developing xerostomia [28].

7.4 Symptoms

Dryness is one of the most unpleasant oral symptoms that adversely affects all oral
functions and compromises oral health. Patients may experience dry oral mucosa
and thick, sticky saliva requiring them to adjust their diet and keep the mouth moist
with water [1, 29].
Both acute and chronic xerostomia induce functional alterations such as chew-
ing, swallowing, speaking, burning, and pain, with a propensity to bacterial and
fungal infection, demineralization of teeth, and increase in caries, dysgeusia, gag-
ging sensations, a fear of choking, and odynophagia. The patient may have bad
breath secondary to food stagnation in the oral mucosa, gingiva, teeth, or tongue
[30, 31].
Cheilitis, a fissuring or ulceration in the angles of the mouth and erythematous
tongue due to damage to the dorsal epithelium, can also be seen in patients with
xerostomia [29, 30].
Quality of life significantly worsens along with the severity of xerostomia. With
each milliliter decrease in saliva secretion, the quality of life score decreases by
2.25% [32].
7.6 Scoring 83

Fig. 7.1 Grade 2


xerostomia in tongue
cancer 1 month after
radiotherapy

7.5 Diagnosis

Xerostomia is a clinical diagnosis based on patient signs and symptoms (Fig. 7.1).
The objective tests of salivary flow are not usually used for diagnosis because there
is little correlation between patient symptoms and these tests. Therefore, clinical
management should be based on patient symptoms [33].
There are some differential diagnoses including oral infections, bad oral condi-
tions, and mucositis, which may be ruled out with a detailed history and clinical
exam. However, they can all occur in association with each other.

7.6 Scoring

For measuring the severity of xerostomia, there are assessment tools based on
patient- and observer-reported data.
Observer-based toxicity scoring is generally based on the Radiation Therapy
Oncology Group (RTOG)/European Organization for Research and Treatment of
84 7 Xerostomia

Table 7.1 RTOG/EORTC xerostomia scoring


Definition
Score 0 No change over baseline
Score 1 Mild mouth dryness/slightly thickened saliva/ may have slightly altered taste such as
metallic taste/these changes not reflected in alteration in baseline feeding behavior,
such as increased use of liquids with meal
Score 2 Moderate to complete dryness/thick, sticky saliva/markedly altered taste
Score 3 –
Score 4 Acute salivary gland necrosis

Table 7.2 Michigan University xerostomia questionnaire


0 1 2 3 4 5 6 7 8 9 10
Rate your difficulty in talking due to dryness
Rate your difficulty in chewing due to dryness
Rate your difficulty in swallowing solid food due to dryness
Rate the frequency of your sleeping problems due to dryness
Rate your mouth or throat dryness when eating food
Rate your mouth or throat dryness while not eating
Rate the frequency of sipping liquids to aid swallowing food
Rate the frequency of sipping liquids for oral comfort when
not eating

Cancer (EORTC) grading scale (Table 7.1). Because of the low correlation between
the measured salivary output and observer-reported xerostomia [34] and underesti-
mation of the severity of xerostomia with these scoring systems [35], several xero-
stomia questionnaires have been developed to permit patient self-reporting.
One of the most validated patient self-reported questionnaires is xerostomia
questionnaire (XQ), which was developed by the University of Michigan (Table 7.2).
It consists of eight questions; patients rate each item on a scale from 0 to 10. Higher
scores are related to more severe xerostomia [35].

7.7 Prevention

There are some preventive approaches for radiation-induced xerostomia, including


more conformal radiation delivery technology, radioprotective agents, and even pre-
irradiation surgical techniques [36, 37].

7.7.1 Amifostin

Available data show that amifostine significantly reduces the incidence of acute and
long-term xerostomia. Amifostine, an organic thiophosphate agent, is a prodrug that is
dephosphorylated by alkaline phosphatase in tissues after administration, converting it
into its active form. This active form enters the cells and acts as a scavenger against free
radicals. Amifostine concentrations in tumor cells are lower than normal tissue due to
7.7 Prevention 85

lower alkaline phosphatase levels and the pH of tumors, and therefore normal tissue
protection is provided [20]. However, amifostine’s effect on tumor cell protection is
still a concern that precludes it from extensive administration as a radioprotective agent.
Brizel et al. in a phase III trial randomized 303 patients that received conventional
radiation therapy for head and neck cancers (both postoperative and as primary treat-
ment) to receive amifostine daily before each fraction (200 mg/m2 intravenously). They
found that amifostine significantly reduced the incidence of grade 2 acute xerostomia
from 78% to 51% and reduced the incidence of grade 2 chronic xerostomia from 57%
to 34% without a difference in disease control or survival [38]. Amifostine was conse-
quently approved by the US Food and Drug Administration (FDA).
Amifostine can be administered only with standard fractionated radiation ther-
apy without chemotherapy and only when ≥75% of both parotid glands are exposed
to radiation in the postoperative setting. Amifostine should not be administered in
patients receiving definitive radiation therapy, except in the context of a clinical
trial, because of insufficient data to exclude a tumor-protective effect in this setting
[39]. Amifostine administration in the setting of concurrent chemotherapy with
radiation therapy and in the setting of accelerated and hyperfractionated therapy has
not been systematically studied [40–43].

7.7.1.1 Prescription
Each vial contains 500 mg of amifostine on the anhydrous basis, requiring reconsti-
tution for intravenous infusion [44]. Prior to intravenous injection, amifostine is
reconstituted with 9.7 mL of sterile 0.9% sodium chloride. The reconstituted solu-
tion (500 mg Amifostine/10 mL) is chemically stable for up to 5 h at room tempera-
ture (approximately 25 °C) or up to 24 h under refrigeration (2 °C–8°C). Amifostine
is prepared in polyvinylchloride (PVC) bags and is available at concentrations rang-
ing from 5 mg/mL to 40 mg/mL [45].
Standard amifostine intravenous administration: 200 mg/m2 over 3 min once
daily 15–30 min prior to radiation therapy [46, 47].
Amifostine can also be administered subcutaneously (unlabeled route): 500 mg
once daily prior to radiation therapy [46, 48, 49].
Note: Because of rapid clearance from the blood and tissue, the drug needs to be
delivered shortly before radiation therapy.

7.7.1.2 Contraindications
Hypersensitivity to aminothiol compounds or any component of the formulation.

7.7.1.3 Advers effects


Several adverse effects have been reported for amifostine including hypotension,
nausea and vomiting, hypocalcemia, and cutaneous reactions.
Blood pressure should be monitored every 5 min during the infusion and there-
after as clinically indicated. The infusion of amifostine should be interrupted if the
systolic blood pressure decreases significantly from baseline [45, 48].
Amifostine is a moderately to highly emetogenic agent, and antiemetic medica-
tions (including dexamethasone 20 mg I.V. and a serotonin 5-HT3 receptor antago-
nist) should be administered prior to and in conjunction with amifostine [50].
86 7 Xerostomia

Serum calcium levels at baseline should be checked and monitored in patients at


risk for hypocalcemia, such as those with nephrotic syndrome or patients receiving
multiple doses of amifostine for injection [45, 48].
Cutaneous evaluation of the patient prior to each administration and permanent dis-
continuation for serious or severe cutaneous reactions should be performed [44, 45].

7.7.2 IMRT

The volume of salivary tissue irradiated is related to the occurrence of xerostomia.


With introduction of novel radiation therapy delivery techniques including inten-
sity-modulated radiation therapy (IMRT), partial sparing of salivary glands may
become possible, and thus acute and late xerostomia are significantly reduced [51,
52]. Mean dose to the parotid glands should be reduced as much as clinically pos-
sible. A mean dose of less than 20 Gy for at least one parotid gland or a mean dose
of less than 25 Gy for both glands may prevent severe long-term xerostomia [53]. It
has been suggested that the mean parotid dose for both sides together has a higher
predictive value over considering each side separately [27].
The effect of amifostine combined with IMRT is not clear. It seems that the protective
effect of IMRT in sparing saliva is much greater than the effect of amifostine [54, 55].
In patients that need bilateral radiation therapy of the head and neck including
bilateral parotids within the radiation fields, the effect of IMRT in prevention of
xerostomia is limited.
IMRT provides a way to spare all salivary glands and improves xerostomia-­
related quality of life during meals and at rest; however, the largest effect is still on
xerostomia during meals [56]. As noted previously, the parotid glands are largely
responsible for the stimulated saliva output, whereas the minor salivary glands and
submandibular glands are mainly responsible for unstimulated saliva (lubrication in
rest). The risk of xerostomia decreases or is even eliminated with sparing of at least
one parotid gland and reduces with sparing of at least one submandibular gland
[53]. It has been reported that a lower mean dose to the oral cavity (<40 Gy) and
contralateral submandibular gland (<50 Gy) is each associated with lower patient-­
reported and observer-rated xerostomia [51].

7.7.3 Submandibular gland transfer

Another preventive approach is submandibular gland transfer. The submandibular


gland is responsible for most of the unstimulated salivary volume, which is impor-
tant in the subjective symptoms of xerostomia and oral homeostasis. Some argue
that sparing the submandibular gland is preferable to sparing the parotid gland [29].
In this approach, a single submandibular gland is transferred into the submental
space during a surgical procedure referred to as the Seikaly-Jha procedure (SJP).
The borders of the transferred gland are marked with wire to help identify it during
7.7 Prevention 87

radiation therapy and can be shielded from the radiation [57, 58]. The surgical
method of transfer is a quick, easy, and simple procedure that can be done during
surgical treatment of the primary tumor [20, 59–61].
The submandibular gland may be damaged during the transfer, and complete
prevention cannot be achieved, although it is still superior to some other preventive
options including pilocarpine in regard to median salivary flow and saliva consis-
tency. It has been estimated that the submandibular gland transfer could reduce
69% of the risk of acute xerostomia. However, after radiation therapy, the salivary
gland repaired itself and the rate of late xerostomia can reach 19% [62]. Local/
regional recurrence and survival outcome after submandibular gland transfer seem
not to be compromised; however there is some controversy and need for more
evaluation [41, 63].
Submandibular gland transfer should be conducted on the gland of the contralat-
eral side of the primary cancer. The procedure should not be performed for patients
with cancer of the oral cavity, bilateral neck lymph node involvement, submandibu-
lar or submental neck lymph node involvement, or advanced neck disease (N3). For
accurate selection of patients that are candidates for submandibular gland transfer,
suspicious nodes and all level I lymph nodes (submental and submandibular) are dis-
sected and sent for frozen section evaluation before transfer. If any of the nodes are
involved with cancer, the transfer procedure should be abandoned [62].
Collectively, quality of life in head and neck cancer patients that are at risk of
radiation-induced xerostomia may improve with submandibular gland transfer [58,
59, 63]. However, further studies are still needed to confirm the safety of this
surgery.

7.7.4 Pilocarpine

Pilocarpine has been approved for postradiation xerostomia. The administration of


pilocarpine during radiation therapy has been studied to prevent the subsequent
development of xerostomia.
Pilocarpine hydrochloride is a muscarinic-cholinergic agent that can stimulate
salivary glands [64–66].
Several studies have shown positive results of pilocarpine in the prevention of
radiation-induced xerostomia, especially when the doses of whole parotid glands
are more than 45 Gy. However, others have concluded that oral pilocarpine could
not be recommended to prevent xerostomia in patients receiving radiation therapy
for head and neck cancers [67].
It has been found that pilocarpine administration during radiation therapy can
result in a significant improvement in unstimulated salivary flow at the end of treat-
ment but not in stimulated flow. This observation can be explained with the mecha-
nism of pilocarpine’s protective effects [68]. Pilocarpine stimulates the residual
function of salivary tissues outside the radiation fields, including non-irradiated
parts of parotid and other major salivary glands/minor salivary glands, and it has no
88 7 Xerostomia

protective effect in the glands that are completely irradiated [69–72]. Prominently
unstimulated salivary flow is preserved.
The other hypothesis proposed for the mechanism of pilocarpine’s protective effect is
related to indirect inhibition of radiation-induced oxidative damage. Pilocarpine reduces
heavy metals such as zinc, manganese, and iron, which are found in secretary granules,
leading to reduction in intracellular leakage of proteolytic enzymes in secretary granules
after radiation damage and reduction in subsequent serous cell autolysis [67, 70].
Data based on in vitro studies indicate that pretreatment pilocarpine administration
does not protect tumor cells and has no effect on radiosensitivity of cell lines [73].

7.7.4.1 Prescription
Start pilocarpine 5 mg three times daily with irradiation, and continue until 3 months
after the end of radiotherapy [74].
Available tablets: 5 mg, 7.5 mg
Taking with a high-fat meal reduces pilocarpine absorption. Give the medication
with food if GI distress occurs.)

7.7.4.2 Contraindications
Hypersensitivity to pilocarpine or any component of the formulation, uncontrolled
asthma, acute iritis or glaucoma, and severe hepatic impairment (adjust dose with
moderate hepatic impairment) [75]
Pilocarpine should be used with extreme caution in patients that have chronic
obstructive pulmonary disease and cardiovascular disease.

7.7.5 Acupuncture

Acupuncture has been investigated in the management of patients with xerostomia


and demonstrated some benefits for improving salivary flow rates and reducing
xerostomia-related symptoms. Based on these observations, the preventive approach
of acupuncture has been recently addressed with promising results [76].
Low-level laser therapy refers to local application of a high-density monochromatic
narrowband light source with the output power range from 5 to 500 mW and a wave-
length between 600 and 1000 nanometers (like helium/neon with wavelength 632.8 nm
or diode laser with various wavelengths 630–680, 700–830, and 900 nm) [77].
Low-level laser therapy may result in improvement of the salivary flow by stimu-
lating salivary glands and the regenerative effect with an increase in the number of
mitosis [78]. Although the use of a low-power laser may prevent xerostomia, it
needs further study to be better implemented [78, 79].
Patients with Sjögren syndrome have been studied and shown that low-level laser
therapy can be safely and effectively used in these patients to reduce xerostomia
[80, 81]. However for detection of low-level laser therapy efficacy on radiation-­
induced xerostomia, long-term follow-up is necessary.
7.8 Management 89

7.8 Management

Therapeutic interventions include supportive care, saliva supplementation, and the


use of procholinergic salivary secretagogues.

7.8.1 Supportive Care

7.8.1.1 Oral Rinses


Oral rinses are recommended to keep the mouth moist and clean by removing
debris.
Patients are informed to use 1 tablespoon (15 mL) of oral rinse such as normal
saline (NS) or 1/2 teaspoon (2.5 mL) of salt in 240 mL of water to swish in the
oral cavity for 30 s and then spit out [82]. Commercially available alcohol-con-
taining oral rinses should be avoided due to their drying effect. Limited data have
proposed that all Biotène products, which include a range of toothpastes, mouth-
washes, mouth sprays, chewing gum, and gels, can improve many of the symp-
toms of radiation-­induced xerostomia, but larger studies are needed to evaluate
efficacy [83].

7.8.1.2 Chewing to Stimulate Secretion of Residual Glandular Tissue


Chewing is a stimulus for inducing salivary glands to secret more saliva, and chew-
ing several times a day can be helpful in reducing symptoms [40].

7.8.1.3 Maintaining Adequate Nutrition


Patients may benefit from a nutritional consult and should be encouraged to moisten
their foods by adding butter, mayonnaise, vegetable oil, yogurt ,or sauces and have
a sip of a liquid after each bite, which helps chewing and swallowing. Patients
should avoid foods high in sugar due to their susceptibility to dental caries, dry or
spicy food, and excessively hot or cold beverages [40].
Xylitol is a natural sweetener product that differs chemically from other sweet-
eners like sorbitol, fructose, or glucose. It actually interferes with the growth of
bacteria associated with tooth decay, and it is approved as a therapeutic sweetener
by the Food and Drug Administration [40].
These patients should avoid irritating foods that are astringents or that may stick
to the teeth [40].
Alcohol, tobacco, and large amounts of caffeine should be avoided due to their
drying effect [82].

7.8.1.4 Drink Adequate Fluids


Patients should be instructed to always carry water with them and keep themselves
well hydrated [82].
90 7 Xerostomia

7.8.1.5 Protection from Dry Lips


Using topically applied water- or aloe-based lubricant after oral care, at bedtime,
and as often as required is helpful [82].

7.8.1.6 Alleviating Nocturnal Symptoms


A mouth guard, a cold air humidifier in the form of a bedside vaporizer or household
humidifier, or applying a small amount of dentifrice on smooth dental surfaces
(especially using anti-xerostomia dentifrices) can alleviate the nocturnal oral dry-
ness [15].

7.8.1.7 Keeping Appropriate Oral Hygiene and Dental Care


Patients should be instructed on tooth brushing, dental flossing, tongue brushing,
and oral mouth rinsing with an appropriate antibacterial mouth rinse such as
chlorhexidine and hexitidine several times a day.

7.8.2 Saliva Supplementation

Replacement therapy with artificial saliva or saliva substitutes can be used to lubri-
cate the mouth. Various substitutes are available commercially that are formulated
to mimic natural saliva [41]. They have short-term activity with no stimulating
effect on salivary glands.
A variety of forms of products including solutions, sprays, gels, and lozenges are
available. In general, they contain an agent to increase viscosity, such as carboxy-
methylcellulose or hydroxyethylcellulose, minerals such as calcium and phosphate
ions and fluoride, preservatives such as methylparaben or propylparaben, and fla-
voring and related agents [41].
Artificial substitutes do not replace the antibacterial and immunologic protection
of saliva and do not exclude the need for regular dental care and appropriate oral
hygiene [11].

7.8.3 Acupuncture

Acupuncture may have a stimulating effect on saliva production and may be a useful
treatment for some patients. The possible mechanism of acupuncture is related to
parasympathetic central nervous system processes, which increase the concentra-
tion of salivary neuropeptides and modulate the complex process of salivary secre-
tion. Another possible mechanism of acupuncture is stimulation of minor salivary
glands present in non-irradiated buccal mucosa [15, 84, 85].
There are some studies that provide encouraging results for using acupuncture in
xerostomia [84, 86–90], while others do not provide a statistically significant effect
on the increase of salivary flow [91, 92]. Further research on acupuncture is neces-
sary prior to the recommendation for widespread clinical implementation in xero-
stomia treatment.
7.8 Management 91

7.8.4 Drugs

Patients with radiation-induced xerostomia may have a minimal response (mainly with
an increase in resting saliva) to systemic sialagogues, but small increases in saliva may
translate into subjective patient willingness [93]. Several therapeutic drugs have been
used to treat xerostomia. Pilocarpine and cevimeline are two systemic FDA-approved
sialagogues for the treatment of xerostomia [94]. In respect to radiation-induced xero-
stomia, pilocarpine is the sole sialagogue agent approved by the FDA [20].
Administration: 5 mg 3 times/day, titration up to 10 mg 3 times/day may be con-
sidered for patients that have not responded adequately (not to exceed 30 mg/day).
After the administration of pilocarpine, salivary output increases rapidly, usually
reaching a maximum within 1 h and returning to baseline at 3 h [31, 95–99].
Systemic administration of pilocarpine is associated with an increased risk of
side effects (e.g., mild-to-moderate sweating, flushing, headache, nausea, urinary
frequency, lacrimation, and rhinitis). Local administration of pilocarpine (topical
pilocarpine) may be an appropriate alternative with a potential decrease in systemic
side effects. Local stimulation of saliva more rapidly increases salivary production
rather than systemic pilocarpine. Overall improvement of xerostomia seems to not
be significantly different between oral and systemic pilocarpine. Further investiga-
tion is required to provide more definitive conclusions [72].
Cevimeline is a newer muscarinic agonist that has been studied in patients with
xerostomia after radiation therapy for head and neck cancers with positive results
[100]. Cevimeline (45 mg three times daily) studied in patients with radiation-­
induced xerostomia showed that 69% experienced an adverse effect, mostly mild to
moderate in severity. The most frequent side effects were sweating, dyspepsia, nau-
sea, and diarrhea [101]. There is no significant difference in overall increase of
production of saliva with cevimeline or pilocarpine [102]. Based on animal studies,
the central nervous system effects are more common with cevimeline than pilocar-
pine, but no clear difference was observed in respiratory and cardiovascular effects.
Clinical trials are ongoing to determine the efficacy and side effects of both cevime-
line and pilocarpine in the secretion of saliva for patients with xerostomia.
Bromhexine, a mucolytic agent, has been used by patients with radiation-induced
xerostomia. However, pilocarpine has demonstrated superiority to bromhexine in
improving xerostomia symptoms [103].
Bethanechol, with prolonged muscarinic and nicotinic-cholinergic activity, has
also been used in patients with radiation-induced xerostomia with no significant
difference in efficacy and side effects rather than pilocarpine [93, 104].
Malva sylvestris L and Alcea digitata (Boiss) Alef have been used as herbal reme-
dies in traditional Persian medicine for their antitussive, antioxidant, expectorant, anti-
inflammatory, antimicrobial, and laxative therapeutic effects [105]. They are useful for
lubrication of the throat and lungs and in respiratory disorders. Studies have shown
these plants to be immune stimulants that are useful in mucositis. This compound was
compared with artificial saliva in a randomized trial [106].The herbal group showed a
significant difference between the grade of dry mouth before and after intervention,
but no change was observed for the grade of dry mouth in the artificial saliva group.
92 7 Xerostomia

References
1. Olver IN (2006) Xerostomia: a common adverse effect of drugs and radiation. Aust Prescr
29(4). doi:10.18773/austprescr.2006.063
2. Baigal G, Ranajit K, Agarwal J (2012) Radiation induced xerostomia. Int J Head Neck Surg
3(2):82–86
3. Jensen SB, Pedersen A, Vissink A et al (2010) A systematic review of salivary gland hypo-
function and xerostomia induced by cancer therapies: prevalence, severity and impact on
quality of life. Sup Care Cancer 18(8):1039–1060
4. Lee T-F, Liou M-H, Huang Y-J, Chao P-J, Ting H-M, Lee H-Y et al (2014) LASSO NTCP
predictors for the incidence of xerostomia in patients with head and neck squamous cell car-
cinoma and nasopharyngeal carcinoma. Sci Rep 4
5. Navazesh M, Christensen C, Brightman V (1992) Clinical criteria for the diagnosis of sali-
vary gland hypofunction. J Dent Res 71(7):1363–1369
6. Salivary glands. allegra comba [Internet]. 2007. Available from flipper. http://flipper.diff.org/
apptagsaccount/items/367
7. Beetz I, Schilstra C, Visink A, van der Schaaf A, Bijl HP, van der Laan BF et al (2013) Role
of minor salivary glands in developing patient-rated xerostomia and sticky saliva during day
and night. Radiother Oncol 109(2):311–316
8. Grundmann O, Mitchell G, Limesand K (2009) Sensitivity of salivary glands to radiation:
from animal models to therapies. J Dent Res 88(10):894–903
9. Witt RL (2005) Salivary gland diseases: surgical and medical management. Thieme, New York
10. Crispian S, Epstein JB (2012) Xerostomia and hyposalivation in patients with cancer. Head
and Neck Surgery 310–323. Avaiable at http://oralmedicinepacific.com/docs/whats-new/
Hyposalivation-in-Cancer-Patients-Chapter-27-OTO-2013.pdf
11. Berk LB, Shivnani AT, Small W Jr (2005) Pathophysiology and management of radiation-­
induced xerostomia. J Support Oncol 3(3):191–200
12. Cheng SCWV, Kwong DL, Ying MT (2011) Assessment of post-radiotherapy salivary glands.
Br J Radiol 84(1001):393–402
13. Shih A, Miaskowski C, Dodd MJ, Stotts NA, MacPhail L (2003) Mechanisms for radiation-­
induced oral mucositis and the consequences. Cancer Nurs 26(3):222–229
14. Konings AW, Coppes RP, Vissink A (2005) On the mechanism of salivary gland radiosensitiv-
ity. Int J Radiat Oncol Biol Phys 62(4):1187–1194
15. Pinna R, Campus GG, Cumbo EMG, Mura II, Milia EP (2015) Xerostomia induced by radio-
therapy: an overview of the physiopathology, clinical evidence, and management of the oral
damage. Ther Clin Risk Manag 11:171–188
16. Ben-Aryeh H, Gutman D, Szargel R, Laufer D (1975) Effects of irradiation on saliva in can-
cer patients. Int J Oral Surg 4(5):205–210
17. Dreizen S, Brown LR, Handler S, Levy BM (1976) Radiation-induced xerostomia in cancer
patients. Effect on salivary and serum electrolytes. Cancer 38(1):273–278
18. Marks JE, Davis CC, Gottsman VL, Purdy JE, Lee F (1981) The effects of radiation on
parotid salivary function. Int J Radiat Oncol Biol Phys 7(8):1013–1019
19. Valdez IH (1991) Radiation-induced salivary dysfunction: clinical course and significance.
Spec Care Dentist 11(6):252–255
20. Dirix P, Nuyts S, Van den Bogaert W (2006) Radiation-induced xerostomia in patients with
head and neck cancer. Cancer 107(11):2525–2534
21. Liu C-C, Xia R, Guadagnolo A, Cormier JN, Du XL (2011) Risk of xerostomia in association
with the receipt of radiation therapy in older patients with head and neck cancer. Am J Ther
18(3):206–215
22. Poulsen M (ed) (2006) Altered fractionation in locally advanced head and neck cancer-an
update. Cancer Forum, The Cancer Council Australia
23. Miah AB, Gulliford SL, Bhide SA, Zaidi SH, Newbold KL, Harrington KJ et al (2013) The
effect of concomitant chemotherapy on parotid gland function following head and neck
IMRT. Radiother Oncol 106(3):346–351
References 93

24. Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB, Jones CU, Sur R, Raben D,
Jassem J, Ove R, Kies MS, Baselga J, Youssoufian H, Amellal N, Rowinsky EK, Ang KK
(2006) Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N
Engl J Med 354(6):567–578
25. Ship JA, Nolan NE, Puckett SA (1995) Longitudinal analysis of parotid and submandibular
salivary flow rates in healthy, different-aged adults. J Gerontol Ser A Biol Med Sci
50(5):M285–M2M9
26. Ricchetti F, Wu B, McNutt T, Wong J, Forastiere A, Marur S et al (2011) Volumetric change
of selected organs at risk during IMRT for oropharyngeal cancer. Int J Radiat Oncol Biol
Phys 80(1):161–168
27. Sanguineti G, Ricchetti F, Wu B, McNutt T, Fiorino C (2015) Parotid gland shrinkage during
IMRT predicts the time to Xerostomia resolution. Radiat Oncol 10(1):1
28. Osorio EMV, Hoogeman MS, Al-Mamgani A, Teguh DN, Levendag PC, Heijmen BJ (2008)
Local anatomic changes in parotid and submandibular glands during radiotherapy for oro-
pharynx cancer and correlation with dose, studied in detail with nonrigid registration. Int
J Radiat Oncol Biol Phys 70(3):875–882
29. Kalmykow B (2012) Management of radiation induced xerostomia. University of
Pennsylvania School of Nursing
30. Guchelaar H-J, Vermes A, Meerwaldt J (1997) Radiation-induced xerostomia: pathophysiol-
ogy, clinical course and supportive treatment. Sup Care Cancer 5(4):281–288
31. Mravak-Stipetić M (2012) Xerostomia-diagnosis and treatment. Rad Hrvatske Akademije
Znanosti i Umjetnosti Medicinske Znanosti 511(38)
32. Mohammadalizadeh S, Pourdamghan N, Nakhaei M, Bahador M (2012) Xerostomia after
radiotherapy and its effect on quality of life in head and neck cancer patients. Arch Iran Med
15(4):214
33. Visvanathan V, Nix P (2010) Managing the patient presenting with xerostomia: a review. Int
J Clin Pract 64(3):404–407
34. Eisbruch A (2007) Reducing xerostomia by IMRT: what may, and may not, be achieved.
J Clin Oncol 25(31):4863–4864
35. Meirovitz A, Murdoch-Kinch CA, Schipper M, Pan C, Eisbruch A (2006) Grading xerosto-
mia by physicians or by patients after intensity-modulated radiotherapy of head-and-neck
cancer. Int J Radiat Oncol Biol Phys 66(2):445–453
36. Chambers MS, Rosenthal DI, Weber RS (2007) Radiation-induced xerostomia. Head Neck
29(1):58–63
37. Chambers MS, Garden AS, Kies MS, Martin JW (2004) Radiation-induced xerostomia in
patients with head and neck cancer: pathogenesis, impact on quality of life, and management.
Head Neck 26(9):796–807
38. Brizel DM, Wasserman TH, Henke M, Strnad V, Rudat V, Monnier A et al (2000) Phase III
randomized trial of amifostine as a radioprotector in head and neck cancer. J Clin Oncol
18(19):3339–3345
39. Amifostine - FDA prescribing information, side effects and uses. [Internet]. Available from
https://www.drugs.com/pro/adderall.html
40. Antonadou D, Pepelassi M, Synodinou M, Puglisi M, Throuvalas N (2002) Prophylactic use
of amifostine to prevent radiochemotherapy-induced mucositis and xerostomia in head-and-­
neck cancer. Int J Radiat Oncol Biol Phys 52(3):739–747
41. Vacha PFF, Mahlmann B et al (2003) Randomized phase III trial of postoperative amifostine
+/− chemoradiotherapy in head and neck cancer. Is there evidence for radioprotection?
Strahlenther Onkol 179:385–389
42. Buentzel J, Micke O, Adamietz IA, Monnier A, Glatzel M, de Vries A (2006) Intravenous
amifostine during chemoradiotherapy for head-and-neck cancer: a randomized placebo-­
controlled phase III study. Int J Radiat Oncol Biol Phys 64(3):684–691
43. Haddad R, Sonis S, Posner M, Wirth L, Costello R, Braschayko P et al (2009) Randomized
phase 2 study of concomitant chemoradiotherapy using weekly carboplatin/paclitaxel with or
without daily subcutaneous amifostine in patients with locally advanced head and neck can-
cer. Cancer 115(19):4514–4523
94 7 Xerostomia

44. Ethyol dosage [Internet]. Available from www.drug.com


45. Drug: Amifostine - Ethyol® Oncology - intravenous dilution data [Internet]. Available from
www.globalrph.com/amifostine.htm
46. Amiofostine drug information [Internet]. Available from http://cursoenarm.net/UPTODATE/
contents/mobipreview.htm?21/31/22014?source=see_link
47. Bardet E, Martin L, Calais G, Alfonsi M, Feham NE, Tuchais C et al (2011) Subcutaneous
compared with intravenous administration of amifostine in patients with head and neck can-
cer receiving radiotherapy: final results of the GORTEC2000-02 phase III randomized trial.
J Clin Oncol 29(2):127–133
48. Samuels MA, Chico IM, Hirsch RL, Fullmer K, Ethyol SQ Safety Study Group (2003)
Ongoing prospective multicenter safety study of the cytoprotectant amifostine given subcuta-
neously: overview of trial design. Semin Oncol 30(6 Suppl 18):94–95
49. Koukourakis MI, Kyrias G, Kakolyris S, Kouroussis C, Frangiadaki C, Giatromanolaki A
et al (2000) Subcutaneous administration of amifostine during fractionated radiotherapy: a
randomized phase II study. J Clin Oncol 18(11):2226–2233
50. RxMed: pharmaceutical information—ETHYOL [Internet]. Available from www.rxmed.
com.
51. Little M, Schipper M, Feng FY, Vineberg K, Cornwall C, Murdoch-Kinch C-A et al (2012)
Reducing xerostomia after chemo-IMRT for head-and-neck cancer: beyond sparing the
parotid glands. Int J Radiat Oncol Biol Phys 83(3):1007–1014
52. Nutting CM, Morden JP, Harrington KJ, Urbano TG, Bhide SA, Clark C et al (2011) Parotid-­
sparing intensity modulated versus conventional radiotherapy in head and neck cancer
(PARSPORT): a phase 3 multicentre randomised controlled trial. Lancet Oncol 12(2):127–136
53. Deasy JO, Moiseenko V, Marks L, Chao KC, Nam J, Eisbruch A (2010) Radiotherapy dose–
volume effects on salivary gland function. Int J Radiat Oncol Biol Phys 76(3):S58–S63
54. Eisbruch A (2011) Amifostine in the treatment of head and neck cancer: intravenous admin-
istration, subcutaneous administration, or none of the above. J Clin Oncol 29(2):119–121
55. Rudat V, Münter M, Rades D, Grötz KA, Bajrovic A, Haberkorn U et al (2008) The effect of
amifostine or IMRT to preserve the parotid function after radiotherapy of the head and neck
region measured by quantitative salivary gland scintigraphy. Radiother Oncol 89(1):71–80
56. Van Rij C, Oughlane-Heemsbergen W, Ackerstaff A, Lamers E, Balm A, Rasch C (2008)
Parotid gland sparing IMRT for head and neck cancer improves xerostomia related quality of
life. Radiat Oncol 3(1):1
57. Seikaly H, Jha N, McGaw T, Coulter L, Liu R, Oldring D (2001) Submandibular gland trans-
fer: a new method of preventing radiation-induced xerostomia. Laryngoscope
111(2):347–352
58. Jha N, Seikaly H, Harris J, Williams D, Sultanem K, Hier M et al (2009) Phase III random-
ized study: oral pilocarpine versus submandibular salivary gland transfer protocol for the
management of radiation-induced xerostomia. Head Neck 31(2):234–243
59. Sood AJ, Fox NF, O’Connell BP, Lovelace TL, Nguyen SA, Sharma AK et al (2014) Salivary
gland transfer to prevent radiation-induced xerostomia: A systematic review and meta-­
analysis. Oral Oncol 50(2):77–83
60. Liu XK, Su Y, Jha N, Hong MH, Mai HQ, Fan W et al (2011) Submandibular salivary gland
transfer for the prevention of radiation-induced xerostomia in patients with nasopharyngeal
carcinoma: 5-Year outcomes. Head Neck 33(3):389–395
61. Liu X, Zeng Z, Hong M, Cui N, Su Y, Mai H et al (2005) Primary effect of submandibular
salivary gland transfer in preventing radiation-induced xerostomia of nasopharyngeal carci-
noma. AiZheng 24(5):577–581
62. Wu F, Weng S, Li C, Sun J, Li L, Gao Q (2015) Submandibular gland transfer for the preven-
tion of postradiation xerostomia in patients with head and neck cancer: a systematic review
and meta-analysis. ORL 77(2):70–86
63. Zhang Y, Guo CB, Zhang L, Wang Y, Peng X, Mao C et al (2012) Prevention of radiation-­
induced xerostomia by submandibular gland transfer. Head Neck 34(7):937–942
64. Basu T, Laskar SG, Gupta T, Budrukkar A, Murthy V, Agarwal JP (2012) Toxicity with radio-
therapy for oral cancers and its management: a practical approach. J Cancer Res Ther 8(6):72
References 95

65. Mandel I, Katz R, Zengo A, Kutscher A, Greenberg R, Katz S et al (1967) The effect of
pharmacologic agents on salivary secretion and composition in man. I. Pilocarpine, atropine
and anticholinesterases. J Oral Ther Pharmacol 4(3):192–199
66. Fox PC, van der Ven PF, Baum BJ, Mandel ID (1986) Pilocarpine for the treatment of xero-
stomia associated with salivary gland dysfunction. Oral Surg Oral Med Oral Pathol
61(3):243–248
67. Atri RDA, Kaushal V (2014) Pilocarpine and prevention of radiation induced xerostomia in
HNSCC. J Cancer Prev Curr Res 1(3)
68. Scarantino C, LeVeque F, Swann RS, White R, Schulsinger A, Hodson DI et al (2006) Effect
of pilocarpine during radiation therapy: results of RTOG 97-09, a phase III randomized study
in head and neck cancer patients. J Support Oncol 4(5):252–258
69. Taweechaisupapong S, Pesee M, Aromdee C, Laopaiboon M, Khunkitti W (2006) Efficacy of
pilocarpine lozenge for post-radiation xerostomia in patients with head and neck cancer. Aust
Dent J 51(4):333–337
70. Horiot J-C, Lipinski F, Schraub S, Maulard-Durdux C, Bensadoun RJ, Ardiet JM et al (2000)
Post-radiation severe xerostomia relieved by pilocarpine: a prospective French cooperative
study. Radiother Oncol 55(3):233–239
71. Fisher J, Scott C, Scarantino CW, Leveque FG, White RL, Rotman M et al (2003) Phase III
quality-of-life study results: impact on patients’ quality of life to reducing xerostomia after
radiotherapy for head-and-neck cancer—RTOG 97-09. Int J Radiat Oncol Biol Phys
56(3):832–836
72. Lajtman Z, Krajina Z, Krpan D, Vincelj J, Borcić V, Popović-Kovacić J (1999) Pilocarpine in
the prevention of postirradiation xerostomia. Acta Med Croatica 54(2):65–67
73. Kim KH, Kim JY, Sung MW, Kim CW (1991) The effect of pilocarpine and atropine admin-
istration on radiation-induced injury of rat submandibular glands. Acta Otolaryngol
111(5):967–973
74. Haddad P, Karimi M (2002) A randomized, double-blind, placebo-controlled trial of con-
comitant pilocarpine with head and neck irradiation for prevention of radiation-induced xero-
stomia. Radiother Oncol 64(1):29–32
75. Pilocarpine hydrochloride [Internet]. Available from www.drug.com
76. Braga FPF, Lemos Junior CA, Alves FA, Migliari DA (2011) Acupuncture for the prevention
of radiation-induced xerostomia in patients with head and neck cancer. Braz Oral Res
25(2):180–185
77. S B. A skeptical look at low level laser therapy. Science base medicine wiki 2014.
78. Lončar B, Mravak Stipetić M, Baričević M, Risović D (2011) The effect of low-level laser
therapy on salivary glands in patients with xerostomia. Photomed Laser Surg
29(3):171–175
79. Lopes CO, Mas JRI, Zângaro RA (2006) Low level laser therapy in the prevention of
radiotherapy-­induced xerostomia and oral mucositis. Radiol Bras 39(2):131–136
80. Pavlić V (2012) The effects of low-level laser therapy on xerostomia (mouth dryness). Med
Pregl 65(5–6):247–250
81. Vidović Juras D, Lukač J, Cekić-Arambašin A, Vidović A, Canjuga I, Sikora M et al (2010)
Effects of low-level laser treatment on mouth dryness. Coll Antropol 34(3):1039–1043
82. Symptom Management Guidelines: XEROSTOMIA [Internet]. Available from www.bccan-
cer.bc.ca/nursin
83. Warde P, Kroll B, O’sullivan B, Aslanidis J, Tew-George E, Waldron J et al (2000) A phase II
study of Biotene in the treatment of postradiation xerostomia in patients with head and neck
cancer. Sup Care Cancer 8(3):203–208
84. Johnstone PA, Peng YP, May BC, Inouye WS, Niemtzow RC (2001) Acupuncture for
pilocarpine-­resistant xerostomia following radiotherapy for head and neck malignancies. Int
J Radiat Oncol Biol Phys 50(2):353–357
85. Chien T-J, Liu C-Y, Hsu C-H (2013) Integrating acupuncture into cancer care. J Trad
Complement Med 3(4):234
86. Braga F, Sugaya N, Hirota S, Weinfeld I, Magalhaes M, Migliari D (2007) The effect of acu-
puncture on salivary flow rates in patients with radiation-induced xerostomia. Minerva
Stomatol 57(7–8):343–348
96 7 Xerostomia

87. Johnstone PA, Niemtzow RC, Riffenburgh RH (2002) Acupuncture for xerostomia. Cancer
94(4):1151–1156
88. Blom M, Lundeberg T (2000) Long-term follow-up of patients treated with acupuncture for
xerostomia and the influence of additional treatment. Oral Dis 6(1):15–24
89. Blom M, Dawidson I, Fernberg J-O, Johnson G, Angmar-Månsson B (1996) Acupuncture
treatment of patients with radiation-induced xerostomia. Eur J Cancer B Oral Oncol
32(3):182–190
90. Blom M, Dawidson I, Angmar-Månsson B (1992) The effect of acupuncture on salivary flow
rates in patients with xerostomia. Oral Surg Oral Med Oral Pathol 73(3):293–298
91. Zhuang L, Yang Z, Zeng X, Zhua X, Chen Z, Liu L et al (2013) The preventive and therapeu-
tic effect of acupuncture for radiation-induced xerostomia in patients with head and neck
cancer: a systematic review. Integrat Cancer Therap 12:197–205
92. Jedel E (2005) Acupuncture in xerostomia—a systematic review. J Oral Rehabil
32(6):392–396
93. Gorsky M, Epstein JB, Parry J, Epstein MS, Le ND, Silverman S (2004) The efficacy of
pilocarpine and bethanechol upon saliva production in cancer patients with hyposalivation
following radiation therapy. Oral Surg Oral Med Oral Pathol Oral Radiol Endod
97(2):190–195
94. Villa A, Connell CL, Abati S (2015) Diagnosis and management of xerostomia and hyposali-
vation. Ther Clin Risk Manag 11:45
95. Wiseman LR, Faulds D (1995) Oral pilocarpine: a review of its pharmacological properties
and clinical potential in xerostomia. Drugs 49(1):143–155
96. Sangthawan D, Watthanaarpornchai S, Phungrassami T (2001) Randomized double blind,
placebo-controlled study of pilocarpine administered during head and neck irradiation to
reduce xerostomia. J Med Assoc Thail 84(2):195–203
97. Johnson JT, Ferretti GA, Nethery WJ, Valdez IH, Fox PC, Ng D et al (1993) Oral pilocarpine
for post-irradiation xerostomia in patients with head and neck cancer. N Engl J Med
329(6):390–395
98. Niedermeier W, Matthaeus C, Meyer C, Staar S, Müller R-P, Schulze H-J (1998) Radiation-­
induced hyposalivation and its treatment with oral pilocarpine. Oral Surg Oral Med Oral
Pathol Oral Radiol Endod 86(5):541–549
99. Aframian D, Helcer M, Livni D, Robinson S, Markitziu A, Nadler C (2007) Pilocarpine treat-
ment in a mixed cohort of xerostomic patients. Oral Dis 13(1):88–92
100. Chambers MS, Posner M, Jones CU, Biel MA, Hodge KM, Vitti R et al (2007) Cevimeline
for the treatment of postirradiation xerostomia in patients with head and neck cancer. Int
J Radiat Oncol Biol Phys 68(4):1102–1109
101. Wolff A, C Fox P, Porter S, T Konttinen Y (2012) Established and novel approaches for the
management of hyposalivation and xerostomia. Curr Pharm Des 18(34):5515–5521
102. Comparing systemic medications for treatment of dry mouth. [Internet]. Available from
www.medscape.com
103. Abbasi F, Farhadi S, Esmaili M (2013) Efficacy of pilocarpine and bromhexine in improving
radiotherapy-induced xerostomia. J Dent Res Dent Clin Dent Prospects 7(2):86
104. Epstein JB, Burchell JL, Emerton S, Le ND, Silverman S (1994) A clinical trial of bethanechol
in patients with xerostomia after radiation therapy: a pilot study. Oral Surg Oral Med Oral
Pathol 77(6):610–614
105. Ameri A, Heydarirad G, Mahdavi Jafari J, Ghobadi A, Rezaeizadeh H, Choopani R (2015)
Medicinal plants contain mucilage used in traditional Persian medicine (TPM). Pharm Biol
53(4):615–623
106. Ameri A, Heydarirad G, Rezaeizadeh H, Choopani R, Ghobadi A, Gachkar L (2016)
Evaluation of efficacy of an herbal compound on dry mouth in patients with head and neck
cancers a randomized clinical trial. J Evid Based Complementary Altern Med 21:30–33
107. Omori Y, Asari T, Maruyama K, Kusama H, Kojima M, Shibata N (2003) Effects of pilocar-
pine hydrochloride and cevimeline on submandibular/sublingual salivation in rat xerostomia
model produced by X-ray irradiation. Arzneimittelforschung 53(05):342–350
Loss of Taste
8

A reduction in taste sensitivity (hypogeusia), an absence of taste sensation (ageu-


sia), or a distortion of normal taste (dysgeusia) are well-known side effects in cancer
patients that receive radiation therapy to the head and neck areas, and it has been
reported in up to 100% of these patients [1–4]. The generic term of dysgeusia more
commonly refers to any alteration in taste perception [5].

8.1 Mechanism

There are four basic taste intensities: salt, sour, bitter, and sweet. In addition to these
basic tastes, a novel taste that is referred to by the Japanese word umami, which
means delicious, has come to be recognized as a “fifth taste.” Umami is found in a
diversity of foods (e.g., fish, meat, milk, tomato, and some vegetables), which is
created by the combination of glutamate with 5′-ribonucleotides [6–8].
The sense of taste is mediated by the taste buds. They are found in the oral cavity,
primarily on the dorsum of the tongue in the circumvallate, fungiform, and foliate
papillae and on the palate, lips, cheeks, pharynx, epiglottis, larynx, and upper part
of the esophagus [9, 10].
The taste bud is an onion-shaped epithelial structure with 50–100 tightly packed
cells, including taste receptor cells, supporting cells, and basal cells [11]. Each taste
bud opens to the epithelial surface via a small opening called the taste pore [12].
Taste receptor cells have short microvilli, which emerge from the apical region
(outer end) of the taste cells to a taste pit below the inner taste pore. Taste receptor
sites are located on membranes of microvilli. The microvilli are the portion of the
cell that is exposed to the oral cavity [13]. The pore enables molecules and ions
taken into the mouth to reach the receptor cells inside [12].
The taste receptor cells contact with afferent sensory neurons at their inner ends.

© Springer International Publishing AG 2017 97


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_8
98 8 Loss of Taste

A nutritional, or trophic, interaction (i.e., one cell emits a substance that a second
cell needs to grow) between the nerve fibers and taste buds exists. The interruption
of the nerve fibers results in the disappearance of the taste buds [12].
Each of the different tastes is perceived in an individual taste bud with spe-
cific receptor cells that respond to distinct chemical stimuli. Furthermore, the
diverse sites of the tongue surface are sensitive to a distinctive taste. For exam-
ple, the anterior (tip) tongue is most sensitive to sweet or salty stimuli, while the
lateral edges and posterior aspects of the tongue respond predominantly to sour
and bitter substances, respectively. However, there is a concept that each taste
cell and each site of the tongue surface may be sensitive to more than one taste
[14], and all four different tastes can be perceived in all areas where taste buds
are located [15].
The mechanisms involved in taste loss during radiation therapy are complex.
Functional loss of taste buds occurs following radiation therapy due to the taste-cell
microvilli or their membrane injuries. Membrane damage causes interruption of the
synaptic contacts, impairment of taste bud trophic function of the nerve, and taste
bud atrophy [12, 16, 17]. Functional loss of taste buds precedes cell loss, which
occurs later following radiation therapy [12, 18].
Direct mucosal and epithelial cell damage of radiation results in desquamation of
the surface epithelium. The epithelial thickness is increased, the taste pores are cov-
ered, and the total area of the taste pores is also decreased [14, 19].

8.2 Timing

Alteration in taste is an early and rapid response to radiation and often precedes
mucositis [20]. Increase in taste thresholds begins from treatment with as little as
2–4 Gy and rises exponentially with a cumulative dose of about 30 Gy (3 weeks),
2 Gy per fraction. Rate of loss then slows down as the patients’ acuity approaches
nil at >30 Gy [18]. Subjective complaints of taste also are started early after the
beginning of the treatment, approximately 1–2 weeks after the initiation of treat-
ment [3, 21, 22]. Perception of bitter and acid flavors is more susceptible to impair-
ment than perception of salty and sweet flavors [23].
The clinical impairment pattern of umami taste has been investigated. Umami
taste declines during the third week after the start of radiation therapy and improves
after treatment conclusion [6, 24].
Loss of taste is usually transient. Patients often experience normal or near-­normal
levels of taste within 1 year after radiation therapy, although it can sometimes take
a few years, or even a residual reduction in taste acuity may permanently remain
[20, 23, 25–32].
A permanent loss of taste can occur at the dose of about 60 Gy [28, 33]. However,
due to the adaptation of the patient to the sensory loss, a minority of patients com-
plain of taste loss [11].
8.5 Diagnosis 99

8.3 Risk Factors

The proportion of the tongue that is contained within the radiation fields and the irra-
diation dose delivered has a significant correlation with the taste loss [12, 15, 34, 35].
A correlation between the tongue area that irradiated and the taste sensation mostly
lost has been observed [18].
The presence of saliva plays a significant role in the normal taste acuity by trans-
port and solubility of gustatory stimulants and protection of the taste receptors.
Saliva production may be reduced by radiation therapy and affects taste sensitivity
[11, 13, 20, 26, 34, 36].
Other factors that have some influence on the incidence or severity of loss of
taste caused by irradiation are malnutrition or specific vitamin or mineral defi-
ciencies like zinc deficiency [37] and destruction of the taste buds by the tumor
[9]. Head and neck surgery by a reduction of the total number of taste buds or
around the chorda tympani or glossopharyngeal nerve [2, 38] can also affect
acuity of taste. Chemotherapy drugs [12, 39], oral mucositis [40], and infection
[5] can exacerbate radiation-induced dysgeusia. Systemic disease like liver and
kidney disorders, endocrine disorders, diabetes mellitus, psychological disor-
ders, and central nervous system disorders, smoking, and alcoholism can also
decrease taste [2, 41].

8.4 Symptoms

Irradiation of the taste buds typically leads to partial (hypogeusia) or complete


(ageusia) inability to taste or an abnormal taste (dysgeusia) [18]. These taste abnor-
malities lead to the decreased hedonic aspect of food intake, decreased appetite, and
reduced nutrient intake, leading to anorexia and weight loss [2, 18, 21, 37, 42].
The nature of the taste sensation modifies the volume and character of saliva.
Loss of taste and failure of adequate salivation may explain difficulty in swallowing
reported by some affected patients [37].

8.5 Diagnosis

Loss of taste may be a subjective response, which patients may indicate a presence
of any subjective awareness of it.
Subjective awareness of taste loss and the presence of any distress caused by taste
impairment (e.g., decreased enjoyment of food and appetite) can be assessed by
using taste questionnaires [2], and objective detection of taste loss and recognition of
the thresholds for each taste quality can be determined in each patient by the increase
in threshold above the upper limit of normal and the lowest concentration of a solute
that the patient distinguishes as different from water, respectively [9, 21, 43–45].
100 8 Loss of Taste

Table 8.1 CTCAE. V4.03 for taste


Definition
Grade 1 Altered taste but no change in diet
Grade 2 Altered taste with change in diet (e.g., oral supplements); noxious or unpleasant
taste; loss of taste

8.6 Scoring

Common Toxicity Criteria for Adverse Effect (CTCAE), version 4.03, scoring sys-
tem has just defined two grades for taste abnormalities [46] (Table 8.1).

8.7 Prevention

Prevention of taste loss can be obtained by the modification of radiation therapy


including normal tissue shielding or placement of these tissues outside the radiation
field by means of field arrangements or repositioning prostheses (e.g., tongue
depressor) [47] or advanced radiation therapy technique (e.g., intensity-modulated
radiotherapy) [48, 49].
The effect of amifostine on taste loss is inconclusive. The administration of ami-
fostine may produce reductions in the severity of taste loss. However, the total fre-
quency of dysgeusia can be more frequent among patients given amifostine than
among controls [50, 51]. Further studies are needed in this regard.
All actions for prevention of xerostomia may be effective in reducing radiation-­
induced taste loss [48] (see Chap. 7).
There are a lack of data and controversial results on the effects of zinc supplemen-
tation in prevention/treatment of taste alterations in cancer patients (see Sect. 8.8).

8.8 Management

In most instances, taste gradually returns to normal or near-normal levels within 1


year after radiation therapy. Because of this transitory aspect, there is usually no
need for special treatment [25], but clinicians can help their patients by offering
counseling to improve their distressing symptoms. Dietary counseling and patient
education can result in improved nutrition status and prevent weight loss.
Patients should be instructed about methods to increase taste, including prepar-
ing foods with strong taste and creating an attractive presentation of foods. Patients
should be encouraged to avoid the use of tobacco and/or alcohol and management
of hyposalivation and poor oral hygiene.
The assessment for taste alterations includes a thorough dietary history, eating
habits, current appetite and desire for food, and weight measuring for compari-
son to baseline [52]. Nutritional counseling estimates each patient’s current
nutritional status, calculates increase in energy and protein requirements to
8.8 Management 101

overcome deficits, and provides the therapeutic diet based on personal eating pat-
terns and preferences, which are adjusted to the individual’s needs. Dietary coun-
seling corrects patient diets with appropriate manipulation and consideration of
foods with appealing taste, color, and smell; oral nutritional supplements may be
added to the diet only in patients with inadequate food intake for more than
5 days or BMI < 18.5 [53, 54].
In the setting of significant weight loss (>2% loss in 1 week), patients should be
evaluated by a registered dietician [52]. A dietician provides an individualized and
intensive dietary counseling based on standard nutrition protocol, the Medical
Nutrition Therapy (Cancer/Radiation Oncology) protocol of the American Dietetic
Association (ADA), to maintain and/or improve a patient’s energy and protein
intake [55–57].
In general, 25–30 kilocalories per kilogram body weight per day and 1–1.5 g of
protein per kilogram per day are appropriate for those of normal weight. For those
that are hypermetabolic or need to gain weight, 30–35 kilocalories per kilogram or
greater and 1.5–2.5 g of protein per kilogram may be necessary [58].
Energy requirements can be calculated using various formulas, which give more
precise estimates of resting energy expenditure. The Harris-Benedict equation is
practical and reliable for measuring metabolic rate. The equation is used to estimate
the basal energy expenditure (BEE) based on weight, height, and age [59]. To esti-
mate daily energy requirements, basal requirements were multiplied by a 1.5 activ-
ity factor [60].

In men :
BEE = 66.5 + (13.75 ´ kg ) + ( 5.003 ´ cm ) - ( 6.775 ´ age ) .
In women :
BEE = 655.1 + ( 9.563 ´ kg ) + (1.850 ´ cm ) - ( 4.676 ´ age )

Zinc plays an important role in taste perception [61]. Zinc deficiency results in
structural changes in taste buds cells, changes in the number and size profile of taste
buds, and decrease in related nerve sensitivity [62, 63]. The effects of zinc supple-
mentation in prevention/treatment of taste alterations in cancer patients are contro-
versial. Some have found that the administration of zinc sulfate (45–50 mg orally
three times daily) in cancer patients that had received radiation therapy to the head
and neck region is an effective approach both in the prevention and correction of
taste abnormalities [2, 3, 21, 64]. However, some found no statistically significant
effect of zinc sulfate therapy on radiation-induced taste alterations [65]. Further
studies with longer follow-ups and with different doses of zinc supplementation are
needed in this regard.
The use of megestrol acetate (480 mg/day) during radiation therapy may be
effective in the improvement of loss of taste, appetite, and reversing malnutrition.
Further evaluation of its effect on radiation-related complications and patient out-
comes is needed [66].
102 8 Loss of Taste

References
1. Huang HY, Wilkie DJ, Schubert MM, Ting LL (2000) Symptom profile of nasopharyngeal
cancer patients during radiation therapy. Cancer Pract 8(6):274–281
2. Redda MGR, Allis S (2006) Radiotherapy-induced taste impairment. Cancer Treat Rev
32(7):541–547
3. Ripamonti C, Zecca E, Brunelli C, Fulfaro F, Villa S, Balzarini A et al (1998) A randomized,
controlled clinical trial to evaluate the effects of zinc sulfate on cancer patients with taste
alterations caused by head and neck irradiation. Cancer 82(10):1938–1945
4. Kearvell RKR, Preston R, Tanner P, Buck H, Hedges R (2004) Acute radiation toxicity assessment
of a 3-D conformal head and neck radiation treatment technique. Australas Radiol 48:358–363
5. Irune E, Dwivedi RC, Nutting CM, Harrington KJ (2014) Treatment-related dysgeusia in head
and neck cancer patients. Cancer Treat Rev 40(9):1106–1117
6. Shi HBMM, Umezaki T et al (2004) Irradiation impairment of umami taste in patients with
head and neck cancer. Auris Nasus Larynx 31:401–406
7. Schiffman S, Frey A, Luboski J, Foster M, Erickson R (1991) Taste of glutamate salts in young
and elderly subjects: role of inosine 5′-monophosphate and ions. Physiol Behav 49(5):843–854
8. Yamaguchi S, Ninomiya K (2000) Umami and food palatability. J Nutr 130(4):921S–926S
9. Maes AHI, Weltens C, Vandevelde G, Delaere P, Evers G, Van den Bogaert W (2002) De
Gustibus: time scale of loss and recovery of tastes caused by radiotherapy. Radiother Oncol
63(2):195–201
10. Schiffman SS, Gatlin CA (1993) Clinical physiology of taste and smell. Annu Rev Nutr
13(1):405–436
11. Feng P, Huang L, Wang H (2013) Taste bud homeostasis in health, disease, and aging. Chem
Senses 39:3–16
12. Nelson GM (1998) Biology of taste buds and the clinical problem of taste loss. Anat Rec
253(3):70–78
13. Matsuo R (2000) Role of saliva in the maintenance of taste sensitivity. Crit Rev Oral Biol Med
11(2):216–229
14. Mirza N, Machtay M, Devine PA, Troxel A, Abboud SK, Doty RL (2008) Gustatory impair-
ment in patients undergoing head and neck irradiation. Laryngoscope 118(1):24–31
15. Yamashita H, Nakagawa K, Nakamura N, Abe K, Asakage T, Ohmoto M et al (2006) Relation
between acute and late irradiation impairment of four basic tastes and irradiated tongue vol-
ume in patients with head-and-neck cancer. Int J Radiat Oncol Biol Phys 66(5):1422–1429
16. Shatzman AR, Mossman KL (1982) Radiation effects on bovine taste bud membranes. Radiat
Res 92(2):353–358
17. Conger AD, Wells MA (1969) Radiation and aging effect on taste structure and function.
Radiat Res 37(1):31–49
18. Conger AD (1973) Loss and recovery of taste acuity in patients irradiated to the oral cavity.
Radiat Res 53(2):338–347
19. Just T, Pau HW, Bombor I, Guthoff RF, Fietkau R, Hummel T (2005) Confocal microscopy of
the peripheral gustatory system: comparison between healthy subjects and patients suffering
from taste disorders during radiochemotherapy. Laryngoscope 115(12):2178–2182
20. Vissink A, Jansma J, Spijkervet F, Burlage F, Coppes R (2003) Oral sequelae of head and neck
radiotherapy. Crit Rev Oral Biol Med 14(3):199–212
21. Mossman KL, Henkin RI (1978) Radiation-induced changes in taste acuity in cancer patients.
Int J Radiat Oncol Biol Phys 4(7–8):663–670
22. Faramarzi E, Mahdavi R, Mohammad-Zadeh M, Ghaeammaghami J (2007) Incidence of eat-
ing problems, taste changes and food preference of cancer patients during radiotherapy. Res
J Biol Sciences 2(5):551–556
23. Toljanic JA, Saunders VW (1984) Radiation therapy and management of the irradiated patient.
J Prosthet Dent 52(6):852–858
References 103

24. Yamashita HNK, Hosoi Y, Kurokawa A, Fukuda Y, Matsumoto I, Misaka T, Abe K (2009)
Umami taste dysfunction in patients receiving radiotherapy for head and neck cancer. Oral
Oncol 45(3):e19–e23
25. Tomita Y, Osaki T (1990) Gustatory impairment and salivary gland pathophysiology in rela-
tion to oral cancer treatment. Int J Oral Maxillofac Surg 19(5):299–304
26. Beumer J, Curtis T, Harrison RE (1979) Radiation therapy of the oral cavity: sequelae and
management, part 1. Head Neck Surg 1(4):301–312
27. Phillips TL, Benak S (1972) Radiation modalities in treatment of cancer of the oral cavity.
J Prosthet Dent 27(4):413–418
28. Rubin RL, Doku HC (1976) Therapeutic radiology—the modalities and their effects on oral
tissues. J Am Dent Assoc 92(4):731–739
29. de Graeff A, de Leeuw JRJ, Ros WJ, Hordijk GJ, Blijham GH, Winnubst JA (2000) Long-term
quality of life of patients with head and neck cancer. Laryngoscope 110(1):98–106
30. Saito T, Miyake M, Kawamori J, Fukushima S, Furuhashi S, Yoshinobu T et al (2001) Buccal
mucosal cancer patient who failed to recover taste acuity after partial oral cavity irradiation.
Radiat Med 20(5):257–260
31. Toda Y (2002) Evaluation of xerostomia and taste disturbance after radiotherapy of patients
with head and neck lesion. Kurume Igakkai Zasshi 65(8–10):195–202
32. Sandow P, Hejrat-Yazdi M, Heft M (2006) Taste loss and recovery following radiation therapy.
J Dent Res 85(7):608–611
33. Stein JJ, James AG, King ER (1970) The management of the teeth, bone, and soft tissues in
patients receiving treatment for oral cancer. Am J Roentgenol 108(2):257–268
34. Fernando I, Patel T, Billingham L, Hammond C, Hallmark S, Glaholm J et al (1995) The effect
of head and neck irradiation on taste dysfunction: a prospective study. Clin Oncol 7(3):173–178
35. Umeyama M, Suzaki H (2001) Effects of radiotherapy on the sense of taste. Pract
Otorhinolaryngol (Basel) 94:617–625
36. Frank R, Herdly J, Philippe E (1965) Acquired dental defects and salivary gland lesions after
irradiation for carcinoma. J Am Dent Assoc 70(4):868–883
37. WD DW (1974) Abnormalities of taste as a remote effect of a neoplasm. Ann N Y Acad Sci
230(1):427–434
38. Urade M, Igarashi T, Sugi M, Matsuya T, Fukuda T (1987) Functional recovery of swallowing,
speech, and taste in an oral cancer patient with subtotal glossectomy. J Oral Maxillofac Surg
45(3):282–285
39. Rehwaldt M, Wickham R, Purl S, Tariman J, Blendowski C, Shott S et al (2009) Self-care
strategies to cope with taste changes after chemotherapy. Oncol Nurs Forum 36:E47–E56
40. Biswa MB (2008) Current trends in the management of oral mucositis related to cancer treat-
ment Malays. J Med Sci 15(3):4–13
41. Su N, Ching V, Grushka M (2013) Taste disorders: a review. J Can Dent Assoc 79:d86
42. Bolze MS, Fosmire G, Stryker J, Chung C, Flipse B (1982) Taste acuity, plasma zinc levels,
and weight loss during radiotherapy: a study of relationships. Radiology 144(1):163–169
43. Henkin RI, Gill JR Jr, Bartter FC (1963) Studies on taste thresholds in normal man and in
patients with adrenal cortical insufficiency: the role of adrenal cortical steroids and of serum
sodium concentration. J Clin Investig 42(5):727
44. Henkin RI, Schechter PJ, Hoye R, Mattern CF (1971) Idiopathic hypogeusia with dysgeusia,
hyposmia, and dysosmia: a new syndrome. JAMA 217(4):434–440
45. Mossman K (1986) Gustatory tissue injury in man: radiation dose response relationships and
mechanisms of taste loss. Br J Cancer Suppl 7:9
46. Common Terminology Criteria for Adverse Events (CTCAE) [Internet] (2010) August 6. Available
from ­evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-­14_QuickReference_8.5x11.pdf
47. Vissink ABF, Spijkervet FK, Jansma J, Coppes RP (2003) Prevention and treatment of the
consequences of head and neck radiotherapy. Crit Rev Oral Biol Med 14(3):213–225
104 8 Loss of Taste

48. Epstein JB, Barasch A (2010) Taste disorders in cancer patients: pathogenesis, and approach
to assessment and management. Oral Oncol 46(2):77–81
49. McLaughlin L, Mahon S (2014) A meta-analysis of the relationship among impaired taste and
treatment, treatment type, and tumor site in head and neck cancer treatment survivors. Oncol
Nurs Forum 41:E194–E202
50. Büntzel J, Schuth J, Küttner K, Glatzel M (1998) Radiochemotherapy with amifostine cyto-
protection for head and neck cancer. Support Care Cancer 6(2):155–160
51. Komaki R, Lee JS, Milas L, Lee HK, Fossella FV, Herbst RS et al (2004) Effects of amifostine
on acute toxicity from concurrent chemotherapy and radiotherapy for inoperable non–small-­
cell lung cancer: report of a randomized comparative trial. Int J Radiat Oncol Biol Phys
58(5):1369–1377
52. Grant M, Kravits K (2000) Symptoms and their impact on nutrition. Semin Oncol Nurs
16:113–121
53. Ravasco P, Monteiro-Grillo I, Marques Vidal P, Camilo ME (2005) Impact of nutrition on
outcome: a prospective randomized controlled trial in patients with head and neck cancer
undergoing radiotherapy. Head Neck 27(8):659–668
54. Ravasco P, Camilo M, Monteiro GI (2001) Does nutrition care enhances quality of life in can-
cer patients undergoing radiotherapy. Clin Nutr 20(suppl 3):30
55. The American Dietetic Association and Morrison Health Care (1998) Medical nutrition ther-
apy across the continuum of care. 2nd ed. The American Dietetic Association, Chicago, IL
56. Association AD (1999) Medical nutrition therapy protocols: an introduction. J Am Diet Assoc
99(3):351
57. Nutrition support in adults: oral supplements, enteral and parenteral feeding: The Royal
College of Surgeons of England (2005)
58. Theresa AF (2008) Enteral nutrition for patients with head and neck cancer. Today’s Dietitian
10(6):46
59. Colasanto JM, Prasad P, Nash MA, Decker RH, Wilson LD (2005) Nutritional support of
patients undergoing radiation therapy for head and neck cancer. Oncology (Williston Park)
19(3):371–379; discussion 80–2, 87
60. Ravasco P, Monteiro-Grillo I, Vidal PM, Camilo ME (2005) Dietary counseling improves
patient outcomes: a prospective, randomized, controlled trial in colorectal cancer patients
undergoing radiotherapy. J Clin Oncol 23(7):1431–1438
61. Keast RS (2003) The effect of zinc on human taste perception. J Food Sci 68(5):1871–1877
62. Chou H-C, Chien C, Huang H, Lu K (2001) Effects of zinc deficiency on the vallate papillae
and taste buds in rats. J Formo Med Associ 100(5):326–335
63. Komai M, Goto T, Suzuki H, Takeda T, Furukawa Y (2000) Zinc deficiency and taste dysfunc-
tion; contribution of carbonic anhydrase, a zinc-metalloenzyme, to normal taste sensation.
Biofactors 12(1–4):65–70
64. Najafizade N, Hemati S, Gookizade A, Berjis N, Hashemi M, Vejdani S et al (2013) Preventive
effects of zinc sulfate on taste alterations in patients under irradiation for head and neck can-
cers: a randomized placebo-controlled trial. J Res Med Sci 18(2):123–126
65. Halyard MY, Jatoi A, Sloan JA, Bearden JD, Vora SA, Atherton PJ et al (2007) Does zinc
sulfate prevent therapy-induced taste alterations in head and neck cancer patients? Results of
phase III double-blind, placebo-controlled trial from the North Central Cancer Treatment
Group (N01C4). Int J Radiat Oncol Biol Phys 67(5):1318–1322
66. Erkurt E, Erkisi M, Tunali C (2000) Supportive treatment in weight losing cancer patients due
to the additive adverse effects of radiation treatment. J Exp Clin Cancer Res 19:431–439
Laryngeal Edema
9

Laryngeal edema occurs when the larynx is included in the treatment field and may
impact the voice. Laryngeal edema accompanied by hoarseness is a common acute
side effect in radiation therapy of laryngeal and hypopharyngeal carcinoma and
whenever the neck is irradiated [1]; however, no studies have reported its incidence
as an acute side effect. It has been estimated that 15–59% of patients with head and
neck cancers develop grade 2 or higher laryngeal edema within 2 years after radia-
tion therapy [2].

9.1 Mechanism

Ionizing radiation generates free radicals and produces various reactive oxygen
species (ROS) that result in DNA damage and changes in the local microenviron-
ment through the activation of cytokine cascades and influx of inflammatory cells
[3, 4]. These processes cause inflammation, hyperemia, and erythema of the
laryngeal mucosa.
The larynx has many mixed serous-mucinous-type glands that lubricate the lar-
ynx with thin mucus secretion and is essential to phonation. Radiation induces atro-
phic changes in laryngeal glands leading to changes in the quantity and quality of
secretions, poor lubrication of the vocal folds, and subsequent voice problems [5, 6].

9.2 Timing

Laryngeal edema may occur during the first 2–3 weeks of radiation therapy and
continue to increase to the end of treatment. Recovery begins 3 weeks after treat-
ment completion and may require 6–12 months to subside.

© Springer International Publishing AG 2017 105


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_9
106 9 Laryngeal Edema

9.3 Risk Factors

The incidence of acute laryngeal edema increases with total dose, field size, tumor
stage, smoking, supraglottic laryngectomy, and chemotherapy administration [2,
7–10].

9.4 Symptoms

In the acute phase, laryngeal edema related to radiation causes phonation dysfunc-
tion (Fig. 9.1) that rarely is severe enough to cause serious dysphagia or compro-
mise the airway [11]. Dryness of the laryngeal mucosa makes it sensitive to develop
referral ear or throat pain.

9.5 Scoring

RTOG Common Toxicity Criteria for acute laryngeal side effects is based on clini-
cal criteria and has been shown in Table 9.1 [12].

Fig. 9.1 Laryngeal edema 7 days after full dose 3DCRT for recurrent true vocal cord SCC

Table 9.1 RTOG common toxicity criteria for laryngeal toxicity criteria
Definition
Grade 0 No change over baseline
Grade 1 Mild or intermittent hoarseness/cough not requiring antitussive/erythema of mucosa
Grade 2 Persistent hoarseness but able to vocalize/referred ear pain, sore throat, patchy
fibrinous exudate, or mild arytenoid edema not requiring narcotic/cough requiring
antitussive
Grade 3 Whispered speech, throat pain, or referred ear pain requiring narcotic/confluent
fibrinous exudate, marked arytenoid edema
Grade 4 Marked dyspnea, stridor, or hemoptysis with tracheostomy or intubation necessary
References 107

9.6 Prevention

The volume of larynx in radiation field receiving high dose of radiation should be
kept as low as possible to minimize the edema. The investigators suggested that the
percentage of laryngeal volume receiving more than 50 Gy and the mean laryngeal
dose should be ideally constrained to less than 27% and 43.5 Gy, respectively [13].
Smoking and alcohol consumption will increase the risk of severe laryngeal edema,
and patients should be encouraged to cease smoking and alcohol consumption.

9.7 Management

Exacerbating behavior such as smoking and alcohol consumption should be discon-


tinued. Voice rest is recommended to all patients.
In patients with moderate to severe or persistent laryngeal edema unresponsive
to conservative treatment methods, treatment with steroids and occasionally antibi-
otics may be necessary [2].
In patients with severe laryngeal edema leading to compromise airway, tempo-
rary tracheostomy should be considered [10].

References
1. Mardini G, Salgado C, Chen H (2010) Esophagus and hypopharyngeal reconstruction. Semin
Plast Surg 24(2):127–136
2. Bae JS, Roh JL, Lee SW, Kim SB, Kim JS, Lee JH, Choi SH, Nam SY, Kim SY (2012)
Laryngeal edema after radiotherapy in patients with squamous cell carcinomas of the larynx
and hypopharynx. Oral Oncol 48(9):853–858
3. Barnett G, West C, Dunning A, Elliott R, Coles C, Pharoah P, Burnet N (2009) Normal tissue reac-
tions to radiotherapy towards tailoring treatment dose by genotype. Nat Rev Cancer 9(2):134–142
4. Di Maggio F, Minafra L, Forte GI, Cammarata F, Lio D, Messa C, Gilardi M, Bravatà V (2015)
Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 12:14
5. Lazarus C (2009) Effects of chemoradiotherapy on voice and swallowing. Surgery 17(3):172–178
6. Sato K, Nakashima T (2008) Effect of irradiation on the human laryngeal glands. Ann Otol
Rhinol Laryngol 117(10):734–739
7. Fu KK, Woodhouse RJ, Quivey JM, Phillips TL, Dedo HH (1982) The significance of laryn-
geal edema following radiotherapy of carcinoma of the vocal cord. Cancer 49(4):655–658
8. Sanguineti G, Adapala P, Endres EJ et al (2007) Dosimetric predictors of laryngeal edema. Int
J Radiat Oncol Biol Phys 68(3):741–749
9. KK F, Woofhouse RJ, Quivey JM et al (1982) The significance of laryngeal edema following
radiotherapy of carcinoma of the vocal cord. Cancer 49:6555–6558
10. Mendenhall WM, Parsons JT, Stringer SP, Cassisi NJ, Million RR (1990) The role of radiation
therapy in laryngeal cancer. CA Cancer J Clin 40(3):150–165
11. Cèfaro GA, Genovesi D, Perez CA (2013) Delineating organs at risk in radiation therapy.
Springer Verlag Italia
12. Cox JD et al (1995) Toxicity criteria of the radiation therapy oncology group (RTOG) and the
european organization for research and treatment of cancer (EORTC). Int J Radiat Oncol Biol
Phys 31(5):1341–1346
13. Rancati T, Schwarz M, Allen AM, Feng F, Popovtzer A, Mittal B, Eisbruch A (2010) Radiation
dose-volume effects in the larynx and pharynx. Int J Radiat Oncol Biol Phys 76:S64–S69
Radiation Pneumonitis
10

Radiation pneumonitis (RP), the acute manifestation of radiation-induced lung


injury, is one of the main dose-limiting toxicities among patients receiving thoracic
radiation therapy. It has been reported to occur in 13–37% of patients receiving
definitive external beam radiation therapy for lung cancer [1]. Radiation pneumoni-
tis development following radiation therapy for other cancers actually depends on
lung volume, which is included in various irradiated volumes and dose parameters.
Some other factors such as smoking, background lung disease, or combined modal-
ity treatments are also important risk factors for radiation-induced pneumonitis. The
incidence of radiation pneumonitis is lower in Hodgkin disease (3%) [2] and breast
cancer patients (1%) [3] compared with lung cancer patients. Diagnosis and indica-
tions for therapeutic intervention are in a major controversy that will be discussed
in the next few paragraphs.

10.1 Mechanism

The first changes that are induced by radiation are increased vascular permeability
and exudation of proteinaceous material in the alveolar space [4]. Capillary endo-
thelial cells are very sensitive to ionizing radiation, and their damages are mani-
fested by detachment of cells from their basement membrane, which is followed by
blood flow turbulence and thrombosis formation. Following endothelial cell injury,
capillary permeation increases, fibrin-rich exudate leaks into the alveoli, and a hya-
line membrane is formed, which impairs gas exchange. Ionizing radiation can also
damage alveolar cells, which are manifested by depletion of type I pneumocytes
and hyperplasia of type II pneumocytes, in the context of the alveolar epithelium
regeneration process [5]. On the other hand, type II pneumocytes are also known to
be damaged by radiation therapy, resulting in release of surfactant into the alveolar
space and detachment of the pneumocytes from their basement membrane [6].
Later, the alveolar exudate clears, fibroblasts migrate into the alveolar walls, and the
alveolar septa are thickened.

© Springer International Publishing AG 2017 109


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_10
110 10 Radiation Pneumonitis

Fig. 10.1 Radiation-­


induced pneumonitis in a
patient with bulky Hodgkin
disease at 2 months after
mediastinal radiation
therapy

10.2 Timing

Although pathologic changes in lung tissue occur in the initial 24–48 h after radia-
tion, those changes are undetectable both clinically and radiologically. Radiation
pneumonitis occurs typically within 6 months after a course of radiation with a peak
onset at 1–3 months but may be seen as early as 1 week, especially in patients
receiving high total dose [7]. In some cases, no symptoms are present, and the diag-
nosis is made by imaging alone (Fig. 10.1).

10.3 Risk Factors

10.3.1 Volume and Dose Parameters

Nontarget lung dose and irradiated volume should be lowered as much as possible.
Different dose and volume parameters (mean lung dose, V5, V10, D15, V20, etc.)
have been evaluated to predict probabilities of radiation pneumonitis, but no defini-
tive dose and volume limits have been accepted as an optimal limit or parameter.
The percentage of lung volume exceeding 20 Gy (V20), percentage of lung volume
exceeding 30 Gy (V30), and mean lung dose (MLD) are the most predictive param-
eters evaluated in several studies. It has been shown that the risk of radiation pneu-
monitis is less than 20% when MLD is less than 20 Gy [8], less than 8% when V20
is 31% or lower [9], and less than 6% when V30 is 18% or lower [10]. These param-
eters are in respect of both lung volumes; however, a significant correlation was
recently found between these parameters when ipsilateral lung volume is consid-
ered and radiation pneumonitis rates have been reported [11].

10.3.2 Fractionation Schedule

The use of twice-daily fractionation has been shown to reduce the risk of radiation
pneumonitis compared with administration of the same total daily dose as a single
fraction [12].
10.4 Symptoms 111

Most dose volume parameters have been studied in areas of conventional frac-
tionated radiotherapy of lung cancer, and with increasing use of stereotactic body
radiation therapy with a large fraction size for lung cancer, well-designed studies
are needed to define new parameters for radiation-induced pneumonitis.

10.3.3 Chemotherapy/Hormone Therapy

The administration of chemotherapy concurrently or before radiation therapy


appears to increase the risk of radiation pneumonitis compared to radiation
therapy alone [13, 14]. It has been shown that concurrent taxane-based chemo-
radiation therapy increases the radiation pneumonitis risk more than cisplatin-
based chemotherapy regimens [15]. Additionally, chemotherapy administration
with certain agents (e.g., taxanes, anthracyclines, gemcitabine, etoposide, or
vinorelbine), following radiation therapy, can cause radiation-induced pneu-
monitis that is an inflammatory reaction within the previously treated radiation
field [16].
There is conflicting data about correlation of hormonal therapy including tamox-
ifen and aromatase inhibitors and radiation pneumonitis risk. Some found no posi-
tive effect of these agents on radiation pneumonitis risk; however, some reported a
significant correlation [14, 17, 18].

10.3.4 Smoking

There are also different results reported about smoking effects on radiation pneumo-
nitis risks [10, 19]. Surprisingly, some have shown that active smokers had a lower
frequency of radiation-induced pneumonitis.

10.3.5 Other Factors

Adequate data are not available for some variables including age, sex, tumor site,
Karnofsky performance status, comorbid lung disease, pulmonary function test, and
biological markers such as plasma cytokine levels and transforming growth factor
beta 1 (TGF-β1).

10.4 Symptoms

Dyspnea is the most common symptom, occurring in as many as 90% of cases.


Dyspnea is frequently accompanied by a dry cough, which occurs in about 50–60%
of cases. A low-grade fever is occasionally reported, which can be more pronounced
in severe cases. Some patients complain of chest pain with breathing, malaise, and
weight loss. Sensation of chest fullness usually develops 1–3 months after comple-
tion of radiation therapy.
112 10 Radiation Pneumonitis

10.5 Diagnosis

Radiation pneumonitis is a diagnosis of exclusion. Physical findings are usually not


prominent, but occasionally moist crackles, a pleural friction rub, or evidence of
consolidation may be present. There is no specific lab test to predict the develop-
ment of RP. Some patients might have a mild polymorphonuclear leukocytosis,
elevated ESR, serum LDH, and CRP, but these findings are nonspecific.
Chest X-ray and computed tomography are the most commonly used modalities
for assessing RP. The most common finding on CXR in early phases is perivascular
haziness, which often progresses to patchy alveolar-filling densities [20]. Pleural
effusions or atelectases are also sometimes seen. These changes occur shortly after
completion of radiation therapy, peak at 6 months, and become stable by 12 months.
One of the most characteristic features of radiation pneumonitis and fibrosis is that
these radiologic changes are confined to the outlines of the field of radiation
(Fig. 10.1). However, the use of oblique beam angles and the development of newer
irradiation techniques such as three-dimensional conformal radiation therapy and
intensity-modulated radiation therapy can result in an unusual distribution of these
findings [21].
Computed tomography (CT) is more sensitive than chest radiograph in detecting
subtle lung injury following radiation treatment but is not required to make the
diagnosis of RP. The most common findings on CT are ground-glass opacities in the
acute phase and traction bronchiectasis, volume loss, and consolidation in the late
phase [22].
Pulmonary function test (PFT) in patients with radiation pneumonitis usually
reveals a reduction in lung volumes and diffusing capacity of the lungs for carbon
monoxide (DLCO), but these changes are nonspecific [23].
Bronchoalveolar lavage and transbronchial biopsy are not useful in the diagnosis
of radiation pneumonitis but can be used to exclude other causes of pulmonary
infiltrates.

10.6 Scoring

Common Toxicity Criteria for Adverse Effect (CTCAE) (Table 10.1) [24] and
Radiation Therapy Oncology Group (RTOG) (Table 10.2) [25] have defined five
grades for radiation-induced pneumonitis with some differences.

Table 10.1 CTCAE v4.03 for pneumonitis


Grade 1 Asymptomatic; clinical or diagnostic observations only; intervention not indicated
Grade 2 Symptomatic; medical intervention indicated; limiting instrumental activity of daily
life (ADL)
Grade 3 Severe symptoms; limiting self-care ADL; oxygen indicated
Grade 4 Life-threatening respiratory compromise; urgent intervention indicated (e.g.,
tracheotomy or intubation)
Grade 5 Death
10.7 Prevention 113

Table 10.2 RTOG grading for radiation-induced pneumonitis


Grade 1 Asymptomatic or mild symptoms (dry cough); slight radiographic changes
Grade 2 Moderate symptomatic fibrosis or pneumonitis (severe cough); low-grade fever;
patchy radiographic changes
Grade 3 Severe symptomatic fibrosis or pneumonitis; dense radiographic changes
Grade 4 Severe respiratory insufficiency; continuous oxygen therapy/assisted ventilation
Grade 5 Death

10.7 Prevention

There are several agents reported to have preventive effects on radiation pneumonitis.
Amifostine is a thio-organic prodrug that is believed to scavenge harmful free
radicals and shield normal tissues from the toxic effects of chemotherapy and radio-
therapy. It is approved for use as a cytoprotectant, relieving problems of dry mouth
(xerostomia) in patients with head and neck cancers undergoing radiotherapy.
Multiple randomized trials have assessed the role of amifostine in preventing RP,
with contradicting results [26–28]. The largest study was performed by the Radiation
Therapy Oncology Group; the incidence of grade ≥3 pulmonary toxicity was not
statistically different between patients receiving amifostine and those that did not
[29]. Current guidelines do not advise amifostine for prevention of radiation-­
induced pneumonitis.
Pentoxifylline is an immunomodulator that is used primarily for patients with
intermittent claudication. In one small randomized study, the use of pentoxifylline
(400 mg three times daily) resulted in a noticeable reduction in grade 2 or 3 pulmo-
nary toxicity (20% vs. 50%), as well as the measured diffusing capacity after
6 months of follow-up [30].
Captopril is an angiotensin-converting enzyme inhibitor that has been shown to
reduce the development of radiation-induced fibrosis in rats [31], although no such
protective effect has been demonstrated in humans.
There is lack of high-quality evidence on the management of radiation pneumo-
nitis, and no prospective controlled studies have evaluated the efficacy of current
therapies for radiation pneumonitis in humans.
Treatment for acute radiation pneumonitis is mainly supportive. Patients that are
asymptomatic with radiographic abnormalities do not require treatment, and patients
with mild symptoms are generally treated with cough suppressants including
codeine and benzonatate.
Despite the lack of solid evidence, oral corticosteroids have been the mainstay of
therapy for radiation pneumonitis, often with dramatic results. Although the starting
dose and tapering schedule are undefined, based upon clinical experience, dose of
approximately 60 mg or 1 mg/kg of prednisone is given for 1–2 weeks, followed by
a slow taper over 4–8 weeks. The dose is tapered slowly because some patients
experience a rebound pneumonitis with a faster tapering schedule. In some cases,
symptoms and radiographic abnormalities tend to recur with discontinuation of
therapy, and patients might need to maintain a low-dose prednisone schedule for
more extended periods of time [32].
114 10 Radiation Pneumonitis

When prednisone dose exceeds 20 mg a day for greater than a month, prophy-
laxis for Pneumocystis pneumonia is recommended. It is worthy to mention that
established fibrosis is irreversible and will not improve with glucocorticoid
therapy.
Azathioprine and cyclosporine may be considered in patients that do not tolerate
glucocorticoids or that have disease refractory to glucocorticoid therapy based on
small case series [33].

References
1. Rodrigues G, Lock M, D’Souza D, Yu E, Van Dyk J (2004) Prediction of radiation pneumonitis
by dose-volume histogram parameters in lung cancer—a systematic review. Radiother Oncol
71(2):127–138
2. Koh E-S, Sun A, Tran TH, Tsang R, Pintilie M, Hodgson DC et al (2006) Clinical dose-volume
histogram analysis in predicting radiation pneumonitis in Hodgkin’s lymphoma. Int J Radiat
Oncol Biol Phys 66(1):223–228
3. Taghian AG, Assaad SI, Niemierko A, Kuter I, Younger J, Schoenthaler R et al (2001) Risk of
pneumonitis in breast cancer patients treated with radiation therapy and combination chemo-
therapy with paclitaxel. J Natl Cancer Inst 93(23):1806–1811
4. Tsoutsou PG, Koukourakis MI (2006) Radiation pneumonitis and fibrosis: mechanisms under-
lying its pathogenesis and implications for future research. Int J Radiat Oncol Biol Phys
66(5):1281–1293
5. Coggle J (1987) Proliferation of type II pneumonocytes after X-irradiation. Int J Radiat Biol
Relat Stud Phys Chem Med 51(3):393–399
6. Rubin P, Shapiro D, Finkelstein J, Penney D (1979) The early release of surfactant following
lung irradiation of alveolar type II cells. Int J Radiat Oncol Biol Phys 5:55–56
7. Choi YW, Munden RF, Erasmus JJ, Park KJ, Chung WK, Jeon SC et al (2004) Effects of radia-
tion therapy on the lung: radiologic appearances and differential diagnosis 1. Radiographics
24(4):985–997
8. Marks LB, Bentzen SM, Deasy JO, Bradley JD, Vogelius IS, El Naqa I et al (2010) Radiation
dose-volume effects in the lung. Int J Radiat Oncol Biol Phys 76(3):S70–S76
9. Graham MV, Purdy JA, Emami B, Harms W, Bosch W, Lockett MA et al (1999) Clinical dose-­
volume histogram analysis for pneumonitis after 3D treatment for non-small cell lung cancer
(NSCLC). Int J Radiat Oncol Biol Phys 45(2):323–329
10. Hernando ML, Marks LB, Bentel GC, Zhou S-M, Hollis D, Das SK et al (2001) Radiation-­
induced pulmonary toxicity: a dose-volume histogram analysis in 201 patients with lung can-
cer. Int J Radiat Oncol Biol Phys 51(3):650–659
11. Ramella S, Trodella L, Mineo TC, Pompeo E, Stimato G, Gaudino D et al (2010) Adding
ipsilateral V 20 and V 30 to conventional dosimetric constraints predicts radiation pneumonitis
in stage IIIA-B NSCLC treated with combined-modality therapy. Int J Radiat Oncol Biol Phys
76(1):110–115
12. Roach M, Gandara DR, Yuo H-S, Swift PS, Kroll S, Shrieve DC et al (1995) Radiation pneu-
monitis following combined modality therapy for lung cancer: analysis of prognostic factors.
J Clin Oncol 13(10):2606–2612
13. Lingos TI, Recht A, Vicini F, Abner A, Silver B, Harris JR (1991) Radiation pneumonitis in
breast cancer patients treated with conservative surgery and radiation therapy. Int J Radiat
Oncol Biol Phys 21(2):355–360
14. Agrawal S (2013) Clinical relevance of radiation pneumonitis in breast cancers. South Asian
J Cancer 2(1):19–20
15. Palma DA, Senan S, Tsujino K, Barriger RB, Rengan R, Moreno M et al (2013) Predicting
radiation pneumonitis after chemoradiation therapy for lung cancer: an international individual
patient data meta-analysis. Int J Radiat Oncol Biol Phys 85(2):444–450
References 115

16. Ding X, Ji W, Li J, Zhang X, Wang L (2011) Radiation recall pneumonitis induced by chemo-
therapy after thoracic radiotherapy for lung cancer. Radiat Oncol 6(1):1
17. Dörr W, Bertmann S, Herrmann T (2005) Radiation induced lung reactions in breast cancer
therapy. Strahlenther Onkol 181(9):567–573
18. Kubo A, Osaki K, Kawanaka T, Furutani S, Ikushima H, Nishitani H (2009) Risk factors for
radiation pneumonitis caused by whole breast irradiation following breast-conserving surgery.
J Med Invest 56(3, 4):99–110
19. Johansson S, Bjermer L, Franzen L, Henriksson R (1998) Effects of ongoing smoking on the
development of radiation-induced pneumonitis in breast cancer and oesophagus cancer
patients. Radiother Oncol 49(1):41–47
20. Monson JM, Stark P, Reilly JJ, Sugarbaker DJ, Strauss GM, Swanson SJ et al (1998) Clinical
radiation pneumonitis and radiographic changes after thoracic radiation therapy for lung car-
cinoma. Cancer 82(5):842–850
21. Aoki T, Nagata Y, Negoro Y, Takayama K, Mizowaki T, Kokubo M et al (2004) Evaluation of
lung injury after three-dimensional conformal stereotactic radiation therapy for solitary lung
tumors: CT appearance 1. Radiology 230(1):101–108
22. Ikezoe J, Takashima S, Morimoto S, Kadowaki K, Takeuchi N, Yamamoto T et al (1988) CT
appearance of acute radiation-induced injury in the lung. Am J Roentgenol 150(4):765–770
23. Miller KL, Zhou S-M, Barrier RC, Shafman T, Folz RJ, Clough RW et al (2003) Long-term
changes in pulmonary function tests after definitive radiotherapy for lung cancer. Int J Radiat
Oncol Biol Phys 56(3):611–615
24. U.S. Department of Health and Human Services and National Cancer Institute. Common
Terminology Criteria for Adverse Events (CTCAE). Version 403: June 14, 2010. 2014
25. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
26. Antonadou D, Petridis A, Synodinou M, Throuvalas N, Bolanos N, Veslemes M et al (eds)
(2003) Amifostine reduces radiochemotherapy-induced toxicities in patients with locally
advanced non-small cell lung cancer. Semin Oncol 30(6 Suppl 18):2–9
27. Movsas B, Scott C, Langer C, Werner-Wasik M, Nicolaou N, Komaki R et al (2005)
Randomized trial of amifostine in locally advanced non-small-cell lung cancer patients receiv-
ing chemotherapy and hyperfractionated radiation: Radiation therapy oncology group trial
98-01. J Clin Oncol 23(10):2145–2154
28. Sasse AD, Clark LG, Sasse EC, Clark OA (2006) Amifostine reduces side effects and improves
complete response rate during radiotherapy: results of a meta-analysis. Int J Radiat Oncol Biol
Phys 64(3):784–791
29. Antonadou D, Coliarakis N, Synodinou M, Athanassiou H, Kouveli A, Verigos C et al (2001)
Randomized phase III trial of radiation treatment ± amifostine in patients with advanced-stage
lung cancer. Int J Radiat Oncol Biol Phys 51(4):915–922
30. Ozturk B, Egehan I, Atavci S, Kitapci M (2004) Pentoxifylline in prevention of radiation-­
induced lung toxicity in patients with breast and lung cancer: a double-blind randomized trial.
Int J Radiat Oncol Biol Phys 58(1):213–219
31. Ward WF, Molteni A, Ts’ao CH (1989) Radiation-induced endothelial dysfunction and fibrosis
in rat lung: modification by the angiotensin converting enzyme inhibitor CL242817. Radiat
Res 117(2):342–350
32. Katayama N, Sato S, Katsui K, Takemoto M, Tsuda T, Yoshida A et al (2009) Analysis of fac-
tors associated with radiation-induced bronchiolitis obliterans organizing pneumonia syn-
drome after breast-conserving therapy. Int J Radiat Oncol Biol Phys 73(4):1049–1054
33. Muraoka T, Bandoh S, Fujita J, Horiike A, Ishii T, Tojo Y et al (2002) Corticosteroid refractory
radiation pneumonitis that remarkably responded to cyclosporin A. Intern Med
41(9):730–733
Pericarditis
11

Pericarditis is the most common acute presentation of radiation-induced heart dis-


ease. The incidence of pericarditis in patients with irradiation for different targets
within and around the thorax is estimated around 5% on average and even less;
indeed, with improvement in radiation treatment techniques, its incidence has
decreased from 25% to 2% [1–3]. Clinically significant pericarditis is observed just
in a limited percentage of patients after mediastinal radiation therapy [4].
Radiation-induced pericarditis most commonly occurs in Hodgkin and non-­
Hodgkin lymphoma and breast cancer patients [2]. It’s probably related to the
incidence and survival of these malignancies. The risk of pericarditis is higher
when the target volume for radiation therapy is in the mediastinum, and it will
decrease with increasing distance between the target volume and the mediastinum
and heart. Site and size of mediastinal lymphadenopathies are important factors in
the incidence of pericarditis for lymphoma patients. With effective chemothera-
pies, target volumes and pericarditis rate have decreased in patients with lym-
phoma after mediastinal radiation. Besides these, pericardial manifestations have
also decreased in lymphoma as well as breast cancer by radiation technique
improvements [5, 6].
Pericardial effusion has been reported in one third of esophageal cancer patients
following definitive chemoradiotherapy [7]. Wei et al. showed that pericardial effu-
sion will develop in 73% of patients if more than 40% of the heart volume receives
more than 30 Gy of radiation [8].
Although acute pericarditis is usually self-limited, the crucial point is the pro-
gression into chronic pericarditis and/or constrictive pericarditis in 10–20% of
patients. However, chronic pericarditis may occur in some patients without any his-
tory of acute pericarditis [9, 10].

© Springer International Publishing AG 2017 117


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_11
118 11 Pericarditis

11.1 Mechanism

After mediastinal radiation therapy, there are some acute and chronic effects on the
heart. Pericarditis is a dominant side effect among acute radiation-induced cardiac
complications, and the essential factors in the acute phase are tumor necrosis factor
(TNF) and interleukins (IL) (i.e., IL-1, IL-6, and IL-8) that cause neutrophil infiltra-
tion within the tissues [11]. Delayed radiation-induced pericarditis is generally the
consequence of an acute inflammatory process followed by fibrin deposition. Injury
is primarily initiated by microvascular damage, which causes episodic ischemia.
The next stage is neovascularization with abnormal and permeable vessels, which
lead to worsening of ischemia and fibrosis progression [12].
Fibrous thickening, pericardial adhesion, and effusion are pathological findings
after radiation to the heart and pericardium. These findings are secondary to micro-
vascular and mesothelial cell injuries. On the other hand, plasminogen activator
decreases, and considering its usual role in fibrinolysis, high levels of fibrin are
expected [13]. Pericardial effusions have high amounts of protein and fibrous adhe-
sions and are also accompanied by high levels of serum inflammatory markers dur-
ing the acute phase [10]. Mononuclear cells infiltrate the pericardium 2 days after
radiation exposure in optic microscopy. Other histologic findings are bizarre fibro-
blasts (with abnormalities in the nucleus and cytoplasm), sclerosis of small vessels,
and fat necrosis of the pericardium [14].

11.2 Timing

Acute pericarditis is uncommon, and it can present during or after radiation. It is


usually detected by subclinical pericardial effusion [11, 15]. The presentation of
pericarditis during the first year after radiation therapy, known as acute pericarditis,
is usually a self-limited and benign disease [16], although it has been named
radiation-­induced late pericardial disease by others [15].

11.3 Risk Factors

Several factors increase the risk of radiation-induced pericarditis, but the per-
centage of irradiated pericardial volume and mean pericardial dose are the most
important [3]. The percentage of cardiac silhouette irradiated could be as an
index of pericardial volume. Other risk factors are the nature of radiation source
and duration and fractionation of radiotherapy. There are some studies that deter-
mine the predictive parameters for radiation-induced pericarditis and pericardial
effusion. Daily dose per fraction more than 3 Gy has been shown to correlate
with pericardial complications [17, 18]. Mean dose to pericardium is a predictive
factor for pericardium injury, and 27 Gy is a proper cutoff point in most studies
[6, 8, 17].
11.5 Diagnosis 119

11.4 Symptoms

As a common manifestation of acute pericarditis, patients present with chest pain,


which usually includes characteristic pleuritic chest pain. This is accompanied by
low-grade fever, dyspnea, and tachycardia [10]. Pericardial friction rub can be found
on physical examination [13].
Patient signs and symptoms could be categorized into six groups depending on
time elapsed from radiation exposure [19]:

1. Acute pericarditis during radiation therapy, which presents with nonspecific


symptoms including chest pain, fever, and EKG abnormalities.
2. Late pericardial disease, which occurs during the first year of radiation therapy
[15].
3. Delayed pericarditis, which presents from months to years after radiation treat-
ment (most references include the second half of late pericarditis timing). In this
group of patients, the most common presentation is dyspnea; some others
develop pericardial pain. Tamponade may also rarely occur in these patients [2].
Pulsus paradoxus (decreasing peripheral arterial pulse paradoxically during
inspiration) is characteristic of tamponade [4].
4. Pancarditis, which manifests as heart failure, is difficult to control. It is a rare
compilation of heart radiation during the acute phase because of the delayed
effect of radiation on the myocardium.
5. Constrictive pericarditis usually develops several years after radiation [15].
Kussmaul’s sign (jugular venous distention on inspiration) is seen in constrictive
pericarditis and less frequently in cardiac tamponade [4].
6. Asymptomatic pericardial effusion, which is an incidental finding by
echocardiography.

11.5 Diagnosis

Considering positional pleuritic chest pain as a hallmark feature of acute pericardi-


tis, an EKG is usually the first diagnostic study obtained. Diffuse ST elevation with
or without PR interval depression could be seen, but sinus tachycardia may also be
seen [15].
Chest radiography in patients with pericardial effusion may show
cardiomegaly.
It is easy to detect loculated or generalized pericardial effusion by echocardiog-
raphy by measurement of echo-free pericardial space. There are other signs on
echocardiography (e.g., collapsing right atrium and ventricle in end-diastolic and
diastolic phase, respectively) [4].
By using computed tomography (CT), pericardial effusion and thickening are
showed and, in comparison to MRI, is more sensitive to detect constrictive pericar-
ditis [4]. Normal thickness of the pericardium is less than 2 mm on CT scan and
120 11 Pericarditis

MRI. Thickened pericardium could be enhanced after IV contrast injection, which


is a sign of inflammation and is used to differentiate between constrictive pericardi-
tis and restrictive cardiomyopathy [20].
Serum inflammation markers, such as white blood cell count, erythrocyte sedi-
mentation rate (ESR), and serum C-reactive protein (CRP), are usually elevated;
serum cardiac troponin I levels may be minimally elevated. CRP as an acute phase
protein can help in treatment monitoring in most patients that have elevated levels
of this marker [21].
In patients with malignant tumors and pericardial effusion, at least three dif-
ferential diagnoses should be considered. One of them is heart failure with peri-
cardial effusion, which is secondary to low cardiac output. Another is malignant
pericardial effusion, which could be accompanied by other metastases and is
confirmed by positive cytological examination for malignant cells. A third dif-
ferential diagnosis is hypothyroidism, which may be due to radiation to the
thyroid [2].

11.6 Scoring

Pericardial effusions and pericarditis have been scored in Common Toxicity Criteria
for Adverse Effect (CTCAE v4.03) as shown in Tables 11.1 and 11.2 [22]. In this
scoring system, no grade 1 has been described for pericardial effusion, and pericar-
dial tamponade is grade 4 at least.
RTOG scoring also classifies acute radiation heart effect into four grades
(Table 11.3) [23].

Table 11.1 CTCAE, version 4.03, scoring system for pericardial effusion
Definition
Grade 1
Grade 2 Asymptomatic effusion size, small to moderate
Grade 3 Effusion with physiologic consequences
Grade 4 Life-threatening consequences with urgent intervention indicated
Grade 5 Death

Table 11.2 CTCAE, version 4.03, scoring system for pericarditis


Definition
Grade 1 Asymptomatic, EKG, or physical findings (e.g., friction rub) consistent with
pericarditis
Grade 2 Symptomatic pericarditis (e.g., chest pain)
Grade 3 Pericarditis with physiologic consequences (e.g., pericardial constriction)
Grade 4 Life-threatening consequences with urgent intervention indicated
Grade 5 Death
11.7 Prevention 121

Table 11.3 RTOG/EORTC radiation toxicity grading for cardiac toxicity


Definition
Grade 1 Asymptomatic but objective evidence of EKG changes or pericardial abnormalities
without evidence of other heart disease
Grade 2 Symptomatic with EKG changes and radiological findings of congestive heart failure
or pericardial disease/no specific treatment required
Grade 3 Congestive heart failure, angina pectoris, and pericardial disease responding to
therapy
Grade 4 Congestive heart failure, angina pectoris, pericardial disease, and arrhythmias not
responsive to nonsurgical measures

11.7 Prevention

Multidisciplinary approach should be considered in the management of patients


with cancer. In some centers, cancer patients are evaluated by cardiologists with
respect to cardio-oncology history taking, physical exam, chest X-ray, and electro-
cardiography with strain and are routine, and based on these patients, risk is deter-
mined [1].
Lowering the pericardium-irradiated volume and dose without any defect in
treatment is an essential step in reducing the probability of pericardial side effect
development. Emami et al. published data on normal tissue tolerance by using TD
5/5 and TD 50/5 parameters. TD 5/5 is the dose associated with a 5% risk of com-
plications within 5 years, and TD 50/5 is the same but in 50% of patients. Considering
pericarditis as the end point, TD 5/5 in conventional radiation therapy is about
60 Gy for one third of the heart, 45 Gy for two third, and 40 Gy for the whole vol-
ume of the heart. For TD 50/5, these doses, respectively, change to 70, 55, and
50 Gy. In patients with predicted long survival, TD 5/5 is determined much lower
(25 Gy), corresponding to other cardiac complications [15].
If we consider two-dimensional planning of radiation therapy, by radiation to
more than 50% of the heart, we will observe an increase in the risk of pericarditis.
In three-dimensional planning, the volume of pericardium that receives ≥30 Gy
(V30) is a main determinant. In order to prevent pericardial injury, it is recom-
mended to keep mean pericardial dose less than 26 Gy or pericardial V30 less than
46% [24] although there are some challenges in defining heart volume [9].
Radiation-induced pericarditis occurs in less than 15% of patients by keeping the
mean dose to the pericardium less than 26 Gy [11].
Using modern techniques in radiation therapy, cardiac exposure has changed. In
breast cancer, irradiation on deep inspiration (unassisted or by using some devices
for active breathing control) and even breast board significantly reduces dose and
volume of irradiated heart. Another factor for decreasing heart dose is using elec-
tron beam irradiation, especially for internal mammary nodes if indicated [13].
Breast radiation therapy in prone position is also helpful to reduce irradiated peri-
cardium volume [5].
By using IMRT, more heart volume is irradiated by lower dose, but high-dose
volume decreases [1].
122 11 Pericarditis

11.8 Treatment

Acute pericarditis is usually self-limited, and half of the patients do not require any
intervention.
Patients are usually treated in the outpatient setting unless they have high-risk
features such as fever (temperature >38°C), leukocytosis, a large pericardial effu-
sion (echo-free space >20 mm), cardiac tamponade, acute trauma, immunosup-
pressed state, concurrent oral anticoagulation, failure of nonsteroidal
anti-inflammatory drug (NSAID) therapy, elevated troponin levels, and recurrent or
incessant pericarditis, all of which mandate hospitalization [21].
Forty percent of symptomatic patients are good responders to the rest and
NSAIDs.
Colchicine is suggested for patients with acute pericarditis as an adjunct to
NSAID therapy and sometimes is sufficient in relieving pain in patients with acute
pericarditis and preventing recurrences [11, 16, 21].
The important practical point is that this presentation is not a reason for radiation
therapy discontinuation, although a reduction in dose should be considered [11, 19].
Different NSAIDs with various dosages are recommended:

Aspirin: 500–1000 mg every 6–8 h (range 1.5–4 g/day).


Ibuprofen: 600 mg every 8 h (range 1200–2400 mg/day).
Indomethacin: 25–50 mg every 8 h; start at the lower end of dosing range and titrate
upward to avoid headaches and dizziness.
Naproxen: 500–1000 mg every 12 h may be used if tolerated and clinically indi-
cated and may increase to 1500 mg daily for a limited time (<6 months).

For geriatric patients, the lowest dose and frequency are recommended.
In patients with renal impairment and a creatinine clearance (CrCl) ≤30 mL/min,
NSAIDs are not recommended. Aspirin is slightly safer than others in these patients,
but it’s not recommended if CrCl is less than 10 mL/min.
NSAIDs should be also used with caution in hepatic impairment. Prophylaxis
with proton pump inhibitors is also an important treatment to implement.
Uncomplicated treatment continues for 1–2 weeks. However, symptom and CRP
normalization should be considered [25].
Pericardiocentesis may be performed for patients with persistent symptomatic
pericardial effusions. The approach to recurrent pericardial effusions would be sur-
gical by either a pericardial window or pericardiectomy [2].

References
1. Herrmann J, Lerman A, Sandhu NP, Villarraga HR, Mulvagh SL, Kohli M (eds) (2014)
Evaluation and management of patients with heart disease and cancer: cardio-oncology. Mayo
Clin Proc 89(9):1287–1306
2. Mann DL, Zipes DP, Libby P, Bonow RO (2014) Braunwald’s heart disease: a textbook of
cardiovascular medicine. Elsevier Health Sciences, Philadelphia
References 123

3. Marx J, Walls R, Hockberger R (2013) Rosen’s emergency medicine-concepts and clinical


practice. Elsevier Health Sciences, Philadelphia
4. Sellke F, Pedro J, Swanson SJ (2015) Sabiston and Spencer Surgery of the chest. Elsevier
Health Sciences, Philadelphia
5. Nitsche M, Pahl R, Huber K, Eilf K, Dunst J (2015) Cardiac toxicity after radiotherapy for
breast cancer: myths and facts. Breast Care (Basel) 10:131–135
6. Carmel RJ, Kaplan HS (1976) Mantle irradiation in Hodgkin’s disease. An analysis of tech-
nique, tumor eradication, and complications. Cancer 37(6):2813–2825
7. Fukada J, Shigematsu N, Takeuchi H, Ohashi T, Saikawa Y, Takaishi H et al (2013)
Symptomatic pericardial effusion after chemoradiation therapy in esophageal cancer patients.
Int J Radiat Oncol Biol Phys 87(3):487–493
8. Wei X, Liu HH, Tucker SL, Wang S, Mohan R, Cox JD et al (2008) Risk factors for pericardial
effusion in inoperable esophageal cancer patients treated with definitive chemoradiation ther-
apy. Int J Radiat Oncol Biol Phys 70(3):707–714
9. Gagliardi G, Constine LS, Moiseenko V, Correa C, Pierce LJ, Allen AM et al (2010) Radiation
dose–volume effects in the heart. Int J Radiat Oncol Biol Phys 76(3):S77–S85
10. Jaworski C, Mariani JA, Wheeler G, Kaye DM (2013) Cardiac complications of thoracic irra-
diation. J Am Coll Cardiol 61(23):2319–2328
11. Madan R, Benson R, Sharma D, Julka P, Rath G (2015) Radiation induced heart disease:
pathogenesis, management and review literature. J Egypt Natl Canc Inst 27(4):187–193
12. Taunk NK, Haffty BG, Kostis JB, Goyal S (2015) Radiation-induced heart disease: pathologic
abnormalities and putative mechanisms. Front Oncol 5:39
13. Senkus-Konefka E, Jassem J (2007) Cardiovascular effects of breast cancer radiotherapy.
Cancer Treat Rev 33(6):578–593
14. Willis MS, Homeister JW, Stone JR (2014) Cellular and molecular pathobiology of cardiovas-
cular disease. Academic Press, London
15. Carver JR (2011) Management of cardiac and pulmonary treatment-related side effects.
J Support Oncol 9:67–94
16. Byhardt RW (2010) Radiation oncology: rational, technique, results. Elsevier Health Sciences,
Philadelphia, pp 411–423
17. Martel MK, Sahijdak WM, Ten Haken RK, Kessler ML, Turrisi AT (1998) Fraction size and
dose parameters related to the incidence of pericardial effusions. Int J Radiat Oncol Biol Phys
40(1):155–161
18. Cosset J, Henry-Amar M, Meerwaldt J (1991) Long-term toxicity of early stages of Hodgkin’s
disease therapy: the EORTC experience. Ann Oncol 2:77–82
19. Yusuf SW, Sami S, Daher IN (2011) Radiation-induced heart disease: a clinical update. Cardiol
Res Pract 2011:317659
20. Benveniste MF, Gomez D, Carter BW, Cuellar SLB, De Groot PM, Marom EM (eds) (2016)
Radiation effects in the mediastinum and surroundings: imaging findings and complications.
Semin Ultrasound CT MR 37(3):268–280
21. Khandaker MH, Espinosa RE, Nishimura RA, Sinak LJ, Hayes SN, Melduni RM et al (eds)
(2010) Pericardial disease: diagnosis and management. Mayo Clin Proc 85(6):572–593
22. CTCAE, Version 4.03, Scoring System.
23. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
24. Halperin EC, Brady LW, Perez CA, Wazer DE (2013) Perez & Brady’s principles and practice
of radiation oncology. Philadephia, 6th eddition, Wolters Kluwer/ Lippincot Williams &
Wilkins, p 1028
25. Task FM, Adler Y, Charron P, Imazio M, Badano L, Barón-Esquivias G et al (2015) 2015 ESC
Guidelines for the diagnosis and management of pericardial diseases: The Task Force for the
Diagnosis and Management of Pericardial Diseases of the European Society of Cardiology
(ESC) Endorsed by: The European Association for Cardio-Thoracic Surgery (EACTS). Eur
Heart J 36(42):2921–2964
Esophagitis
12

Radiation esophagitis is an acute dose-limiting toxicity associated with radiation


treatment that decreases the patient’s compliance for treatment completion.
Incidence of acute esophagitis has been reported in about 29% of patients with
thoracic radiation therapy [1] and in 93% of patients undergoing thoracic chemora-
diation therapy [2]. Severe esophagitis requiring nutritional support or radiation
therapy breaks has been reported in 1–4% and 22–45% of patients that are treated
with radiation therapy alone or chemoradiation therapy, respectively [3, 4].

12.1 Mechanism

The luminal side of the normal esophagus is lined by mucosa. The mucosal layer
contains epithelium, lamina propria, and muscularis mucosae. The esophageal epi-
thelium is a nonkeratinizing, stratified, squamous epithelium and contains rapidly
dividing cells situated in the basal layer [5].
Radiation therapy affects the basal epithelial cell layer and limits the prolifera-
tion rate of the basal epithelium, causing mucosal thinning, ulceration, and initia-
tion of the inflammatory response resulting in congestion, edema, or erosion [6].

12.2 Timing

Acute radiation esophagitis begins on the second to third week after initiation of
irradiation (20–30 Gy). Generally, with single daily fractions of 2.0 Gy radiation
therapy, grade 1 esophagitis first appears in the second week, and grade 2 and higher

© Springer International Publishing AG 2017 125


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_12
126 12 Esophagitis

esophagitis begins during the third week of the treatment. As treatment continues,
the rate of symptomatic esophagitis increases. Patients experience grade 3 esopha-
gitis during the fifth week of treatment [7].
Esophagitis often recovers within 4–6 weeks of treatment completion. The dura-
tion of symptomatic esophagitis is longer for intensive treatment (hyperfractionated
radiation therapy or concurrent chemotherapy) [8].

12.3 Risk Factors

The most significant predictors of acute esophagitis are concurrent chemotherapy


[9–13] and hyperfractionated radiation therapy, especially in the setting of concur-
rent chemotherapy with hyperfractionated radiation therapy [12, 14–17].
It has been reported that the use of chemotherapy concurrent with irradiation is
associated with a nearly 12-fold greater risk of developing severe esophagitis [18].
Sequential chemotherapy seems not to significantly increase the risk of esophageal
radiation injury [3, 4, 14, 19]. The incidence of esophagitis in chemoradiotherapy is
also dependent on chemotherapeutic agents used. Cisplatin-based regimens are gen-
erally associated with a lower rate of significant esophagitis compared with proto-
cols using paclitaxel [7, 20, 21].
Radiation regimen schedule is an important factor in the rate of esophagitis.
Higher radiotherapy dose per fraction [22] and increasing the number of daily frac-
tions induce more severe esophagitis.
A number of studies evaluating dosimetric factors have shown that percentage
of esophagus volume receiving 10–60 Gy [7, 9, 23–30], mean esophageal dose
[24, 25, 31], the maximal esophageal dose [11, 13, 32], esophageal surface area
receiving 55 Gy [9], and higher length of the esophagus in the radiation treatment
field (with some contradictory results) [6, 15, 33] are all associated with acute
esophagitis.
A recent systematic literature review concluded that the valuable dosimetric
parameters predicting the occurrence of acute radiation esophagitis in patients
receiving concurrent chemoradiotherapy include maximum esophageal dose,
mean esophageal dose, and esophageal volume receiving 20, 30, 50, and 55 Gy
[34].
It is well known that with the similar dosimetric parameter consideration, only a
small proportion of patients develop esophageal toxicity. Individual variability in
radiosensitivity should be further investigated to identify genetic variations modu-
lating the development of radiation esophagitis. In this regard, it has been observed
that patients with the transforming growth factor-beta1 (TGFβ1)509 CC genotype
[35], heat shock protein beta-1 (HSPB1) CC genotype [36], and single nucleotide
polymorphisms (SNPs) in inflammation-related genes including three PTGS2
12.5 Scoring 127

(COX2) variants rs20417, rs5275, and rs689470 [37] are at greater risk for develop-
ing radiation esophagitis.
It has been shown that one of the most significant risk factors for dysphagia dur-
ing chemoradiation is the maximal grade of neutropenia. Indeed, the nadir of the
neutrophil granulocytes during treatment is strongly associated with developing and
severity of esophagitis [17, 38]. Patients with higher pretreatment platelet counts
and lower hemoglobin levels as a preexisting systemic inflammatory state have
greater rates of radiation esophagitis [39].
There are individual case reports demonstrated that patients with human immu-
nodeficiency virus (HIV) may experience unusually severe radiation-induced
esophagitis [40, 41].
Other factors proposed to be associated with acute esophagitis include the
­pretreatment body mass index [30].
Patient’s gender, age, performance status, and histology are not significantly
­correlated with esophageal toxicity [6, 42].

12.4 Symptoms

Radiation-induced esophagitis presents with retrosternal or substernal burning sen-


sation, pain, odynophagia, dysphagia, and anorexia. Weight loss could occur and
nutritional support may be needed. Rarely, with severe esophagitis, patients may
develop obstruction, perforation, or fistulas.

12.5 Scoring

The Radiation Therapy Oncology Group (RTOG) and European Organization for
Research and Treatment of Cancer (EORTC) have defined four grades for radiation-­
induced esophagitis (Table 12.1) [43].

Table 12.1 RTOG/EORTC esophagitis grading


Definition
Grade 1 Mild dysphagia or odynophagia (may require topical anesthetic or nonnarcotic
analgesics and a soft diet)
Grade 2 Moderate dysphagia or odynophagia (may require narcotic analgesics and puree or
liquid diet)
Grade 3 Severe dysphagia or odynophagia with dehydration or weight loss >15% from
pretreatment baseline requiring NG feeding tube, IV fluids, or hyperalimentation
Grade 4 Complete obstruction, ulceration, perforation, and fistulization
128 12 Esophagitis

12.6 Diagnosis

Radiation esophagitis is a clinical diagnosis in patients with dysphagia or odyno-


phagia, that is, receiving thoracic irradiation. In patients with symptoms progress-
ing after completion of radiation therapy despite the optimal treatment, endoscopy
should be considered to establish the diagnosis and rule out other etiologies like
infectious or fungal esophagitis.

12.7 Prevention

Prediction of esophagitis severity allows prevention of esophagitis with treatment


deintensification and other measurements. Various dose constraints are proposed to
predict radiation-induced esophageal toxicity that should be considered in treatment
planning.
It has been shown that esophagitis as an inflammatory process results in elevated
18
F-fluordeoxyglucose (FDG) positron emission tomography (PET) uptake in the
postradiation period, which correlates to the treatment planning dose distribution.
Using posttreatment 18F-FDG-PET scans allows for developing a quantitative bio-
logical model to improve the accuracy of esophagitis prediction based on planned
dose [44]. Also an increase in FDG uptake during radiation therapy from pre-radia-
tion may predict radiation esophagitis and provides the opportunity for treatment
planning modification [45].
Advances in the radiation delivery with intensity-modulated radiation therapy
(IMRT) may allow radiation oncologists to prescribe higher doses to tumors with
normal structures being spared such and keep the incidence of radiation-induced
esophagitis quite low when the esophagus is not the treatment target [11, 46].
Amifostine is an organic thiophosphate compound that offers selectively normal
cell protection from radiation-induced damage and also chemotherapeutic agents by
scavenging oxygen-derived free radicals [47]. It seems that amifostine administra-
tion can protect the normal esophageal mucosa from radiation-induced injury and
delay the onset of esophagitis and reduce its severity and incidence.
In a multicenter trial of 146 patients with advanced lung cancer treated with a
daily fractionated radiation therapy to a total of 55–60 Gy with or without daily
amifostine administration reported that the incidence of esophagitis grade 2 or
higher was 42% in the radiation therapy alone group versus 4% in the group with
amifostine administration during 4 weeks of treatment [48].
Amifostine administration seems to reduce severe esophagitis and analgesic
intake in patients undergoing chemoradiation therapy without compromising treat-
ment outcomes [20, 49–51]. Further studies are required to determine the optimal
amifostine combination with therapeutic strategy.
Glutamine supplement administration during thoracic irradiation appears to
postpone onset and reduce the severity of radiation esophagitis [52]. Further inves-
tigation of this agent may therefore be warranted.
References 129

Oral sucralfate solution has been compared with placebo as prophylaxis in a


prospective trial and didn’t prevent esophagitis development, although it had signifi-
cant gastrointestinal side effects [53].

12.8 Management

Acute esophagitis is managed symptomatically.


Dietary modification that should be recommended to all patients are:

Eat frequent meals with small pieces throughout the day instead of three large
meals.
Eat soft foods that are warm or at room temperature.
Drink enough liquids.
Avoid hot or spicy foods, acidic foods, and hard and crunchy foods.
Avoid alcohol and tobacco.

Pain is managed with topical analgesics (e.g., viscous lidocaine), NSAIDs, or


narcotics.
If patients complain of reflux symptoms, antacids, proton-pump inhibitors, or H2
receptor blockers could be effective.
In the setting of severe dysphagia or odynophagia with significant weight loss,
short breaks from radiation therapy have been proposed. Feeding tube or parenteral
nutrition may be needed for these patients (see Sect. 6.7.5).

References
1. Le Chevalier T, Arriagada R, Quoix E, Ruffle P, Martin M, Tarayre M et al (1991) Radiotherapy
alone versus combined chemotherapy and radiotherapy in nonresectable non-small-cell lung
cancer: first analysis of a randomized trial in 353 patients. J Natl Cancer Inst 83(6):417–423
2. Wei X, Liu HH, Tucker SL, Liao Z, Hu C, Mohan R, Cox JD, Komaki R (2006) Risk factors for
acute esophagitis in non-small-cell lung cancer patients treated with concurrent chemotherapy
and three-dimensional conformal radiotherapy. Int J Radiat Oncol Biol Phys 66(1):100–107
3. Dillman RO, Seagren SL, Propert KJ, Guerra J, Eaton WL, Perry MC et al (1990) A random-
ized trial of induction chemotherapy plus high-dose radiation versus radiation alone in stage
III non-small-cell lung cancer. N Engl J Med 323(14):940–945
4. Curran WJ, Paulus R, Langer CJ, Komaki R, Lee JS, Hauser S et al (2011) Sequential vs con-
current chemoradiation for stage III non-small cell lung cancer: randomized Phase III trial
RTOG 9410. J Natl Cancer Inst 103(19):1452–1460
5. Squier CA, Kremer MJ (2000) Biology of oral mucosa and esophagus. J Natl Cancer Inst
Monogr 29:7–15
6. Choy H, LaPorte K, Knill-Selby E, Mohr P, Shyr Y (eds) (1999) Esophagitis in combined modality
therapy for locally advanced non-small cell lung cancer. Semin Radiat Oncol 9(2 Suppl 1):90–96
7. Ozgen A, Hayran M, Kahraman F (2012) Mean esophageal radiation dose is predictive of the
grade of acute esophagitis in lung cancer patients treated with concurrent radiotherapy and
chemotherapy. J Radiat Res 53(6):916–922
130 12 Esophagitis

8. Ball D, Bishop J, Smith J, Crennan E, O’Brien P, Davis S et al (1995) A phase III study of
accelerated radiotherapy with and without carboplatin in nonsmall cell lung cancer: an interim
toxicity analysis of the first 100 patients. Int J Radiat Oncol Biol Phys 31(2):267–272
9. Bradley J, Deasy JO, Bentzen S, El Naqa I (2004) Dosimetric correlates for acute esophagitis
in patients treated with radiotherapy for lung carcinoma. Int J Radiat Oncol Biol Phys
58(4):1106–1113
10. Choy H, Akerley W, Safran H, Graziano S, Chung C, Williams T et al (1998) Multiinstitutional
phase II trial of paclitaxel, carboplatin, and concurrent radiation therapy for locally advanced
non-small-cell lung cancer. J Clin Oncol 16(10):3316–3322
11. Qiao W-B, Zhao Y-H, Zhao Y-B, Wang R-Z (2005) Clinical and dosimetric factors of radiation-­
induced esophageal injury: radiation-induced esophageal toxicity. World J Gastroenterol
11(17):2626–2629
12. Maguire PD, Sibley GS, Zhou S-M, Jamieson TA, Light KL, Antoine PA et al (1999) Clinical
and dosimetric predictors of radiation-induced esophageal toxicity. Int J Radiat Oncol Biol
Phys 45(1):97–103
13. Singh AK, Lockett MA, Bradley JD (2003) Predictors of radiation-induced esophageal toxic-
ity in patients with non-small-cell lung cancer treated with three-dimensional conformal radio-
therapy. Int J Radiat Oncol Biol Phys 55(2):337–341
14. Byhardt R, Scott C, Sause W, Emami B, Komaki R, Fisher B et al (1998) Response, toxicity, failure
patterns, and survival in five radiation therapy oncology group (RTOG) trials of sequential and/or
concurrent chemotherapy and radiotherapy for locally advanced non-small-cell carcinoma of the
lung. Int J Radiat Oncol Biol Phys 42(3):469–478
15. Werner-Wasik M, Pequignot E, Leeper D, Hauck W, Curran W (2000) Predictors of severe
esophagitis include use of concurrent chemotherapy, but not the length of irradiated esopha-
gus: a multivariate analysis of patients with lung cancer treated with nonoperative therapy. Int
J Radiat Oncol Biol Phys 48(3):689–696
16. Werner-Wasik M, Paulus R, Curran WJ, Byhardt R (2011) Acute esophagitis and late lung
toxicity in concurrent chemoradiotherapy trials in patients with locally advanced non-small-­
cell lung cancer: analysis of the radiation therapy oncology group (RTOG) database. Clin
Lung Cancer 12(4):245–251
17. Turrisi AT, Kim K, Blum R, Sause WT, Livingston RB, Komaki R et al (1999) Twice-daily
compared with once-daily thoracic radiotherapy in limited small-cell lung cancer treated con-
currently with cisplatin and etoposide. N Engl J Med 340(4):265–271
18. Werner-Wasik M, Scott C, Graham ML, Smith C, Byhardt RW, Roach M et al (1999)
Interfraction interval does not affect survival of patients with non-small cell lung cancer treated
with chemotherapy and/or hyperfractionated radiotherapy: a multivariate analysis of 1076
RTOG patients. Int J Radiat Oncol Biol Phys 44(2):327–331
19. Belderbos J, Heemsbergen W, Hoogeman M, Pengel K, Rossi M, Lebesque J (2005) Acute
esophageal toxicity in non-small cell lung cancer patients after high dose conformal radio-
therapy. Radiother Oncol 75(2):157–164
20. Leong SS, Tan EH, Fong KW, Wilder-Smith E, Ong YK, Tai BC et al (2003) Randomized
double-blind trial of combined modality treatment with or without amifostine in unresectable
stage III non-small-cell lung cancer. J Clin Oncol 21(9):1767–1774
21. Maraz A, Furak J, Varga Z, Fodor E, Egyued Z, Borzási E et al (2013) Acute oesophageal
toxicity related to paclitaxel-based concurrent chemoradiotherapy for non-small cell lung can-
cer. Anticancer Res 33(4):1737–1741
22. Bar-Ad V, Leiby B, Witek M, Xiao Y, Cui Y, Dai Y et al (2014) Treatment-related acute esoph-
agitis for patients with locoregionally advanced non-small cell lung cancer treated with
involved-field radiotherapy and concurrent chemotherapy. Am J Clin Oncol 37(5):433–437
23. Takeda K, Nemoto K, Saito H, Ogawa Y, Takai Y, Yamada S (2006) Predictive factors for acute
esophageal toxicity in thoracic radiotherapy. Tohoku J Exp Med 208(4):299–306
24. Watkins JM, Wahlquist AE, Shirai K, Garrett-Mayer E, Aguero EG, Fortney JA et al (2009) Factors
associated with severe acute esophagitis from hyperfractionated radiotherapy with concurrent che-
motherapy for limited-stage small-cell lung cancer. Int J Radiat Oncol Biol Phys 74(4):1108–1113
References 131

25. Zehentmayr F, Söhn M, Exeli A-K, Wurstbauer K, Tröller A, Deutschmann H et al (2015) Normal
tissue complication models for clinically relevant acute esophagitis (≥ grade 2) in patients treated
with dose differentiated accelerated radiotherapy (DART-bid). Radiat Oncol 10(1):121
26. Rodríguez N, Algara M, Foro P, Lacruz M, Reig A, Membrive I et al (2009) Predictors of acute
esophagitis in lung cancer patients treated with concurrent three-dimensional conformal radio-
therapy and chemotherapy. Int J Radiat Oncol Biol Phys 73(3):810–817
27. Zhang Z, Xu J, Zhou T, Yi Y, Li H, Sun H et al (2014) Risk factors of radiation-induced acute
esophagitis in non-small cell lung cancer patients treated with concomitant chemoradiother-
apy. Radiat Oncol 9(1):54
28. Kuroda Y, Sekine I, Sumi M, Sekii S, Takahashi K, Inaba K et al (2013) Acute radiation esoph-
agitis caused by high-dose involved field radiotherapy with concurrent cisplatin and vinorel-
bine for stage III non-small cell lung cancer. Technol Cancer Res Treat 12(4):333–339
29. Palma DA, Senan S, Oberije C, Belderbos J, de Dios NR, Bradley JD et al (2013) Predicting
esophagitis after chemoradiation therapy for non-small cell lung cancer: an individual patient
data meta-analysis. Int J Radiat Oncol Biol Phys 87(4):690–696
30. Patel AB, Edelman MJ, Kwok Y, Krasna MJ, Suntharalingam M (2004) Predictors of acute
esophagitis in patients with non-small cell lung carcinoma treated with concurrent chemo-
therapy and hyperfractionated radiotherapy followed by surgery. Int J Radiat Oncol Biol Phys
60(4):1106–1112
31. Huang EX, Bradley JD, El Naqa I, Hope AJ, Lindsay PE, Bosch WR et al (2012) Modeling the
risk of radiation-induced acute esophagitis for combined Washington University and RTOG
trial 93-11 lung cancer patients. Int J Radiat Oncol Biol Phys 82(5):1674–1679
32. Gong B, Jiang N, Yan G, Wang S, Deng C, Wei S et al (2016) Predictors for severe acute
esophagitis in lung cancer patients treated with chemoradiotherapy: a systematic review. Curr
Med Res Opin 32(10):1701–1708
33. Langer CJ, Movsas B, Hudes R, Schol J, Keenan E, Kilpatrick D et al (eds) (1997) Induction
paclitaxel and carboplatin followed by concurrent chemoradiotherapy in patients with unre-
sectable, locally advanced non-small cell lung carcinoma: report of Fox Chase Cancer Center
study 94-001. Semin Oncol 24(4 Suppl 12):S12-89–S12-95
34. Yu Y, Guan H, Dong Y, Xing L, Li X (2016) Advances in dosimetry and biological predictors
of radiation-induced esophagitis. Onco Targets Ther 9:597–603
35. Ellingrod VL, Schipper M, Stringer KA, Cai X, Hayman JA, Yu J et al (2013) Genetic varia-
tions in TGFβ1, tPA, and ACE and radiation-induced thoracic toxicities in patients with non-­
small-­cell lung cancer. J Thorac Oncol 8(2):208–213
36. Guerra JLL, Wei Q, Yuan X, Gomez D, Liu Z, Zhuang Y et al (2011) Functional promoter
rs2868371 variant of HSPB1 associates with radiation-induced esophageal toxicity in patients
with non-small-cell lung cancer treated with radio (chemo) therapy. Radiother Oncol
101(2):271–277
37. Hildebrandt MA, Komaki R, Liao Z, Gu J, Chang JY, Ye Y et al (2010) Genetic variants in
inflammation-related genes are associated with radiation-induced toxicity following treatment
for non-small cell lung cancer. PLoS One 5(8):e12402
38. Everitt S, Duffy M, Bressel M, McInnes B, Russell C, Sevitt T et al (2016) Association of
oesophageal radiation dose volume metrics, neutropenia and acute radiation oesophagitis in
patients receiving chemoradiotherapy for non-small cell lung cancer. Radiat Oncol 11(1):20
39. Tang C, Liao Z, Zhuang Y, Levy LB, Hung C, Li X et al (2014) Acute phase response
before treatment predicts radiation esophagitis in non-small cell lung cancer. Radiother
Oncol 110(3):493–498
40. Leigh BR, Lau DH (1998) Severe esophageal toxicity after thoracic radiation therapy for lung
cancer associated with the human immunodeficiency virus: a case report and review of the
literature. Am J Clin Oncol 21(5):479–481
41. Costleigh BJ, Miyamoto CT, Micaily B, Brady LW (1995) Heightened sensitivity of the esoph-
agus to radiation in a patient with AIDS. Am J Gastroenterol 90(5):812–814
132 12 Esophagitis

42. Chapet O, Kong F-M, Lee JS, Hayman JA, Ten Haken RK (2005) Normal tissue complication
probability modeling for acute esophagitis in patients treated with conformal radiation therapy
for non-small cell lung cancer. Radiother Oncol 77(2):176–181
43. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
44. Nijkamp J, Rossi M, Lebesque J, Belderbos J, van den Heuvel M, Kwint M et al (2013) Relating
acute esophagitis to radiotherapy dose using FDG-PET in concurrent chemo-­radiotherapy for
locally advanced non-small cell lung cancer. Radiother Oncol 106(1):118–123
45. Yuan ST, Brown RK, Zhao L, Ten Haken RK, Gross M, Cease KB et al (2014) Timing and
intensity of changes in FDG uptake with symptomatic esophagitis during radiotherapy or
chemo-radiotherapy. Radiat Oncol 9(1):37
46. Kwint M, Uyterlinde W, Nijkamp J, Chen C, de Bois J, Sonke J-J et al (2012) Acute esophagus
toxicity in lung cancer patients after intensity modulated radiation therapy and concurrent
chemotherapy. Int J Radiat Oncol Biol Phys 84(2):e223–e228
47. Kouvaris JR, Kouloulias VE, Vlahos LJ (2007) Amifostine: the first selective-target and broad-­
spectrum radioprotector. Oncologist 12(6):738–747
48. Antonadou D, Coliarakis N, Synodinou M, Athanassiou H, Kouveli A, Verigos C et al (2001)
Randomized phase III trial of radiation treatment±amifostine in patients with advanced-stage
lung cancer. Int J Radiat Oncol Biol Phys 51(4):915–922
49. Komaki R, Lee J, Kaplan B, Allen P, Kelly J, Liao Z et al (eds) (2002) Randomized phase III
study of chemoradiation with or without amifostine for patients with favorable performance
status inoperable stage II-III non-small cell lung cancer: preliminary results. Semin Radiat
Oncol 12:46–49
50. Lawrence YR, Paulus R, Langer C, Werner-Wasik M, Buyyounouski MK, Komaki R et al
(2013) The addition of amifostine to carboplatin and paclitaxel based chemoradiation in locally
advanced non-small cell lung cancer: long-term follow-up of Radiation Therapy Oncology
Group (RTOG) randomized trial 9801. Lung Cancer 80(3):298–305
51. Werner-Wasik M, Axelrod RS, Friedland DP, Hauck W, Rose LJ, Chapman AE et al (eds)
(2002) Phase II: Trial of twice weekly amifostine in patients with non-small cell lung cancer
treated with chemoradiotherapy. Semin Radiat Oncol 12:34–39
52. Topkan E, Yavuz MN, Onal C, Yavuz AA (2009) Prevention of acute radiation-induced esoph-
agitis with glutamine in non-small cell lung cancer patients treated with radiotherapy: evalua-
tion of clinical and dosimetric parameters. Lung Cancer 63(3):393–399
53. McGinnis WL, Loprinzi CL, Buskirk SJ, Sloan JA, Drummond RG, Frank AR et al (1997)
Placebo-controlled trial of sucralfate for inhibiting radiation-induced esophagitis. J Clin Oncol
15(3):1239–1243
Radiation Gastritis
13

Radiation-induced gastritis occurs in about 30–50% of patients undergoing radia-


tion therapy whenever the stomach is included in radiation fields as the main target
or an organ at risk (e.g., during radiation treatment of lymphoma, neoplasm of lower
esophagus, stomach, biliary tract, pancreas, testis, or including of thoracic stomach
in radiation field after gastric pull-up procedure) [1–4].

13.1 Mechanism

The gastric walls consist of mucosa, submucosa, muscularis, and serosal layers. The
gastric mucosa is the mucous membrane layer of the stomach, which contains the
glands and gastric pits. The epithelium of the mucosa has gastric pits (mucous mem-
brane invaginations), and lamina propria contains gastric glands. There are three
types of gastric glands including cardiac glands, which produce lysozyme and
mucus, fundic and body glands, which produce hydrochloric acid, pepsin, and intrin-
sic factor, and lipase and pyloric glands, which produce lysozyme and mucin [3].
Each fundic gland is divided into three segments: the isthmus, neck, and the
gland proper. The main epithelial cell types of the gland are the precursor mucous
neck cells, the parietal or oxyntic cell (secrete hydrochloric acid), and the chief or
zymogen cell (produce pepsinogen). The surface of the mucous membrane is cov-
ered by a single layer of mucous cells. These cells secrete mucous and develop the
gastric mucosal barrier [5].
Several types of endocrine cells are found throughout the gastric mucosa and
produce histamine, gastrin, and somatostatin.
Irradiation produces marked mucosal and submucosal edema, ulcerative or ero-
sive lesions, and changes in the quality and quantity of gastric secretion [2, 6].
The early changes in gastric mucosa include mucosal and submucosal edema
and inflammation, endothelial cell swelling, and capillary dilation, which could be
demonstrated after 20–25 Gy of radiation dose [3].

© Springer International Publishing AG 2017 133


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_13
134 13 Radiation Gastritis

With higher doses, more severe injuries could occur including gastric erosion or
ulcer. The gastric ulcer developing rate increases with doses above 45 Gy (25–30%
of patients) [6]. The degree of gastric acidity in radiation ulcers is variable. In some
cases, the acidity is somewhat increased; in the majority, it is normal or even sub-
normal [1].
Radiation initially reduces surface mucous cells due to their relatively short life-­
span. So breakdown of the mucosal barrier, net flux of electrolytes into the gastric
lumen, and back diffusion of the luminal hydrogen ion occur. Mucosal histamine is
released and stimulates secretion directly from the glandular cells, and a transient
increase in acid secretion during the radiation therapy exhibits [5, 7].
After the initial gastric hypersecretion, secondary mucosal damage occurs and
induces coagulation necrosis of glandular cells, denudation of glandular epithelium,
cystic dilatation of the glands and a reduction of the gastric glands, and the volume
of gastric secretion and its content change [7]. A significant reduction in the secre-
tion of hydrochloric acid and pepsin develops. The degree and duration of the radia-
tion effect on gastric secretion depends on radiation dose and biologic differences
[8]. It has been reported that gastric acidity may suppress or significantly decrease
with doses of less than 20 Gy [9]. Some have suggested that chief cells are more
radiosensitive than are parietal cells, and gastric acidity suppression occurs first due
to damage to the pepsinogen-secreting chief cells; parietal cells are more resistant
and are depressed later [10]. Another reason for gastric acidity suppression is reduc-
tion in gastric histamine content [11]. A decrease in gastric acidity occurs within
4–6 weeks after completion of radiation therapy [12].

13.2 Risk Factors

The severity and frequency of the acute gastric lesions depends upon the total dose
received by the stomach. The radiosensitivity of stomach seems to be higher than
the small and large intestine [13]. The gastric ulcer never occurs following less than
45 Gy. When the radiation dose exceeds 45 Gy, the incidence of gastric ulcer is
25–30%. With higher radiation dose, the more serious the gastric damage may
occur, leading to penetration hemorrhage and perforation [6]. The risk of perforated
ulcers increases with doses greater than 60 Gy [14].
Chemotherapy may also increase the gastric injuries induced by radiation.
With the exception of unknown factors of individual sensitivity, there is no report
of patient factor effects on frequency of the acute gastric lesions [9, 11, 14].

13.3 Timing

Gastritis will develop in the second and third week of irradiation (20–30 Gy) [15].
Patients may experience nausea, epigastric distress, and uncommon vomiting, and
these symptoms last for only a few hours after each fraction of radiation. Later with
higher radiation doses, more severe symptoms including epigastric pain may occur.
13.7 Management 135

Symptoms usually resolve 1–2 weeks after completion of the radiation therapy.
Occasionally the mild epigastric distress or dyspepsia persists for months or even
years [3].
Ulceration occurs from 1 month to 6 years with an average of 5 months after
radiation therapy [6].

13.4 Symptoms

Epigastric distress, anorexia, nausea, vomiting, and pain are more common symp-
toms of radiation gastritis [10, 16].
Acute ulceration may be seen shortly after radiation therapy. The usual symp-
toms of ulcer are present, but food and antacids usually afford no relief [6]. The
most severe symptoms appear when ulcers are accompanied by perforation or
obstruction, usually 1–2 months after irradiation [14].

13.5 Diagnosis

The diagnosis of radiation-induced gastritis is based on clinical suspicions in


patients that received radiation therapy with stomach at least partially within the
treatment portal. Barium meal examination and gastroscopy may be needed in some
patients with severe symptoms or no response to initial treatment.
Radiation therapy in patients that have a tumor in the stomach like lymphoma may
lead to destruction of deeply infiltrating tumor and result in gastric perforation. These
conditions should be considered in distinction of radiation-induced injuries [17].

13.6 Prevention

There is no prophylactic agent available to mitigate the acute gastric injury. Reducing
the irradiated volume specially for higher doses (when the stomach is not the treat-
ment target) is the best way to minimize gastritis probability, and new techniques
could help radiation oncologists in this way, although there are no definitive data
about dose-volume constraints for partial volume irradiation of stomach. Doses of
45 Gy to the whole stomach are associated with ulceration in 5–7% of patients [18].

13.7 Management

Medical management with antacids (H2 blockers and proton pump inhibitors) is
usually used in patients with gastritis symptoms.
Nausea and vomiting are treated with antiemetics. The management of radiation-­
induced nausea and vomiting is discussed separately (see Chap. 20).
136 13 Radiation Gastritis

Radiation-induced gastric ulceration less often responds to the medical manage-


ment than the usual peptic ulcer disease, and due to higher frequency of complica-
tions in these ulcers, early surgical intervention is indicated in patients who do not
respond to medical management [17].

References
1. Hamilton FE (1948) Gastric ulcer following radiation. Trans West Surg Assoc 54(54 Annual
Meet. (1946 Memphis)):51–56
2. Ohtsubo K, Watanabe H, Mouri H, Yamashita K, Yasumoto K, Yano S (2012) Endoscopic
findings of upper gastrointestinal lesions in patients with pancreatic cancer. JOP
13(4):420–426
3. Fajardo L-G, Berthrong M, Anderson R (2001) Radiation pathology. Oxford University Press,
New York
4. Matsumoto S, Kiyosue H, Komatsu E, Wakisaka M, Tomonari K, Hori Y, Matsumoto A, Mori
H (2004) Radiotherapy combined with transarterial infusion chemotherapy and concurrent
infusion of a vasoconstrictor agent for nonresectable advanced hepatic hilar duct carcinoma.
Cancer 100(11):2422–2429
5. Michalowski A, Hornsey S (1986) Assays of damage to the alimentary canal. Br J Cancer
Suppl 7:1–6
6. Sell A, Jensen TS (1966) Acute gastric ulcers induced by radiation. Acta Radiol Ther Phys
Biol 4(4):289–297
7. Ragins H, Fried M, Dombro R, Dittbrenner M, Liu SM, Johnson L, Sanfilippo L (1967) A
comparison of the radiosensitivity of the gastric mucosa at rest and during stimulation.
Secretory, histochemical, and histologic studies. Am J Dig Dis 12(12):1256–1271
8. Snell AM, Ballman JL (1934) Gastric secretion following irradiation of the exposed stomach
and the upper abdominal viscera by roentgen rays. Am J Dig Dis 1(2):164–168
9. Feiring W, Jampol ML (1950) Perforation of a gastric ulcer following intensive radiation ther-
apy. N Engl J Med 242(19):751–753
10. Novak JM, Collins JT, Donowitz M, Farman J, Sheahan DG, Spiro HM (1979) Effects of
radiation on the human gastrointestinal tract. J Clin Gastroenterol 1(1):9–39
11. Man WK, Gompertz RH, Li SK, Michalowski A, Baron JH, Spencer J (1988) Effect of gastric
irradiation on gastric secretion and histamine in mice. Agents Actions 23(3–4):297–299
12. Rider J, Moeller H, Althausen T, Sheline G (1957) The effect of x-ray therapy on gastric acid-
ity and on 17-hydroxycorticoid and uropepsin excretion. Ann Intern Med 47(4):651–665
13. Elliott AR, Jenkinson EL (1934) Ulcerations of the stomach and small intestine following
roentgen therapy. Radiology. doi:10.1148/23.2.149
14. Goldstein HM, Rogers LF, Fletcher GH, Dodd GD (1975) Radiological manifestations of
radiation-induced injury to the normal upper gastrointestinal tract. Radiology
117(1):135–140
15. Breiter N, Sassy T, Trott KR (1993) The effect of dose fractionation on radiation injury in the
rat stomach. Radiother Oncol 27(3):223–228
16. Nam T, Ahn J, Choi Y, Jeong J, Kim Y, Yoon M, Song J, Ahn S, Chung W (2014) The role of
radiotherapy in the treatment of gastric mucosa-associated lymphoid tissue lymphoma. Cancer
Res Treat 46(1):33–40
17. Keltum JM, Jaffe BM, Calhoun TR, Ballinger WF (1977) Gastric complications after radio-
therapy for Hodgkin’s disease and other lymphomas. Am J Surg 134(3):314–317
18. Kavanagh BD, Pan CC, Dawson LA, Das SK, Li XA, Ten Haken RK, Miften M (2010)
Radiation dose-volume effects in the stomach and small bowel. Int J Radiat Oncol Biol Phys
76(3 Suppl):S101–S107
Radiation-Induced Liver Disease
14

Radiation-induced liver disease (RILD), historically called radiation hepatitis, has


been reported in 6–66% of patients whose livers are irradiated based on radiation
dose, exposed liver volume, and baseline liver function [1, 2]. Abnormalities in
laboratory liver function tests may be found on routine evaluation of patients when
RILD is mild, but in severe cases, liver dysfunction may affect the treatment course
and eventually threaten patient’s lives.

14.1 Mechanism

The histologic hallmark associated with RILD is veno-occlusive disease (VOD). It


is characterized by severe congestion of the sinusoids in the central portion of the
lobules with sparing of the larger veins [3].
The pathogenesis of RILD is unknown. Some possible mechanisms have been
proposed. It has been postulated that radiation damages the sinusoidal and vascular
endothelial cells, which initiates the coagulation cascade leading to fibrin accumu-
lation and clot formation. The fibrin network is replaced by collagen deposition and
resulted in progressive fibrous obliteration of small hepatic veins [3, 4]. Irradiation
induces the dysregulated activation of myofibroblastic hepatic stellate cells, increas-
ing some cytokine expression like TGF-β1 (transforming growth factor) and hedge-
hog (Hh) pathway activation (an essential pathway for tissue remodeling), [5] which
are important in the development of fibrosis following exposure to radiation. GDC-­
0449 targets the Hh signaling pathway, and this suppressed Hh signaling may lead
to reduced proliferation of progenitor cells and fibrosis in irradiated livers [5].
Finally, hepatic vessels occlude, resulting in vascular leakage, distorted dilated
sinusoids, and congestion with erythrocyte trapping, causing central zone hypoxia
and hepatocyte death [6].

© Springer International Publishing AG 2017 137


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_14
138 14 Radiation-Induced Liver Disease

A cluster of NF-κB-regulated cytokines, including TNF-α, are induced by radia-


tion and increase the sensitivity of hepatocytes to radiation and induce cell apopto-
sis [7].
After 4 months, an effective circulatory system within the liver has apparently
developed leading to the disappearance of congestion and restoration of hepatic
cells [8].

14.2 Risk Factors

Radiation treatment factors have a strong correlation with RILD. Mean radiation
dose to the liver and volume of liver exposed to more than 30–35 Gy of radiation are
directly related to RILD occurrence [9, 10]. Emami et al. found a <5% rate of RILD
when the mean whole liver dose is ≤30 Gy in patients without preexisting liver
disease or primary liver cancer. The mean liver dose should be ≤28 Gy in those
patients with preexisting liver disease [11]. However, part of the liver could be
safely treated with higher doses with acceptable complications. Dose per fraction is
an important factor in the development of RILD. Altered fractionated radiation ther-
apy with large fraction sizes decreases liver tolerance [12].
The combination of chemotherapy agents with whole liver radiation seems to
decrease liver tolerance [13, 14], although studies with use of fluoropyrimidines
[15–18] or partial liver radiation have not been reported to significantly increase
hepatic complications of combination therapy.
Baseline liver function is an important factor in predicting the occurrence of
RILD. Cirrhotic livers have a lower tolerance for radiation. It has been reported that
patients with worse Child-Pugh Class (B vs. A) and chronic hepatitis B carriers are
at higher risk for developing RILD [9].
Patients with elevated liver enzymes during radiation therapy are at risk for
developing RILD [12]. Such liver abnormalities are presumably related to self-­
limited liver inflammation [19].
Primary hepatobiliary carcinoma is associated with a significantly increased risk
of RILD compared with a diagnosis of liver metastases; this could be related to
preexisting cirrhosis or hepatitis in patients with hepatobiliary carcinoma [20].
Other factors associated with elevated risk of RILD include prior transcatheter
arterial chemoembolization (TACE) [12], portal vein tumor thrombosis [21], tumor
stage [12], and male gender [22].

14.3 Timing

RILD typically occurs 4–8 weeks after the completion of treatment, although it has
been described as early as 2 weeks and as late as 7 months afterward [3].
14.5 Scoring 139

14.4 Symptoms

In severe cases, patients develop rapid weight gain, increase in abdominal girth,
liver enlargement, right upper quadrant discomfort, ascites, and jaundice [3].
Physical examination reveals ascites and hepatomegaly in moderate to severe cases,
although in mild cases these signs are detectable only by ultrasound or abdominal
CT scan [3]. Serum alkaline phosphatase is predominantly elevated (more than
twice the upper limit of normal or baseline value), with minimal increase or rela-
tively normal levels of aspartate transaminase (AST) and alanine (ALT) (in the
range of twofold above normal) and bilirubin [3, 19].
In patients with intrahepatic cancer, decreases in alkaline phosphatase because of
tumor response may mask alkaline phosphatase elevation resulting from RILD [23].
In patients with hepatitis and cirrhosis, nonclassic-type RILD with markedly
elevated serum transaminases (>5 times the upper limit of normal) rather than ele-
vated alkaline phosphatase or a decline in liver function (measured by a worsening
of Child-Pugh score by 2 or more) and jaundice can occur, indicating severe
radiation-­induced injury to hepatocytes [19].
Reactivation of viral hepatitis has been reported in patient who underwent radia-
tion therapy; however, its role in RILD pathogenesis is unclear. Elevation of trans-
aminases rather than commonly reported increase in alkaline phosphatase is seen in
hepatitis B carrier patients and implies radiation injury of hepatocytes rather than
bile ducts [9]. It has been suggested that the risk of hepatitis B reactivation is
decreased with prophylactic antiretroviral therapy [19].
Thrombocytopenia has been reported in children studies related to congestion of
the portal bed and spleen and secondary hypersplenism [24].

14.5 Scoring

Most research uses the Cancer Therapy Evaluation Program and Common
Terminology Criteria for Adverse Events (CTCAE) to evaluate liver toxicity after
radiation therapy [25]. The Cancer Therapy Evaluation Program, Common
Terminology Criteria for Adverse Events (CTCAE), Version 3.0, defines grades 2,
3, 4, and 5 liver dysfunction as jaundice, asterixis, encephalopathy or coma, and
death, respectively. No grade 1 has been defined (Table 14.1) [19].

Table 14.1 CTCAE.V3 Definition


liver dysfunction criteria
Grade 1 –
Grade 2 Jaundice
Grade 3 Asterixis
Grade 4 Encephalopathy
Grade 5 Death
140 14 Radiation-Induced Liver Disease

14.6 Diagnosis

Patients with typical clinical picture and history of recent radiation therapy to the
liver included in treatment field have a high likelihood of having RILD.
In patients with intrahepatic cancer, progression of cancer should be ruled out
before making the diagnosis of RILD. An abdominal CT scan and paracentesis of
the ascites are typically performed as part of the differential diagnosis [3].
Ascitic fluid obtained by paracentesis is consistent with a transudate feature (the
serum to ascites albumin gradient > 1.1) with cytological negativity for malignant
cells [26].
Irradiated liver in RILD appears hypodense on non-contrast CT scans. This CT
finding is limited to radiation fields with well-defined linear margins in partial liver
irradiation with more than 45 Gy [27, 28]. The sharp margins of demarcation are not
seen in patients that received radiation through several non-axial and non-coplanar
portals. The hypodensity of the irradiated area most likely correlates with changes
of increased water content resulting from either edema or vascular congestion [29].
On contrast CT scans, irradiated areas are hypodense on the portal venous phase due
to hypoperfusion and decreased contrast inflow and become hyperdense on the
delayed phase obtained 4 min after contrast injection due to decreased venous drain-
age and stasis [30]. Maximal effect of radiation on liver appearance in CT images is
seen 2–3 months after completion of therapy and is reversible with return to normal
appearance [29].
The area of low density on CT has high signal intensity on the T2-weighted and
low intensity signal on the T1-weighted sequence of MR images [27, 31]. Contrast
enhancement of the irradiated area could be seen in MRI imaging after administra-
tion of gadopentetate dimeglumine (Gd-DTPA) due to the increased capillary per-
meability and chondroitin sulfate iron colloid (CSIC) due to hypofunction of the
reticuloendothelial system [32].
The irradiation area has diminished function that could be detected with radio-
isotope scan as the area with decreased radioisotope uptake. These functional scans
have the potential to quantify hepatocyte function to distinguish functional regions
of hepatocytes from nonfunctional zones. Technetium (99mTc) sulfur colloid (SC)
single-photon emission computed tomography is used as the imaging modality to
image liver function, and there is a correlation between differential SC uptake and
varying doses of radiation delivered [33].

14.7 Prevention

Prevention of RILD is paramount due to its fatal potential.


Three-dimensional treatment planning offers the potential to determine the liver
radiation dose and volume, RILD risk could be estimated using dose-volume histo-
gram (DVH) parameters, and modification of treatment planning should be consid-
ered if the RILD risk is high enough.
14.8 Management 141

With the availability of intensity-modulated radiotherapy (IMRT), stereotactic


body radiotherapy (SBRT), and image guidance in radiation treatment planning,
radiation can be delivered with better liver sparing.
Coa et al. recently have proposed a risk assessment strategy for radiation treat-
ment planning and re-optimization of the plan during therapy for intrahepatic radia-
tion treatment. The assessment of the patient’s individual portal vein perfusion
dose-response function during therapy and prediction of residual liver function after
radiation could allow for detection of patients at high risk for RILD and adjusting
the treatment plan [34, 35].
Amifostine is an organic thiophosphate that was developed to selectively protect
normal tissues against the toxicities of chemotherapy and radiation. Systemically or
regionally, administration of amifostine effectively protects hepatocytes from ion-
izing radiation damage without compromising the antitumor effect of radiation.
Feng et al. observed that the use of amifostine allows for a higher dose of whole
liver radiation to be safely administered and suggested the possibility of using ami-
fostine in combination with radiation for patients with focal liver disease undergo-
ing intensity-modulated radiotherapy or stereotactic body radiotherapy for focal
liver tumors [36, 37].
Theoretically, anticoagulation may exert a protective effect against acute radia-
tion injury by precluding fibrin clot formation and decrease the hepatic congestion.
Lightdale et al. observed a possible protective effect for warfarin in a small group of
Hodgkin’s disease patients [23]. Further evaluation of anticoagulation effect on
radiation hepatic injury appears warranted.
Exposure to ionizing radiation enhances production of reactive oxygen species
and decreases the levels of antioxidant in liver, whereas selenium and vitamin E
supplementation can modulate the changes in liver. Gençel et al. observed signifi-
cant decrease in oxidative stress in rat liver with selenium and vitamin E administra-
tion prior to a 7 Gy exposure relative to control animals [38].

14.8 Management

Treatment of RILD is primarily supportive and involves the use of diuretics for fluid
retention, analgesics for pain, paracentesis for tense ascites, correction of coagu-
lopathy, and steroids to prevent hepatic congestion [39].
Most patients respond to this therapy and symptoms resolve over the subsequent
l–2 months. A minority of patients develop jaundice, progressive ascites refractory
to paracentesis and diuretics, and coagulopathy. Although some of these patients
may recover, a substantial fraction will die due to liver failure [3].
Thrombolysis using tissue plasminogen activator (rh-tPA) and heparin remains
unproven for the treatment of hepatic veno-occlusive disease due to the risk of
bleeding in VOD and suboptimal response rate [40]. tPA/heparin should not be used
in patients with severe VOD with multi-organ failure (MOF) and should be given
early in VOD [39].
142 14 Radiation-Induced Liver Disease

Defibrotide is a deoxyribonucleic acid derivative extracted from mammalian


organs that has fibrinolytic and antithrombotic properties. It has been successfully
used to treat severe hepatic VOD with MOF in patients that have received cytotoxic
chemotherapy in preparation for bone marrow transplantation. Investigations of
defibrotide appear warranted in the setting of radiation-induced liver injury [39, 41,
42].
Radiation will cause hepatocyte injury and suppress liver regeneration.
Hematopoietic stem cells or hepatocytes with normal regenerative potential would
proliferate and repopulate the liver; it has been suggested that G-CSF-mobilized
CD34+ hematopoietic stem cells (HSCs) [43] and transplanted hepatocytes [44]
may ameliorate radiation-induced damage to liver by the ability to engraft into liver
tissue and generate hepatocytes.

References
1. Munden RF, Erasmus JJ, Smythe WR, Madewell JE, Forster KM, Stevens CW (2005)
Radiation injury to the liver after intensity-modulated radiation therapy in patients with meso-
thelioma: an unusual CT appearance. Am J Roentgenol 184(4):1091–1095
2. Khozouz RF, Huq SZ, Perry MC (2008) Radiation-induced liver disease. J Clin Oncol
26(29):4844–4845
3. Lawrence TS, Robertson JM, Anscher MS, Jirtle RL, Ensminger WD, Fajardo LF (1995)
Hepatic toxicity resulting from cancer treatment. Int J Radiat Oncol Biol Phys
31(5):1237–1248
4. Sempoux C, Horsmans Y, Geubel A, Fraikin J, Van Beers BE, Gigot JF, Lerut J, Rahier
J (1997) Severe radiation-induced liver disease following localized radiation therapy for bilio-
pancreatic carcinoma: activation of hepatic stellate cells as an early event. Hepatology
26(1):128–134
5. Wang S, Lee Y, Kim J, Hyun J, Lee K, Kim Y, Jung Y (2013) Potential role of Hedgehog
pathway in liver response to radiation. PLoS One 8(9):e74141
6. Olsen CC, Welsh J, Kavanagh BD, Franklin W, McCarter M, Cardenes HR, Gaspar LE,
Schefter TE (2009) Microscopic and macroscopic tumor and parenchymal effects of liver ste-
reotactic body radiotherapy. Int J Radiat Oncol Biol Phys 73(5):1414–1424
7. Cheng W, Xiao L, Ainiwaer A, Wang Y, Wu G, Mao R, Yang Y, Bao Y (2015) Molecular
responses of radiation-induced liver damage in rats. Mol Med Rep 11(4):2592–2600
8. Reed GB Jr, Cox AJ Jr (1966) The human liver after radiation injury. A form of veno-occlusive
disease. Am J Pathol 48(4):597–611
9. Cheng JC, JK W, Lee PC, Liu HS, Jian JJ, Lin YM, Sung JL, Jan GJ (2004) Biologic suscep-
tibility of hepatocellular carcinoma patients treated with radiotherapy to radiation-induced
liver disease. Int J Radiat Oncol Biol Phys 60:1502–1509
10. Lawrence TS, Ten Haken RK, Kessler ML, Robertson JM, Lyman JT, Lavigne ML, Brown
MB, DuRoss DJ, Andrews JC, Ensminger WD et al (1992) The use of 3-D dose volume analy-
sis to predict radiation hepatitis. Int J Radiat Oncol Biol Phys 23(4):781–788
11. Emami B, Lyman J, Brown A, Coia L, Goitein M, Munzenrider JE, Shank B, Solin LJ, Wesson
M (1991) Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys
21(1):109–122
12. Liang SX, Zhu XD, ZY X, Zhu J, Zhao JD, HJ L, Yang YL, Chen L, Wang AY, XL F, Jiang
GL (2006) Radiation-induced liver disease in three-dimensional conformal radiation therapy
for primary liver carcinoma: the risk factors and hepatic radiation tolerance. Int J Radiat Oncol
Biol Phys 65(2):426–434
References 143

13. Haddad E, Le Bourgeois JP, Kuentz M, Lobo P (1983) Liver complications in lymphomas
treated with a combination of chemotherapy and radiotherapy: preliminary results. Int J Radiat
Oncol Biol Phys 9(9):1313–1319
14. McCracken JD, Weatherall TJ, Oishi N, Janaki L, Boyer C (1985) Adjuvant intrahepatic che-
motherapy with mitomycin and 5-FU combined with hepatic irradiation in high-risk patients
with carcinoma of the colon: a Southwest Oncology Group phase II pilot study. Cancer Treat
Rep 69(1):129–131
15. Byfield JE, Barone RM, Frankel SS, Sharp TR (1984) Treatment with combined intra-arterial
5-FUdR infusion and whole-liver radiation for colon carcinoma metastatic to the liver.
Preliminary results. Am J Clin Oncol 7(4):319–325
16. Friedman M, Cassidy M, Levine M, Phillips T, Spivack S, Resser KJ (1979) Combined modal-
ity therapy of hepatic metastasis. Cancer 44:906–913
17. Lokich J, Kinsella T, Perri J, Malcolm A, Clouse M (1981) Concomitant hepatic radiation and
intraarterial fluorinated pyrimidine therapy: correlation of liver scan, liver function tests, and
plasma CEA with tumor response. Cancer 48(12):2569–2574
18. Yin H, Lu K, Qiao W-B, Zhang H-Y, Sun D, You Q-S (2014) Whole-liver radiotherapy concur-
rent with chemotherapy as a palliative treatment for colorectal patients with massive and mul-
tiple liver metastases: a retrospective study. Asian Pac J Cancer Prev 15(4):1597–1602
19. Pan CC, Kavanagh BD, Dawson LA, Li XA, Das SK, Miften M, Ten Haken RK (2010)
Radiation-associated liver injury. Int J Radiat Oncol Biol Phys 76(3 Suppl):S94–100
20. Tomé WA, Fenwick JD (2004) Analysis of radiation-induced liver disease using the Lyman
NTCP model: in regard to Dawson et al. IJROBP 2002;53:810-821. Int J Radiat Oncol Biol
Phys 58(4):1318–1319
21. Seong J, Park HC, Han KH, Chon CY (2003) Clinical results and prognostic factors in radio-
therapy for unresectable hepatocellular carcinoma: a retrospective study of 158 patients. Int
J Radiat Oncol Biol Phys 55(2):329–336
22. Dawson LA, Normolle D, Balter JM, McGinn CJ, Lawrence TS, Ten Haken RK (2002)
Analysis of radiation-induced liver disease using the Lyman NTCP model. Int J Radiat Oncol
Biol Phys 53(4):810–821
23. Lightdale CJ, Wasser J, Coleman M, Brower M, Tefft M, Pasmantier M (1979) Anticoagulation
and high dose liver radiation: a preliminary report. Cancer 43(1):174–181
24. Tefft M, Mitus A, Das L, Vawter GF, Filler RM (1970) Irradiation of the liver in children:
review of experience in the acute and chronic phases, and in the intact normal and partially
resected. Am J Roentgenol Radium Ther Nucl Med 108(2):365–385
25. Li G, Wang J, Hu W, Zhang Z (2015) Radiation-induced liver injury in three-dimensional
conformal radiation therapy (3D-CRT) for postoperative or locoregional recurrent gastric can-
cer: risk factors and dose limitations. PLoS One 10(8):e0136288
26. Russell AH, Clyde C, Wasserman TH, Turner SS, Rotman M (1993) Accelerated hyperfrac-
tionated hepatic irradiation in the management of patients with liver metastases: results of the
RTOG dose escalating protocol. Int J Radiat Oncol Biol Phys 27(1):117–123
27. Kwek JW, Iyer RB, Dunnington J, Faria S, Silverman PM (2006) Spectrum of imaging find-
ings in the abdomen after radiotherapy. AJR Am J Roentgenol 187(5):1204–1211
28. Munden RF, Erasmus JJ, Smythe WR, Madewell JE, Forster KM, Stevens CW (2005)
Radiation injury to the liver after intensity-modulated radiation therapy in patients with meso-
thelioma: an unusual CT appearance. AJR Am J Roentgenol 184(4):1091–1095
29. Yamasaki SA, Marn CS, Francis IR, Robertson JM, Lawrence TS (1995) High-dose localized
radiation therapy for treatment of hepatic malignant tumors: CT findings and their relation to
radiation hepatitis. AJR Am J Roentgenol 165(1):79–84
30. Willemart S, Nicaise N, Struyven J, van Gansbeke D (2000) Acute radiation-induced hepatic
injury: evaluation by triphasic contrast enhanced helical CT. Br J Radiol 73(869):544–546
31. Unger EC, Lee JK, Weyman PJ (1987) CT and MR imaging of radiation hepatitis. J Comput
Assist Tomogr 11(2):264–268
144 14 Radiation-Induced Liver Disease

32. Suto Y, Kato T, Yoshida K, Sugihara S, Kamba M, Ohta YMRI (1996) Findings in radiation-­
induced hepatic injuries. Yonago Acta Med 39:127–134
33. Chapman TR, Kumarapeli AR, Nyflot MJ, Bowen SR, Yeung RS, Vesselle HJ, Yeh MM,
Apisarnthanarax S (2015) Functional imaging of radiation liver injury in a liver metastasis
patient: imaging and pathologic correlation. J Gastrointest Oncol 6(3):E44–E47
34. Cao Y, Pan C, Balter JM, Platt JF, Francis IR, Knol JA, Normolle D, Ben-Josef E, Ten Haken
RK, Lawrence TS (2008) Liver function after irradiation based on computed tomographic
portal vein perfusion imaging. Int J Radiat Oncol Biol Phys 70(1):154–160
35. Cao Y, Platt JF, Francis IR, Balter JM, Pan C, Normolle D, Ben-Josef E, Haken RK, Lawrence
TS (2007) The prediction of radiation-induced liver dysfunction using a local dose and regional
venous perfusion model. Med Phys 34(2):604–612
36. Feng M, Smith DE, Normolle DP, Knol JA, Pan CC, Ben-Josef E, Lu Z, Feng MR, Chen J,
Ensminger W, Lawrence TS (2012) A phase I clinical and pharmacology study using amifos-
tine as a radioprotector in dose-escalated whole liver radiation therapy. Int J Radiat Oncol Biol
Phys 83(5):1441–1447
37. Symon Z, Levi M, Ensminger WD, Smith DE, Lawrence TS (2001 Jun 1) Selective radiopro-
tection of hepatocytes by systemic and portal vein infusions of amifostine in a rat liver tumor
model. Int J Radiat Oncol Biol Phys 50(2):473–478
38. Gençel O, Naziroglu M, Celik O, Yalman K, Bayram D (2010) Selenium and vitamin E modu-
lates radiation-induced liver toxicity in pregnant and nonpregnant rat: effects of colemanite
and hematite shielding. Biol Trace Elem Res 135(1–3):253–263
39. Guha C, Kavanagh BD (2011) Hepatic radiation toxicity: avoidance and amelioration. Semin
Radiat Oncol 21(4):256–263
40. Bearman SI, Lee JL, Barón AE, McDonald GB (1997) Treatment of hepatic veno-occlusive
disease with recombinant human tissue plasminogen activator and heparin in 42 marrow trans-
plant patients. Blood 89(5):1501–1506
41. Palmer KJ, Goa KL (1993) Defibrotide. A review of its pharmacodynamic and pharmacoki-
netic properties, and therapeutic use in vascular disorders. Drugs 45(2):259–294
42. Benimetskaya L, Wu S, Voskresenskiy AM, Echart C, Zhou JF, Shin J, Iacobelli M, Richardson
P, Ayyanar K, Stein CA (2008) Angiogenesis alteration by defibrotide: implications for its
mechanism of action in severe hepatic veno-occlusive disease. Blood 112(10):4343–4352
43. Li N, Zhang L, Li H, Fang B (2010) Human CD34+ cells mobilized by granulocyte colony-­
stimulating factor ameliorate radiation-induced liver damage in mice. Stem Cell Res Ther
1(3):22
44. Guha C, Sharma A, Gupta S, Alfieri A, Gorla GR, Gagandeep S, Sokhi R, Roy-Chowdhury N,
Tanaka KE, Vikram B, Roy-Chowdhury J (1999) Amelioration of radiation-induced liver dam-
age in partially hepatectomized rats by hepatocyte transplantation. Cancer Res
59(23):5871–5874
Enteritis
15

Acute gastrointestinal complications occur in 80% of patients in pelvic radiation


therapy [1]. The small intestine appears to be more sensitive to radiation than the
colon and rectum [2] and is also more frequently exposed incidentally to radiation
during treatment of adjacent organs because of its situation within the peritoneal
cavity.
Different intestinal side effects were reported in the Postoperative Radiation
Therapy in Endometrial Cancer (PORTEC-2) Trial, which found that among 214
women with endometrial cancer treated with external beam radiation therapy after
total abdominal hysterectomy and oophorectomy, the rates of diarrhea, fecal leak-
age, rectal blood loss, and bloating were all increased from the baseline at a rate
of 22%, 5.3%, 1.8%, and 0.7% at 1–4 weeks after the end of treatment, respec-
tively [3].
In a study of 107 patients with gynecologic, urologic, or gastrointestinal cancer
within the pelvis that underwent radiation therapy, acute gastrointestinal toxicities
were assessed during the treatment. It was found that 94% of patients developed
altered bowel habits, 80% loose stool, 74% frequency, 65% difficult gas, 60%
pain, more than 48% distress, 44% tenesmus, more than 40% restrictions in daily
activity, 39% urgency, 37% fecal incontinence, and 40% required antidiarrheal
medication [4].

15.1 Mechanism

The intestinal epithelium is a single layer of columnar cells containing crypt-villous


units. Villous protrusions increase the absorptive surface of the small intestine.
Undifferentiated rapidly proliferating stem cells are in crypts that are dividing and
converted to other differentiated cells including enterocytes that migrate to the vil-
lous epithelium. The crypt-villous units are substantially degenerated in organiza-
tion after radiation injury.

© Springer International Publishing AG 2017 145


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_15
146 15 Enteritis

Radiation initially induces mitotic arrest in stem cells. Cryptal surface cell lay-
ers become swollen with nuclear shrinkage and degenerative cytoplasmic changes
and necrosis occur. Villi become shortened with a decrease in enterocyte cell num-
ber and microvilli length and number, leading to absorptive capacity reduction.
Acute inflammatory reaction, leukocyte migration (mostly polymorphonuclear leu-
kocytes and eosinophils), increased microvascular permeability, hyperemia, and
edema all occur causing tissue damage and mucosal breakdown, and, ultimately,
ulceration will develop [2]. Small bowel bacterial overgrowth also may occur dur-
ing pelvic radiation therapy in some patients and is likely related to gastrointestinal
symptoms [5].
Damaged epithelium results in impaired absorption of bile salts, fats, carbohy-
drates, proteins, and vitamin B12 during radiation therapy [6].
The small bowel motility pattern also changes during radiation therapy. The
exact mechanism of this change remains unclear. Some proposed mechanisms are
related to alteration in neurotransmitters release, activity and sensitivity, mucosal
injury, and change in water and electrolyte absorption or alterations in the function
of the smooth muscle cells [7, 8].

15.2 Timing

Symptoms such as cramping and diarrhea usually develop about 1–2 weeks after the
start of radiation therapy (5–12 Gy in fractionated radiation therapy) [9, 10], and its
peak is reached by the fourth to fifth week [10]. Symptoms gradually decrease in
severity within 2–6 weeks after treatment [11, 12], but it could last longer [13].

15.3 Risk Factors

Radiation dose and the volume of small bowel exposed to radiation correlate with
incidence and severity of acute enteritis [14, 15]. Small bowel loops are freely mov-
ing within the peritoneal cavity, and defining exact dose-volume parameters that are
consistent during the entire course of radiation therapy is not possible. Kavanagh
et al. found that severe acute small bowel toxicity could be prevented if the volume
of small bowel receiving radiation doses more than 15 Gy is kept under 120 mL
(D15 < 120 mL) when bowel loops are contoured as normal tissue volume; how-
ever, if the entire peritoneal cavity is considered as small bowel volume, the volume
receiving more than 45 Gy should be kept under 195 mL (D45 < 195 mL) [9].
Chemotherapy concomitant with radiation therapy increases the risk of all grades
of acute enteritis, especially higher grades [16–18]. A systematic review of acute
and late toxicity of concomitant chemoradiation for cervical cancer has reported a
twofold increase in acute grade 3 and 4 gastrointestinal toxicities in patients treated
with chemoradiation therapy rather than radiation alone [18].
Inflammatory bowel disease (IBD) is a known risk factor for small bowel
radiation-­induced toxicity, though very scant data are available. Irradiation can
15.5 Scoring 147

exacerbate IBD symptoms; IBD also increases the acute and late side effects of
radiation therapy significantly. Acute gastrointestinal toxicity that results in radia-
tion cessation was more than 20% in a retrospective study of 28 patients with IBD
that underwent external beam abdominal or pelvic irradiation, and there was no
difference in acute toxicities between patients with Crohn’s disease and ulcerative
colitis. It is recommended that radiation should be used with caution in these
patients [19]. If the radiation therapy is mandatory, efforts should be made to spare
the normal gastrointestinal tissue. Selecting appropriate patient position, computed
tomography (CT) simulator with contrast agent, and using more conformal treat-
ment techniques are effective to determine gastrointestinal tract irradiated volume
and doses.
Previous abdominal surgery is related to risk of acute enteritis due to a larger
volume and fixed loop of small bowel within the radiation field [20, 21]. Type of
surgery may be important in this view. In a retrospective study of 120 patients with
rectal cancer that received chemoradiation, severe radiation-induced diarrhea was
more common in patients with sphincter-preserving procedures versus abdomino-
perineal resection [22].
Low body mass index [14] and female gender [22] have been reported to be
associated with radiation-induced enteritis.
Diabetes, hypertension, age, field design, and number (two-, three-, or four-field
technique) have not been reported to significantly increase acute toxicity of pelvic
radiation [11].

15.4 Symptoms and Diagnosis

Acute injury to small bowel manifests mainly with diarrhea with or without abdom-
inal cramping during or shortly after radiation therapy. The diarrhea secondary to
acute radiation enteritis may be associated with decreased absorption of bile salts,
vitamin B12, lactose, fat, and more rapid small intestinal transit [13]. Nausea and
vomiting, bloating, and loss of appetite may occur [10, 13]. Weight loss can be a
secondary finding. Rarely, obstructive symptoms or bowel perforation due to pro-
found acute enteritis may occur and require surgical intervention, especially in
patients with underlying inflammatory disease [23].

15.5 Scoring

Radiation Therapy Oncology Group (RTOG) and European Organization for


Research and Treatment of Cancer (EORTC) have classified all symptoms
related to lower gastrointestinal and pelvic tissue in the single scoring category
(Table 15.1) [24].
Diarrhea is the main symptom of radiation-induced enteritis, and Table 15.2
shows the Common Terminology Criteria for Adverse Events (CTCAE) v4, which
classifies diarrhea into five grades [25].
148 15 Enteritis

Table 15.1 RTOG/EORTC lower gastrointestinal toxicity


Definition
Grade 1 Increased frequency or change in quality of bowel habits not requiring medication/
rectal discomfort not requiring analgesics
Grade 2 Diarrhea requiring parasympatheticolytic drugs (e.g., Lomotil)/mucous discharge not
necessitating sanitary pads/rectal or abdominal pain requiring analgesics
Grade 3 Diarrhea requiring parenteral support/severe mucous or bloody discharge
necessitating sanitary pads/abdominal distention (flat plate radiograph demonstrates
distended bowel loops)
Grade 4 Acute or subacute obstruction, fistula, or perforation; GI bleeding requiring
transfusion; abdominal pain or tenesmus requiring tube decompression or bowel
diversion

Table 15.2 CTCAE v4, diarrhea scoring


Definition
Grade 1 <4 stools per day over baseline; mild increase in ostomy output compared to baseline
Grade 2 Four to six stools per day over baseline; moderate increase in ostomy output
compared to baseline
Grade 3 ≥7 stools per day over baseline; incontinence; hospitalization indicated; severe
increase in ostomy output compared to baseline; limiting self-care ADL (activities of
daily living)
Grade 4 Life-threatening consequences; urgent intervention indicated
Grade 5 Death

15.6 Prevention

Frequently, different volumes of small bowel have to be included in irradiation por-


tals when abdominal or pelvic tumors are treated with radiation therapy. It should be
attempted to reduce the bowel volume in radiation portals or at least exclude from
high-dose exposure by various techniques such as patient positioning, use of a belly
board, treatment with a full bladder, or using multiple-field techniques [26–28].
Prone positioning on belly-board devices is an appropriate way to spare the small
bowel in both 3D-CRT (conformal radiation therapy) and IMRT (intensity modu-
lated radiotherapy) treatment plans [29, 30], although it may increase large bowel
volume in the treatment field when the limited arc technique is used [29].
With the introduction of new techniques for radiation delivery like IMRT, small
bowel volume with high radiation doses and resultant radiation toxicity have
decreased [31–34].
A number of surgical techniques have been applied for bowel exclusion from the
radiation field in patients with postoperative pelvic radiation therapy that have used
prosthetic materials (e.g., absorbable mesh or saline-filled tissue expanders) or
omentum [35–37]. These interventions are not routinely used in many centers [10].
European Society for Medical Oncology (ESMO) clinical practice guideline rec-
ommends using systemic sulfasalazine at a dose of 500 mg administered orally
twice a day to prevent radiation-induced enteropathy in patients receiving radiation
therapy to the pelvis.
15.7 Management 149

There are many preventive options that have been discussed in the literature but
cannot be recommended due to insufficient evidence [38–46].
Elemental nutritional formulas, low-lactose, low-fat, and low-fiber diets, and
probiotic preparations currently cannot be considered in clinical practice as pro-
phylactic interventions until better determination of their safety and efficacy is
made [42].
Amifostine [39] and vitamin E [40] before radiation therapy as well as angioten-
sin I-converting enzyme inhibitors (ACEi) and 3-hydroxy-methylglutaryl coenzyme
A reductase inhibitors (statins) [41] during radiation therapy appear to provide pro-
tection against lower gastrointestinal toxicity. These primary promising results need
to be established in future studies.
Glutamine is a major source of energy for intestinal epithelial cells and is neces-
sary for cellular proliferation, mucosal cell integrity, and immunological protection
of the small bowel [46, 47]. A protective effect of oral glutamine on the small bowel
mucosa has been suggested in several studies with contradictory results [43–46].
There is insufficient data to draw a definitive conclusion.
Different cytokines and peptides have been evaluated for protecting the gastrointes-
tinal tract from radiation side effects. The protection effects of insulin-like growth
factor-I (IGF-I) on small intestinal mucosal radiation damage have been proposed in
animal studies that need further evaluation [48–51]. Several other agents including
immunomodulators (e.g., orazipone, interleukin-11) [37, 52], trophic agents like gluca-
gon-like peptide-2 and its dipeptidyl peptidase-4 (DPP-IV) resistant analog teduglutide
[53], and other growth factors [54, 55] are proposed to modulate small intestine injury.
A circadian diurnal variation in the number of apoptotic cells in the intestinal
crypt has been shown in animal studies to affect radiation-induced enteritis fre-
quency [56, 57]. In a randomized study of 229 patients with cervical carcinoma that
were treated with radiation therapy in the morning (8:00–10:00 AM) or evening
(6:00–8:00 PM), the overall incidence and severity of diarrhea in patients increased
with the morning treatment as compared with the evening [38]. Currently there is no
recommendation for this observation in clinical practice.

15.7 Management

The key point in the approach to patients with radiation-induced diarrhea is deter-
mining the severity of diarrhea and patient’s general condition. Mild to moderate
diarrhea without any other significant symptoms and signs can be treated in the
outpatient setting with dietary modification, antidiarrheals, and antispasmodics (see
below). All of these patients should be examined every 24 h [58].
Patients with severe diarrhea or persistent mild to moderate diarrhea or presence
of some cautionary factors such as fever, dehydration, severe abdominal cramping,
nausea and vomiting, sepsis, neutropenia, or blood in the stool should be hospitalized
to be immediately worked up with complete blood count, stool exam and culture,
serum electrolyte and renal function test and aggressive treatment with octreotide,
antibiotic therapy, and fluid and electrolyte replacement, as indicated [58].
150 15 Enteritis

15.7.1 Dietary Modification

Patients should be encouraged for maintenance of adequate hydration (35 ml/kg/


day) and eating small, frequent high-protein food. Chocolate, alcohol, caffeine,
sorbitol-containing substances, beans, high osmolar beverage, foods with insoluble
fiber including skins of fruits and raw vegetables, whole-grain and multigrain foods,
strong spices, and extreme hot/cold food should be avoided. A transient lactose
intolerance may occur in up to 45% of patients that respond to the avoidance of milk
and milk products (a lactose-restricted diet) [5, 13, 59]. Fat malabsorption also
occurs in many cases that provides a rationale for the use of low-fat diet [10].
Soluble fiber may help build stool consistency (e.g., fruits without skins, oat, bran,
and barley) [59].

15.7.2 Antidiarrheals

Loperamide is often used as a first-line antidiarrheal agent. If diarrhea does not


respond to the use of loperamide after 48 h, octreotide has been shown to be
effective [10]. It has been reported that octreotide seems to be more
effective than diphenoxylate and atropine in the treatment of radiation-induced
diarrhea [60].
Loperamide should be started with an initial dose of 4 mg, then 2 mg after each
loose stool or every 4 h; treatment should be continued throughout the duration of
radiation therapy with standard dose. If diarrhea does not resolve after 24 h,
increasing the dose 2 mg every 2 h should be considered, not to exceed 16 mg/day
(8 mg/day for self-medication). These patients should be further assessed with
stool analysis and blood work-up. Oral supplementation or intravenous hydration
may be indicated in patients based on their general condition, lab test results, and
dehydration status. Treatment delay may be required based on physician judgment.
If no improvement has been seen within 48 h, loperamide is discontinued and
second-line treatment with octereotide 100 μg subcutaneously (SC) three times
daily will start [10, 59, 61].
Cholestyramine is a nonabsorbable high molecular resin that binds bile salts irre-
versibly. Some studies have supported cholestyramine efficacy in the treatment of
acute and chronic diarrhea in patients treated with radiation therapy [62, 63].
It has been observed that the inhibition of prostaglandin biosynthesis by sul-
phasalazine agents may relieve diarrhea caused by other reasons than radiation, and
this led to assessing aspirin in radiation-induced diarrhea. There is limited evidence
in this regard and no guideline recommended for its use [62, 64, 65].

15.8 Antispasmodics

An anticholinergic antispasmodic agent could alleviate bowel cramping.


References 151

References
1. Andreyev J (2005) Gastrointestinal complications of pelvic radiotherapy: are they of any
importance? Gut 54(8):1051–1054
2. Carr KE (2001) Effects of radiation damage on intestinal morphology. Int Rev Cytol
208:1–119
3. Nout RA, Putter H, Jürgenliemk-Schulz IM, Jobsen JJ, Lutgens LC, van der Steen-Banasik
EM et al (2009) Quality of life after pelvic radiotherapy or vaginal brachytherapy for endome-
trial cancer: first results of the randomized PORTEC-2 trial. J Clin Oncol 27(21):3547–3556
4. Khalid U, McGough C, Hackett C, Blake P, Harrington KJ, Khoo VS et al (2006) A modified
inflammatory bowel disease questionnaire and the Vaizey Incontinence questionnaire are more
sensitive measures of acute gastrointestinal toxicity during pelvic radiotherapy than RTOG
grading. Int J Radiat Oncol Biol Phys 64(5):1432–1441
5. Wedlake L, Thomas K, McGough C, Andreyev H (2008) Small bowel bacterial overgrowth
and lactose intolerance during radical pelvic radiotherapy: an observational study. Eur J Cancer
44(15):2212–2217
6. Berthrong M, Fajardo LF (1981) Radiation injury in surgical pathology: Part II. Alimentary
tract. Am J Surg Pathol 5(2):153–178
7. Otterson MF, Sarna SK, Moulder JE (1988) Effects of fractionated doses of ionizing radiation
on small intestinal motor activity. Gastroenterology 95(5):1249–1257
8. Summers R, Kent TH, Osborne J (1970) Effects of drugs, ileal obstruction, and irradiation on
rat gastrointestinal propulsion. University of Iowa, Iowa City
9. Kavanagh BD, Pan CC, Dawson LA, Das SK, Li XA, Ten Haken RK et al (2010) Radiation
dose-volume effects in the stomach and small bowel. Int J Radiat Oncol Biol Phys
76(3):S101–S1S7
10. Stacey R, Green JT (2014) Radiation-induced small bowel disease: latest developments and
clinical guidance. Ther Adv Chronic Dis 5(1):15–29
11. Creutzberg CL, van Putten WL, Koper PC, Lybeert ML, Jobsen JJ, Wárlám-Rodenhuis CC, De
Winter KA, Lutgens LC, van den Bergh AC, van der Steen-Banasik E, Beerman H, van Lent
M, PORTEC Study Group, The Postoperative Radiation Therapy in Endometrial Carcinoma
(2001) The morbidity of treatment for patients with Stage I endometrial cancer: results from a
randomized trial. Int J Radiat Oncol Biol Phys 51(5):1246–1255
12. Newman A, Blendis L, Katsaris J, Charlesworth M, Walter L (1973) Small-intestinal injury in
women who have had pelvic radiotherapy. Lancet 302(7844):1471–1473
13. Yeoh E, Horowitz M, Russo A, Muecke T, Robb T, Maddox A et al (1993) Effect of pelvic
irradiation on gastrointestinal function: a prospective longitudinal study. Am J Med
95(4):397–406
14. Minsky BD, Conti JA, Huang Y, Knopf K (1995) Relationship of acute gastrointestinal toxicity
and the volume of irradiated small bowel in patients receiving combined modality therapy for
rectal cancer. J Clin Oncol 13(6):1409–1416
15. Roeske JC, Bonta D, Mell LK, Lujan AE, Mundt AJ (2003) A dosimetric analysis of acute
gastrointestinal toxicity in women receiving intensity-modulated whole-pelvic radiation ther-
apy. Radiother Oncol 69(2):201–207
16. Keys HM, Bundy BN, Stehman FB, Muderspach LI, Chafe WE, Suggs CL et al (1999)
Cisplatin, radiation, and adjuvant hysterectomy compared with radiation and adjuvant hyster-
ectomy for bulky stage IB cervical carcinoma. N Engl J Med 340(15):1154–1161
17. Morris M, Eifel PJ, Lu J, Grigsby PW, Levenback C, Stevens RE et al (1999) Pelvic radiation
with concurrent chemotherapy compared with pelvic and para-aortic radiation for high-risk
cervical cancer. N Engl J Med 340(15):1137–1143
18. Kirwan JM, Symonds P, Green JA, Tierney J, Collingwood M, Williams CJ (2003) A system-
atic review of acute and late toxicity of concomitant chemoradiation for cervical cancer.
Radiother Oncol 68(3):217–226
152 15 Enteritis

19. Willett CG, Ooi C-J, Zietman AL, Menon V, Goldberg S, Sands BE et al (2000) Acute and late
toxicity of patients with inflammatory bowel disease undergoing irradiation for abdominal and
pelvic neoplasms. Int J Radiat Oncol Biol Phys 46(4):995–998
20. Gallagher MJ, Brereton HD, Rostock RA, Zero JM, Zekoski DA, Poyss LF et al (1986) A
prospective study of treatment techniques to minimize the volume of pelvic small bowel with
reduction of acute and late effects associated with pelvic irradiation. Int J Radiat Oncol Biol
Phys 12(9):1565–1573
21. Huang E-Y, Sung C-C, Ko S-F, Wang C-J, Yang KD (2007) The different volume effects of
small-bowel toxicity during pelvic irradiation between gynecologic patients with and without
abdominal surgery: a prospective study with computed tomography-based dosimetry. Int
J Radiat Oncol Biol Phys 69(3):732–739
22. Rödel C, Fietkau R, Keilholz L, Grabenbauer G, Kessler H, Martus P et al (1997) The acute
toxicity of the simultaneous radiochemotherapy of rectal carcinoma. Strahlenther Onkol
173(8):415–421
23. Schofield PF, Holden D, Carr N (1983) Bowel disease after radiotherapy. J R Soc Med
76(6):463–466
24. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
25. NCI, NIH, DHHS (2009) Common terminology criteria for adverse events v4.0. National
Cancer Institute, Bethesda, MD
26. Gunderson LL, Russell AH, Llewellyn HJ, Doppke KP, Tepper JE (1985) Treatment planning
for colorectal cancer: radiation and surgical techniques and value of small-bowel films. Int
J Radiat Oncol Biol Phys 11(7):1379–1393
27. Green N (1983) The avoidance of small intestine injury in gynecologic cancer. Int J Radiat
Oncol Biol Phys 9(9):1385–1390
28. Acker JC, Marks LB (1995) The lack of impact of pelvic irradiation on small bowel mobility:
implications for radiotherapy treatment planning. Int J Radiat Oncol Biol Phys
32(5):1473–1475
29. Adli M, Mayr NA, Kaiser HS, Skwarchuk MW, Meeks SL, Mardirossian G et al (2003) Does
prone positioning reduce small bowel dose in pelvic radiation with intensity-modulated radio-
therapy for gynecologic cancer? Int J Radiat Oncol Biol Phys 57(1):230–238
30. Martin J, Fitzpatrick K, Horan G, McCloy R, Buckney S, O’Neill L et al (2005) Treatment
with a belly-board device significantly reduces the volume of small bowel irradiated and
results in low acute toxicity in adjuvant radiotherapy for gynecologic cancer: results of a pro-
spective study. Radiother Oncol 74(3):267–274
31. Urbano MTG, Henrys AJ, Adams EJ, Norman AR, Bedford JL, Harrington KJ et al (2006)
Intensity-modulated radiotherapy in patients with locally advanced rectal cancer reduces vol-
ume of bowel treated to high dose levels. Int J Radiat Oncol Biol Phys 65(3):907–916
32. Kachnic LA (2007) Adjuvant chemoradiation for localized rectal cancer: current trends and
future directions. Gastrointest Cancer Res 1(4 Suppl 2):S64–S72
33. Mundt AJ, Roeske JC, Lujan AE, Yamada SD, Waggoner SE, Fleming G et al (2001) Initial
clinical experience with intensity-modulated whole-pelvis radiation therapy in women with
gynecologic malignancies. Gynecol Oncol 82(3):456–463
34. Mundt AJ, Lujan AE, Rotmensch J, Waggoner SE, Yamada SD, Fleming G et al (2002)
Intensity-modulated whole pelvic radiotherapy in women with gynecologic malignancies. Int
J Radiat Oncol Biol Phys 52(5):1330–1337
35. Nguyen B, Hamper U (1997) Pelvic silicone prosthesis for prevention of radiation enteritis:
US and CT features. Abdom Imaging 22(2):175–177
36. Dasmahapatra KS, Swaminathan AP (1991) The use of a biodegradable mesh to prevent
radiation-­associated small-bowel injury. Arch Surg 126(3):366–369
37. Choi HJ, Lee HS (1995) Effect of omental pedicle hammock in protection against radiation-­
induced enteropathy in patients with rectal cancer. Dis Colon Rectum 38(3):276–280
References 153

38. Shukla P, Gupta D, Bisht SS, Pant MC, Bhatt ML, Gupta R et al (2010) Circadian variation in
radiation-induced intestinal mucositis in patients with cervical carcinoma. Cancer
116(8):2031–2035
39. Athanassiou H, Antonadou D, Coliarakis N, Kouveli A, Synodinou M, Paraskevaidis M et al
(2003) Protective effect of amifostine during fractionated radiotherapy in patients with pelvic
carcinomas: results of a randomized trial. Int J Radiat Oncol Biol Phys 56(4):1154–1160
40. Empey LR, Papp JD, Jewell LD, Fedorak RN (1992) Mucosal protective effects of vitamin E
and misoprostol during acute radiation-induced enteritis in rats. Dig Dis Sci 37(2):205–214
41. Wedlake LJ, Silia F, Benton B, Lalji A, Thomas K, Dearnaley DP et al (2012) Evaluating the
efficacy of statins and ACE-inhibitors in reducing gastrointestinal toxicity in patients receiving
radiotherapy for pelvic malignancies. Eur J Cancer 48(14):2117–2124
42. Wedlake L, Shaw C, Whelan K, Andreyev H (2013) Systematic review: the efficacy of nutri-
tional interventions to counteract acute gastrointestinal toxicity during therapeutic pelvic
radiotherapy. Aliment Pharmacol Ther 37(11):1046–1056
43. Klimberg VS, Souba WW, Dolson DJ, Salloum RM, Hautamaki RD, Plumley DA et al (1990)
Prophylactic glutamine protects the intestinal mucosa from radiation injury. Cancer
66(1):62–68
44. Vidal-Casariego A, Calleja-Fernández A, Cano-Rodríguez I, Cordido F, Ballesteros-Pomar
MD (2015) Effects of oral glutamine during abdominal radiotherapy on chronic radiation
enteritis: a randomized controlled trial. Nutrition 31(1):200–204
45. Conejo IM, Castillejo AR, de Dios NR, Arnalot PF, Latiesas JS, Galán JL et al (2011)
Prevention of acute radiation enteritis: efficacy and tolerance of glutamine. Clin Transl Oncol
13(10):760–763
46. Vidal-Casariego A, Calleja-Fernández A, de Urbina-González JJO, Cano-Rodríguez I,
Cordido F, Ballesteros-Pomar MD (2014) Efficacy of glutamine in the prevention of acute
radiation enteritis: a randomized controlled trial. J Parenter Enteral Nutr 38(2):205–213
47. Rao R, Samak G (2012) Role of glutamine in protection of intestinal epithelial tight junctions.
J Epithel Biol Pharmacol 5(Suppl 1-M7):47–54
48. Vazquez I, Gomez-De-Segura IA, Grande AG, Escribano A, Gonzalez-Gancedo P, Gomez A
et al (1999) Protective effect of enriched diet plus growth hormone administration on radiation-­
induced intestinal injury and on its evolutionary pattern in the rat. Dig Dis Sci
44(11):2350–2358
49. Wilkins HR, Ohneda K, Keku TO, D’Ercole AJ, Fuller CR, Williams KL et al (2002) Reduction
of spontaneous and irradiation-induced apoptosis in small intestine of IGF-I transgenic mice.
Am J Physiol Gastrointest Liver Physiol 283(2):G457–G464
50. Mylonas PG, Matsouka PT, Papandoniou EV, Vagianos C, Kalfarentzos F, Alexandrides TK
(2000) Growth hormone and insulin-like growth factor I protect intestinal cells from radiation
induced apoptosis. Mol Cell Endocrinol 160(1):115–122
51. Howarth G, Fraser R, Frisby C, Schirmer M, Yeoh E (1997) Effects of insulin-like
growth factor-I administration on radiation enteritis in rats. Scand J Gastroenterol
32(11):1118–1124
52. Boerma M, Wang J, Burnett AF, Santin AD, Roman JJ, Hauer-Jensen M (2007) Local admin-
istration of interleukin-11 ameliorates intestinal radiation injury in rats. Cancer Res
67(19):9501–9506
53. Booth C, Booth D, Williamson S, Demchyshyn L, Potten CS (2004) Teduglutide ([Gly2] GLP-­
2) protects small intestinal stem cells from radiation damage. Cell Prolif 37(6):385–400
54. Khan WB, Shui C, Ning S, Knox SJ (1997) Enhancement of murine intestinal stem cell sur-
vival after irradiation by keratinocyte growth factor. Radiat Res 148(3):248–253
55. Matthews MA, Watkins D, Darbyshire A, Carson WE, Besner GE (2013) Heparin-binding
EGF-like growth factor (HB-EGF) protects the intestines from radiation therapy-induced
intestinal injury. J Pediatr Surg 48(6):1316–1322
56. Ijiri K, Potten C (1990) The circadian rhythm for the number and sensitivity of radiation-­
induced apoptosis in the crypts of mouse small intestine. Int J Radiat Biol 58(1):165–175
154 15 Enteritis

57. Ijiri K, Potten CS (1988) Circadian rhythms in the incidence of apoptotic cells and number of
clonogenic cells in intestinal crypts after radiation using normal and reversed light conditions.
Int J Radiat Biol 53(5):717–727
58. Lawrence TS, Rosenberg SA (2015) DeVita, Hellman, and Rosenberg’s cancer: principles &
practice of oncology. Lippincott Williams & Wilkins, Philadelphia, PA
59. Halperin EC, Brady LW, Wazer DE, Perez CA (2013) Perez & Brady’s principles and practice
of radiation oncology. Lippincott Williams & Wilkins, Philadelphia, PA
60. Yavuz MN, Yavuz AA, Aydin F, Can G, Kavgaci H (2002) The efficacy of octreotide in the
therapy of acute radiation-induced diarrhea: a randomized controlled study. Int J Radiat Oncol
Biol Phys 54(1):195–202
61. Loperamide—Medscape Reference. http://reference.medscape.com/drug/imodium-k-pek-ii-
loperamide-342041
62. Heusinkveld RS, Manning MR, Aristizabal SA (1978) Control of radiation-induced diarrhea
with cholestyramine. Int J Radiat Oncol Biol Phys 4(7):687–690
63. Condon JR, Wolverson RL, South M, Brinkley D (1978) Radiation diarrhoea and cholestyr-
amine. Postgrad Med J 54(638):838–839
64. Mennie A, Dalley V (1973) Aspirin in radiation-induced diarrhoea. Lancet 301(7812):1131
65. Mennie A, Dalley V, Dinneen L, Collier H (1975) Treatment of radiation-induced gastrointes-
tinal distress with acetylsalicylate. Lancet 306(7942):942–943
Radiation Cystitis
16

Radiation cystitis is a complication of pelvic radiation therapy that patients encoun-


ter when treated for bladder cancer or other pelvic cancers.
There is a wide range of radiation cystitis incidence because of variation in radia-
tion technique, time-dose-volume parameters, the subjective nature of the symp-
toms, and diversity in collecting and reporting patient data between studies that
evaluated radiation cystitis [1].
Up to 70–90% of patients with bladder cancer experience acute genitourinary
symptoms during radiation therapy [2–19]. Prostate cancer is the second most com-
mon reason for radiation-induced cystitis (41–100%) [20–30], followed by cervical
cancer (20–25%) [31–35] and rectal cancer (35–53%) [36, 37].

16.1 Mechanism

The bladder wall has three well-defined layers including the mucosa (urothelium,
basement membrane, and lamina propria), the muscularis propria, and the adventi-
tia/serosa.
Human urothelium is considered a transitional epithelial tissue that its cellular
differentiation increased from basal cells at basement membrane to surface cells of
superficial or apical layer [38]. Surface cells known as umbrella cells are joined to
each other with tight cell membrane junctions. The apical surface of umbrella cells
is almost entirely covered by rigid plaques (known as the asymmetric unit mem-
brane or AUM) consisting of a group of transmembrane proteins called uroplakins
(UP), i.e., UPIa, UPIb, UPII, and UPIII [39–42].
Between these particles, UPIII is a main subunit of urothelial plaques that play a
key role in urothelial structure [43].
The urothelium also contains a luminal negatively charged glycosaminoglycan
(GAG) called a mucous layer that covers the umbrella cells on top of the apical
membrane [44].

© Springer International Publishing AG 2017 155


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_16
156 16 Radiation Cystitis

GAGs can be in a non-sulfated form like hyaluronic acid (HA) or a sulfated form
like heparan sulfate and heparin, chondroitin sulfate (CS), dermatan sulfate, and
keratan sulfate [45].
Tight junctions between umbrella cells, the urothelial plaques, and the glycos-
aminoglycan (GAG) layer make an efficient barrier against electrolyte and nonelec-
trolyte urine concentrations in bladder mucosa [46, 47].
Data on underlying mechanisms of acute radiation effects in the urinary bladder
are limited. The urothelium barrier of the bladder is affected by radiation. Decrease
in the number of umbrella cells [48], loss of the superficial UPIII layer [48], and
GAG layer damage [49, 50] have been reported in patients with radiation cystitis.
However, due to slow cell turnover rate of bladder epithelium (in the range of
42–350 days) [46, 51], gross tissue breakdown does not occur in early radiation
cystitis [48].
Following the urothelium barrier impairment, urine substances infiltrate the
bladder wall and produce more injuries by induction of the inflammatory response,
mast cell proliferation, activation, degranulation and histamine release, vasodilata-
tion, and also erythematous swelling [48]. Urine ion content, histamine, and other
inflammatory substances irritate nerve terminals within the bladder wall [52]. This
damaged barrier is also susceptible to secondary infection that could produce more
injury [53].
Molecular responses including increased synthesis of intercellular adhesion mol-
ecules (ICAM1) [54] and prostaglandin [55] resulted in increased inflammatory
reactions and detrusor muscle edema and decreased the bladder capacity. All these
events finally produce overactive bladder symptoms.

16.2 Timing

The first symptoms of early radiation effects on the urinary bladder usually occur
4–6 weeks after treatment begins. They are usually mild and transient. It has been
found that grade 2 or higher acute bladder toxicity may take an average of 6 weeks
(range 2–21 weeks) after the completion of treatment to improve [55, 56], but
5–20% of patients may develop persistent late bladder damage from 6 months to
10 years after treatment [57]. The risk of late bladder damage depends on pre-­
radiation therapy genitourinary morbidity [4], smoking history (≥1 pack per day)
[58], body mass index(>30 kg/m2) [58], any kind of procedure before radiation
therapy [59], and a high dose per fraction [60] that go beyond the scope of this book.

16.3 Risk Factors

The risk of acute bladder damage increases with pre-radiation therapy genitourinary
morbidity score [4, 61].
Chemotherapy administered concurrently with radiation therapy has not been
shown to significantly increase the risk of acute bladder complications [12, 34, 62],
16.5 Scoring 157

whereas use of hormonal therapy in prostate cancer has been associated with more
acute genitourinary complications [4, 61].
Radiation total dose, maximum bladder dose, and higher volume of bladder
exposed to radiation are associated with a higher risk of acute toxicity [29, 63, 64].
The treatment time and dose per fraction have not been reported to be related with
acute bladder injury [56, 65].
Other clinical factors such as age, transurethral resection of the prostate before
radiation therapy, diabetes mellitus, and smoking have not been reported to be asso-
ciated with acute bladder toxicity [4, 56], although some of them as previously
mentioned are related to late toxicity.

16.4 Symptoms and Diagnosis

Radiation cystitis symptoms during the early reaction phase include frequency, noc-
turia, urgency, dysuria, and hematuria [66].

Urgency: Sudden and compelling desire to pass urine, which is difficult to defer
Incontinence: Involuntary leakage of urine with the feeling of urgency
Frequency: Voiding more than eight times during the day
Nocturia: Need to wake up more than once at night to void

Bacterial infection may complicate the clinical picture.


Direct examination of the urothelium demonstrates mucosal edema, hyperemia,
and inflammation.

16.5 Scoring

Radiation Therapy Oncology Group (RTOG) radiation induced bladder morbidity


scoring criteria presented in Table 16.1 [67].

Table 16.1 RTOG bladder morbidity scoring


Grade 1 Frequency of urination or nocturia twice pretreatment habit/dysuria, urgency not
requiring medication
Grade 2 Frequency of urination or nocturia that is less frequent than every hour. Dysuria,
urgency, and bladder spasm requiring local anesthetic (e.g., Pyridium)
Grade 3 Frequency with urgency and nocturia hourly or more frequency/dysuria, pelvis pain
or bladder spasm requiring regular, frequent narcotic/gross hematuria with/without
clot passage
Grade 4 Hematuria requiring transfusion/acute bladder obstruction not secondary to clot
passage, ulceration, or necrosis
158 16 Radiation Cystitis

16.6 Prevention

Acute symptoms of radiation injury to the bladder are usually self-limited and man-
ageable [68], but prevention of these symptoms is important because they are asso-
ciated with a marked impact on quality of life and increased occurrence of late
toxicities [56].

16.6.1 Bladder Sparing

With the introduction of novel radiation therapy techniques like intensity-­modulated


radiation therapy (IMRT) or image-guided radiotherapy (IGRT), treatment confor-
mity and normal tissue sparing improve, which minimize the risk of treatment-­
related toxicity. A significant reduction in acute bladder toxicity in prostate, cervical,
and rectal cancer patients treated with this highly conformal treatment versus 3D
conformal radiotherapy (3DCRT) has been found [69–73].
Different dose-volume parameters have been proposed to predict acute genito-
urinary toxicities. For example, maximum dose (Dmax) of bladder in prostate can-
cer study [29] and volume of bladder receiving 35 Gy or more in rectal cancer [36]
have been reported to have significant correlations with the risk of acute bladder
toxicity.

16.6.2 Intravesical Instillation

Intravesical GAG instillations including hyaluronic acid [73] and chondroitin sul-
fate [74] in patients undergoing radiation therapy may reduce overactive bladder
symptoms due to covering of the bladder urothelium and preventing of cellular
damage by urinary irritants.
The proposed treatment scheme of chondroitin sulfate is weekly instillations of
40 mL of 0.2% solution of chondroitin sulfate during radiation therapy in outpatient
clinic after a course of radiation therapy (at least 2 days after chemotherapy). A
single-use catheter is usually used to empty the bladder and then fill it with the
40 mL solution. Patients are asked not to void at least 2 h after the instillation [74].
The proposed treatment scheme of intravesical hyaluronic acid prescription is
instillations of 40 mg/50 mL intravesical hyaluronic acid. Instillations should begin
1 week before starting radiation therapy and then weekly instillations until 4 weeks
after completion of radiation therapy [73].

16.7 Management

Generally, all patients with acute radiation cystitis symptoms should be encouraged
to increase fluid intake. It can result in urine irritant dilution and prevent urinary
tract infections [75].
16.7 Management 159

Acute radiation cystitis often responds to symptomatic therapy. Anticholinergics


have been the mainstay of management for storage symptoms such as frequency.
Other treatment options include analgesics like phenazopyridine and alpha-1
blocker [66].

16.7.1 Anticholinergics

Anticholinergic agents block acetylcholine effects at muscarinic receptors and sup-


press bladder muscle contractions [76]. These agents improve bladder storage and
alleviate symptoms.
Oxybutynin and tolterodine are widely used for overactive bladder symptoms. They
are nonspecific muscarinic receptor blocker and can cause side effects by acting on
other parts of the body (e.g., dry mouth or eyes, constipation, or nausea). Oxybutynin
and tolterodine appear to have similar effects on patients’ symptom improvement but
have a lower risk of withdrawals and dry mouth [76, 77]. Extended release preparations
of these drugs are available and are associated with similar efficacy and less risk of dry
mouth [76, 77]. Hyoscyamine is another anticholinergic agent that is used to treat cys-
titis. Data for comparing hyoscyamine to other anticholinergic drugs are lacking.

Oxybutynin chloride:
Immediate release: 5 mg PO two to three times daily; not to exceed 5 mg PO four
times daily [78]
Extended release: 5–10 mg PO daily; may be increased by 5 mg/day at weekly
intervals, not to exceed 30 mg/day [78]

Tolterodine:
Immediate release: 2 mg PO q12h [79]
Extended release: 2–4 mg PO once daily [79]

Oxybutynin chloride and tolterodine are contraindicated in patients with gastric


or urinary obstruction or retention, paralytic ileus, severe ulcerative colitis, and
uncontrolled narrow-angle glaucoma [78, 79].

16.7.2 Alpha-1 Blocker

Tamsulosin is an α-1A-specific blocker that induces selective relaxation of the


detrusor muscle and improved bladder compliance [80]. Tamsulosin may be the
preferred drug to prescribe over other apha-1 blockers like prazosin because of its
more acceptable side effect profile and greater patient satisfaction [81].

Usage: 0.4 mg PO once daily, 30 min after same meal each day [82];

it should be used with caution in patients with coronary artery disease [82].
160 16 Radiation Cystitis

16.7.3 Analgesics

Phenazopyridine is a urinary analgesic that is excreted into the urine and induces a
local analgesic effect.
Usage: 100–200 mg PO after meals three times daily [83].
Phenazopyridine is contraindicated in hypersensitivity to drug, severe hepatitis,
and renal impairment (CrCl <50 mL/min) [83].

16.7.4 Intravesical GAG

Damage to the GAG layer has been found in radiation cystitis mechanism.
Intravesical GAG could be bound to the damaged bladder surface and reduce radia-
tion cystitis symptoms. The glycosaminoglycan molecule is not absorbed into the
body and doesn’t penetrate the bladder and remain on the mucosal surface [84].
Extensive clinical experience has been gained with formulations containing
either chondroitin sulfate, hyaluronic acid, or a combination of chondroitin sulfate
and hyaluronic acid in late radiation cystitis [50]. More trials are needed to deter-
mine the benefit of these agents in acute radiation cystitis.

References
1. Marks LB, Carroll PR, Dugan TC, Anscher MS (1995) The response of the urinary bladder,
urethra, and ureter to radiation and chemotherapy. Int J Radiat Oncol Biol Phys
31(5):1257–1280
2. Turgeon G-A, Souhami L, Cury FL, Faria SL, Duclos M, Sturgeon J et al (2014)
Hypofractionated intensity modulated radiation therapy in combined modality treatment for
bladder preservation in elderly patients with invasive bladder cancer. Int J Radiat Oncol Biol
Phys 88(2):326–331
3. Jie S, Xia L, Fuquan Z, Ke H, Xiaorong H, Xin L et al (2009) Outcome of three-dimensional
conformal radiotherapy for 109 patients with bladder cancer. Chinese. J Radiat Oncol
18(2):115–119
4. Tester W, Porter A, Asbell S, Coughlin C, Heaney J, Krall J et al (1993) Combined modality
program with possible organ preservation for invasive bladder carcinoma: results of RTOG
protocol 85-12. Int J Radiat Oncol Biol Phys 25(5):783–790
5. Ploussard G, Daneshmand S, Efstathiou JA, Herr HW, James ND, Rödel CM et al (2014)
Critical analysis of bladder sparing with trimodal therapy in muscle-invasive bladder cancer: a
systematic review. Eur Urol 66(1):120–137
6. Arias F, Domı́nguez MA, Martı́nez E, Illarramendi JJ, Miquelez S, Pascual I et al (2000)
Chemoradiotherapy for muscle invading bladder carcinoma. Final report of a single institu-
tional organ-sparing program. Int J Radiat Oncol Biol Phys 47(2):373–378
7. Danesi DT, Arcangeli G, Cruciani E, Altavista P, Mecozzi A, Saracino B et al (2004)
Conservative treatment of invasive bladder carcinoma by transurethral resection, protracted
intravenous infusion chemotherapy, and hyperfractionated radiotherapy. Cancer
101(11):2540–2548
References 161

8. Chen W-C, Liaw C-C, Chuang C-K, Chen M-F, Chen C-S, Lin PY et al (2003) Concurrent
cisplatin, 5-fluorouracil, leucovorin, and radiotherapy for invasive bladder cancer. Int J Radiat
Oncol Biol Phys 56(3):726–733
9. Hussain MH, Glass TR, Forman J, Sakr W, Smith DC, Al-Sarraf M et al (2001) Combination
cisplatin, 5-fluorouracil and radiation therapy for locally advanced unresectable or medically
unfit bladder cancer cases: a Southwest Oncology Group Study. J Urol 165(1):56–61
10. Kragelj B, Zaletel-Kragelj L, Sedmak B, Čufer T, Červek J (2005) Phase II study of radioche-
motherapy with vinblastine in invasive bladder cancer. Radiother Oncol 75(1):44–47
11. Hussain S, Stocken D, Peake D, Glaholm J, Zarkar A, Wallace D et al (2004) Long-term results
of a phase II study of synchronous chemoradiotherapy in advanced muscle invasive bladder
cancer. Br J Cancer 90(11):2106–2111
12. James ND, Hussain SA, Hall E, Jenkins P, Tremlett J, Rawlings C et al (2012) Radiotherapy
with or without chemotherapy in muscle-invasive bladder cancer. N Engl J Med
366(16):1477–1488
13. Mitin T, Hunt D, Shipley WU, Kaufman DS, Uzzo R, Wu C-L et al (2013) Transurethral sur-
gery and twice-daily radiation plus paclitaxel-cisplatin or fluorouracil-cisplatin with selective
bladder preservation and adjuvant chemotherapy for patients with muscle invasive bladder
cancer (RTOG 0233): a randomised multicentre phase 2 trial. Lancet Oncol 14(9):863–872
14. Hagan MP, Winter KA, Kaufman DS, Wajsman Z, Zietman AL, Heney NM et al (2003) RTOG
97-06: initial report of a phase I–II trial of selective bladder conservation using TURBT, twice-­
daily accelerated irradiation sensitized with cisplatin, and adjuvant MCV combination chemo-
therapy. Int J Radiat Oncol Biol Phys 57(3):665–672
15. Kaufman DS, Winter KA, Shipley WU, Heney NM, Wallace HJ, Toonkel LM et al (2009)
Phase I-II RTOG study (99-06) of patients with muscle-invasive bladder cancer undergoing
transurethral surgery, paclitaxel, cisplatin, and twice-daily radiotherapy followed by selective
bladder preservation or radical cystectomy and adjuvant chemotherapy. Urology
73(4):833–837
16. Gogna NK, Matthews JH, Turner SL, Mameghan H, Duchesne GM, Spry N et al (2006)
Efficacy and tolerability of concurrent weekly low dose cisplatin during radiation treatment of
localised muscle invasive bladder transitional cell carcinoma: a report of two sequential Phase
II studies from the Trans Tasman Radiation Oncology Group. Radiother Oncol 81(1):9–17
17. Kaufman DS, Winter KA, Shipley WU, Heney NM, Chetner MP, Souhami L et al (2000) The
initial results in muscle-invading bladder cancer of RTOG 95-06: phase I/II trial of transurethral
surgery plus radiation therapy with concurrent cisplatin and 5-fluorouracil followed by selective
bladder preservation or cystectomy depending on the initial response. Oncologist 5(6):471–476
18. Rödel C, Grabenbauer GG, Kühn R, Papadopoulos T, Dunst J, Meyer M et al (2002) Combined-­
modality treatment and selective organ preservation in invasive bladder cancer: long-term
results. J Clin Oncol 20(14):3061–3071
19. Majewski W, Tarnawski R (2009) Acute and late toxicity in radical radiotherapy for bladder
cancer. Clin Oncol 21(8):598–609
20. Peeters ST, Heemsbergen WD, van Putten WL, Slot A, Tabak H, Mens JW et al (2005) Acute
and late complications after radiotherapy for prostate cancer: results of a multicenter random-
ized trial comparing 68 Gy to 78 Gy. Int J Radiat Oncol Biol Phys 61(4):1019–1034
21. Harsolia A, Vargas C, Yan D, Brabbins D, Lockman D, Liang J et al (2007) Predictors for
chronic urinary toxicity after the treatment of prostate cancer with adaptive three-dimensional
conformal radiotherapy: dose–volume analysis of a phase II dose-escalation study. Int J Radiat
Oncol Biol Phys 69(4):1100–1109
22. Deville C, Both S, Hwang W-T, Tochner Z, Vapiwala N (2010) Clinical toxicities and dosimet-
ric parameters after whole-pelvis versus prostate-only intensity-modulated radiation therapy
for prostate cancer. Int J Radiat Oncol Biol Phys 78(3):763–772
162 16 Radiation Cystitis

23. Matzinger O, Duclos F, Van den Bergh A, Carrie C, Villà S, Kitsios P et al (2009) Acute toxic-
ity of curative radiotherapy for intermediate-and high-risk localised prostate cancer in the
EORTC trial 22991. Eur J Cancer 45(16):2825–2834
24. Bastasch MD, Teh BS, Mai W-Y, McGary JE, Grant IIIWH, Butler EB (2006) Tolerance of
endorectal balloon in 396 patients treated with intensity-modulated radiation therapy (IMRT)
for prostate cancer. Am J Clin Oncol 29(1):8–11
25. Deville C, Both S, Bui V, Hwang W-T, Tan K-S, Schaer M et al (2012) Acute gastrointestinal
and genitourinary toxicity of image-guided intensity modulated radiation therapy for prostate
cancer using a daily water-filled endorectal balloon. Radiat Oncol 7(1):76
26. Zietman AL, DeSilvio ML, Slater JD, Rossi CJ, Miller DW, Adams JA et al (2005) Comparison
of conventional-dose vs high-dose conformal radiation therapy in clinically localized adeno-
carcinoma of the prostate: a randomized controlled trial. JAMA 294(10):1233–1239
27. Viani GA, Stefano EJ, Afonso SL (2009) Higher-than-conventional radiation doses in local-
ized prostate cancer treatment: a meta-analysis of randomized, controlled trials. Int J Radiat
Oncol Biol Phys 74(5):1405–1418
28. Dearnaley DP, Sydes MR, Langley RE, Graham JD, Huddart RA, Syndikus I et al (2007) The
early toxicity of escalated versus standard dose conformal radiotherapy with neo-adjuvant
androgen suppression for patients with localised prostate cancer: results from the MRC RT01
trial (ISRCTN47772397). Radiother Oncol 83(1):31–41
29. Vora SA, Wong WW, Schild SE, Ezzell GA, Halyard MY (2007) Analysis of biochemical
control and prognostic factors in patients treated with either low-dose three-dimensional con-
formal radiation therapy or high-dose intensity-modulated radiotherapy for localized prostate
cancer. Int J Radiat Oncol Biol Phys 68(4):1053–1058
30. Budäus L, Bolla M, Bossi A, Cozzarini C, Crook J, Widmark A et al (2012) Functional out-
comes and complications following radiation therapy for prostate cancer: a critical analysis of
the literature. Eur Urol 61(1):112–127
31. Mitchell PA, Waggoner S, Rotmensch J, Mundt AJ (1998) Cervical cancer in the elderly
treated with radiation therapy. Gynecol Oncol 71(2):291–298
32. Whitney CW, Sause W, Bundy BN, Malfetano JH, Hannigan EV, Fowler WC Jr et al (1999)
Randomized comparison of fluorouracil plus cisplatin versus hydroxyurea as an adjunct to
radiation therapy in stage IIB-IVA carcinoma of the cervix with negative para-aortic lymph
nodes: a Gynecologic Oncology Group and Southwest Oncology Group study. J Clin Oncol
17(5):1339–1348
33. Rose PG, Bundy BN, Watkins EB, Thigpen JT, Deppe G, Maiman MA et al (1999) Concurrent
cisplatin-based radiotherapy and chemotherapy for locally advanced cervical cancer. N Engl
J Med 340(15):1144–1153
34. Morris M, Eifel PJ, Lu J, Grigsby PW, Levenback C, Stevens RE et al (1999) Pelvic radiation
with concurrent chemotherapy compared with pelvic and para-aortic radiation for high-risk
cervical cancer. N Engl J Med 340(15):1137–1143
35. Eifel PJ (ed) (2006) Chemoradiotherapy in the treatment of cervical cancer. Semin Radiat
Oncol 16(3):177–185
36. Appelt AL, Bentzen SM, Jakobsen A, Vogelius IR (2015) Dose-response of acute urinary
toxicity of long-course preoperative chemoradiotherapy for rectal cancer. Acta Oncol
54(2):179–186
37. Minsky BD, Cohen AM, Kemeny N, Enker WE, Kelsen DP, Reichman B et al (1992)
Combined modality therapy of rectal cancer: decreased acute toxicity with the preoperative
approach. J Clin Oncol 10(8):1218–1224
38. Jost SP, Gosling JA, Dixon JS (1989) The morphology of normal human bladder urothelium.
J Anat 167:103–115
39. Kong X-T, Deng F-M, Hu P, Liang F-X, Zhou G, Auerbach AB et al (2004) Roles of uropla-
kins in plaque formation, umbrella cell enlargement, and urinary tract diseases. J Cell Biol
167(6):1195–1204
40. Zhou G, Liang F-X, Wang Z, Liao Y, Ghiso J, Luque-Garcia JL et al (2012) MAL facilitates
the incorporation of exocytic uroplakin-delivering vesicles into the apical membrane of uro-
thelial umbrella cells. Mol Biol Cell 23(7):1354–1366
References 163

41. Kachar B, Liang F, Lins U, Ding M, Wu X-R, Stoffler D et al (1999) Three-dimensional analy-
sis of the 16 nm urothelial plaque particle: luminal surface exposure, preferential head-to-head
interaction, and hinge formation. J Mol Biol 285(2):595–608
42. Kreft ME, Jezernik K, Kreft M, Romih R (2009) Apical plasma membrane traffic in superficial
cells of bladder urothelium. Ann N Y Acad Sci 1152(1):18–29
43. Hu P, Deng F-M, Liang F-X, Hu C-M, Auerbach AB, Shapiro E et al (2000) Ablation of uro-
plakin III gene results in small urothelial plaques, urothelial leakage, and vesicoureteral reflux.
J Cell Biol 151(5):961–972
44. Janssen D, Schalken J, ten Dam G, Heesakkers J (2010) 1036 the urothelial cell-line RT4
expresses a glycosaminoglycan (GAG) layer on its outer surface; an in vitro model for the
bladder GAG-layer. J Urol 183(4):e403
45. Damiano R, Cicione A (2011) The role of sodium hyaluronate and sodium chondroitin sul-
phate in the management of bladder disease. Ther Adv Urol 3(5):223–232
46. Stewart F (1986) Mechanism of bladder damage and repair after treatment with radiation and
cytostatic drugs. Br J Cancer Suppl 7:280–291
47. Chen I, Tong Y (2007) The urothelium: a new target in urology. JTUA 18(1):12–16
48. Jaal J, Dörr W (2006) Radiation-induced damage to mouse urothelial barrier. Radiother Oncol
80(2):250–256
49. Parsons CL, Zupkas P, Parsons JK (2001) Intravesical potassium sensitivity in patients with
interstitial cystitis and urethral syndrome. Urology 57(3):428–432
50. Madersbacher H, van Ophoven A, van Kerrebroeck PE (2013) GAG layer replenishment ther-
apy for chronic forms of cystitis with intravesical glycosaminoglycans—a review.
NeurourolUrodyn 32(1):9–18
51. Stewart FA, Denekamp J, Hirst D (1980) Proliferation kinetics of the mouse bladder after
irradiation. Cell Prolif 13(1):75–89
52. Cervigni M, Natale F, Nasta L, Padoa A, Voi RL, Porru D (2008) A combined intravesical
therapy with hyaluronic acid and chondroitin for refractory painful bladder syndrome/intersti-
tial cystitis. Int Urogynecol J 19(7):943–947
53. Cervigni MKP, Oliveira P, Tarricone R, Guzman S (2014) Glycosaminoglycan-replenishment
therapy: rationale for use and current evidence. EMJ Urol 1:41–47
54. Jaal J, Brüchner K, Hoinkis C, Dörr W (2004) Radiation-induced variations in urothelial
expression of intercellular adhesion molecule 1 (ICAM-1): association with changes in urinary
bladder function. Int J Radiat Biol 80(1):65–72
55. Jaal J, Dörr W (2006) Radiation induced inflammatory changes in the mouse bladder: the role
of cyclooxygenase-2. J Urol 175(4):1529–1533
56. Yoshimura R-i, Iwata M, Shibuya H, Sakai Y, Kihara K (2006) Acute and late genitourinary
toxicity of conformal radiotherapy for prostate cancer. Radiat Med 24(8):553–559
57. Stewart F, Lundbeck F, Oussoren Y, Luts A (1991) Acute and late radiation damage in mouse
bladder: a comparison of urination frequency and cystometry. Int J Radiat Oncol Biol Phys
21(5):1211–1219
58. Eifel PJ, Jhingran A, Bodurka DC, Levenback C, Thames H (2002) Correlation of smoking
history and other patient characteristics with major complications of pelvic radiation therapy
for cervical cancer. J Clin Oncol 20(17):3651–3657
59. Viswanathan AN, Yorke ED, Marks LB, Eifel PJ, Shipley WU (2010) Radiation dose–volume
effects of the urinary bladder. Int J Radiat Oncol Biol Phys 76(3):S116–S122
60. Lindholt J, Hansen P (1986) Prostatic carcinoma: complications of megavoltage radiation
therapy. Br J Urol 58(1):52–54
61. Peeters ST, Hoogeman MS, Heemsbergen WD, Slot A, Tabak H, Koper PC et al (2005)
Volume and hormonal effects for acute side effects of rectum and bladder during conformal
radiotherapy for prostate cancer. Int J Radiat Oncol Biol Phys 63(4):1142–1152
62. Kirwan JM, Symonds P, Green JA, Tierney J, Collingwood M, Williams CJ (2003) A system-
atic review of acute and late toxicity of concomitant chemoradiation for cervical cancer.
Radiother Oncol 68(3):217–226
63. Valicenti RK, Winter K, Cox JD, Sandler HM, Bosch W, Vijayakumar S et al (2003) RTOG
94-06: is the addition of neoadjuvant hormonal therapy to dose-escalated 3D conformal
164 16 Radiation Cystitis

radiation therapy for prostate cancer associated with treatment toxicity? Int J Radiat Oncol
Biol Phys 57(3):614–620
64. Dearnaley D, Hall E, Lawrence D, Huddart R, Eeles R, Nutting C et al (2005) Phase III pilot
study of dose escalation using conformal radiotherapy in prostate cancer: PSA control and side
effects. Br J Cancer 92(3):488–498
65. Horwich A, Dearnaley D, Huddart R, Graham J, Bessell E, Mason M et al (2005) A ran-
domised trial of accelerated radiotherapy for localised invasive bladder cancer. Radiother
Oncol 75(1):34–43
66. Giannitsas K, Athanasopoulos A (2015) Intravesical therapies for radiation cystitis. Current
Urology 8(4):169–174
67. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
68. Smit SG, Heyns CF (2010) Management of radiation cystitis. Nat Rev Urol 7(4):206–214
69. Arbea L, Ramos LI, Martínez-Monge R, Moreno M, Aristu J (2010) Intensity-modulated radi-
ation therapy (IMRT) vs. 3D conformal radiotherapy (3DCRT) in locally advanced rectal can-
cer (LARC): dosimetric comparison and clinical implications. Radiat Oncol 5(1):17
70. Michalski JM, Yan Y, Watkins-Bruner D, Bosch W, Winter K, Galvin JM et al (2013)
Preliminary toxicity analysis of 3DCRT versus IMRT on the high dose arm of the RTOG 0126
Prostate Cancer Trial. Int J Radiat Oncol Biol Phys. doi:10.1016/j.ijrobp.2013.07.041
71. Gill S, Thomas J, Fox C, Kron T, Rolfo A, Leahy M et al (2011) Acute toxicity in prostate
cancer patients treated with and without image-guided radiotherapy. Radiat Oncol 6(1):145
72. Tao Y, Lefkopoulos D, Ibrahima D, Bridier A, Polizzi Mdel P, Wibault P et al (2008)
Comparison of dose contribution to normal pelvic tissues among conventional, conformal and
intensity-modulated radiotherapy techniques in prostate cancer. Acta Oncol 47(3):442–450
73. Patino EG, Garrido NS, Caneda LC, Gomez EC, Pazos AV, Ferreira AV et al (2008) Protective
effect on the urinary bladder mucosa of intravesical hyaluronic acid in cervix cancer patients
treated with pelvic radiotherapy, weekly chemotherapy and high-dose-rate brachytherapy.
Brachytherapy 7(2):152–153
74. Hazewinkel MH, Stalpers LJ, Dijkgraaf MG, Roovers J-PW (2011) Prophylactic vesical instil-
lations with 0.2% chondroitin sulfate may reduce symptoms of acute radiation cystitis in
patients undergoing radiotherapy for gynecological malignancies. Int Urogynecol
J 22(6):725–730
75. Bosch PC, Bosch DC (2014) Treating interstitial cystitis/bladder pain syndrome as a chronic
disease. Rev Urol 16(2):83–87
76. Hesch K (ed) (2007) Agents for treatment of overactive bladder: a therapeutic class review.
Proc (Baylor Univ Med Cent) 20(3):307–314
77. Madhuvrata P, Cody JD, Ellis G, Herbison GP, Hay-Smith EJC (2012) Which anticholinergic
drug for overactive bladder symptoms in adults. Cochrane Libr. doi:10.1002/14651858.
CD005429.pub2
78. oxybutynin—Medscape Reference [Internet]. reference.medscape.com
79. tolterodine—Medscape Reference [Internet]. reference.medscape.com
80. Ruggieri MR, Braverman AS, Pontari MA (2005) Combined use of α-adrenergic and musca-
rinic antagonists for the treatment of voiding dysfunction. J Urol 174(5):1743–1748
81. Hajebrahimi S, Asrbadr YA, Azaripour A, Sadeghi-Bazargani H (2011) Effect of tamsulosin
versus prazosin on clinical and urodynamic parameters in women with voiding difficulty: a
randomized clinical trial. International journal of general medicine 4:35–39
82. tamsulosin—Medscape Reference [Internet]. reference.medscape.com
83. Azo Standard, Pyridium—Medscape Reference [Internet]. reference.medscape.com
84. Kyker KD, Coffman J, Hurst RE (2005) Exogenous glycosaminoglycans coat damaged blad-
der surfaces in experimentally damaged mouse bladder. BMC Urol 5(1):1
Radiation Proctitis
17

The rectum is the most affected organ during pelvic radiation therapy because of its
fixed position in the central part of pelvic cavity [1]. Acute radiation proctitis occurs
in more than 75% of the patients during pelvic radiation therapy [2]. Treatment
interruption due to severe acute radiation proctitis has been reported in 10–20% of
patients undergoing pelvic radiation therapy [3, 4].

17.1 Mechanism

Radiation-induced acute colorectal injury involves all mucosal compartments


including the epithelium, goblet cells, and lamina propria.
Mitotic arrest occurs in rapidly proliferating epithelial stem cells in mucosal
crypts and leads to cell depletion; shortening, narrowing, and loss of crypts; eroded
surface of epithelium; and mucosal break down. Inflammatory cell infiltration of the
epithelium by neutrophilic and eosinophilic cells occurs. Cryptitis due to neutrophil
infiltration through the crypt wall and crypt abscesses by eosinophil accumulation
develop. The number of goblet cells is reduced, and glandular atrophy occurs.
Significant inflammation of the lamina propria with vessel congestion occurs and
leads to hyperemia, edema, and ulceration [5–7].
Inflammatory mediators including eicosanoids are induced in the rectum as an
early response to direct radiation injury or secondary to mucosal damage and lead
to an increase in inflammatory cell infiltration and activation, capillary permeability,
and proctitis severity [8].

17.2 Risk Factors

Several patient-related factors have been reported to be associated with acute radia-
tion proctitis like patients with younger age (less than 60 years) [9], presence of
hemorrhoids, diabetes [10], and inflammatory bowel diseases [11] (see Chap. 15).

© Springer International Publishing AG 2017 165


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_17
166 17 Radiation Proctitis

Treatment-related factors including multiple radiation dosimetric factors (like


the percent volume of rectum receiving more than 30, 35, and 60 Gy, mean rectal
dose, the percent volume of anal canal receiving 15 Gy, the rectal length irradiated
to doses of 5 Gy, and more) [9, 10, 12] and concurrent chemotherapy are proposed
as factors related to probability of acute radiation proctitis [13].

17.3 Timing

Early symptoms can develop during the first or second week of radiation therapy
[14] and increase in frequency and severity throughout the radiation therapy course
[5]. Symptoms including anorectal dysfunction usually resolve within 2–3 months
after treatment completion [4].

17.4 Symptoms

Acute radiation proctitis manifests by increased frequency and urgency of defeca-


tion, rectal pain, tenesmus, mucous discharge, and less commonly bleeding and
fecal incontinence [3, 15]. Fecal urgency and incontinence are associated with alter-
ations in sphincter function and reduction in the minimum basal and mean squeeze
pressures in anorectal manometry [3]. Preexisting patient symptoms or medical
conditions may be worsened during radiation therapy. Preexisting hemorrhoids may
be inflamed and exacerbate patient symptoms.
Perianal skin reactions occur in patients with this area present in radiation portals
that presents with the skin changes that are discussed in Chap. 1.
Edematous mucosa with visible and friable vascular pattern is the most common
morphologic change that is seen endoscopically in its maximal score in the acute
phase of radiation proctitis [5]. Infrequently, spontaneous hemorrhage or visible
ulcers may be seen [14].

17.5 Scoring

Radiation Therapy Oncology Group (RTOG) and European Organization for


Research and Treatment of Cancer (EORTC) radiation toxicity grading classify all
symptoms related to the lower gastrointestinal tract in a single scoring category
(Table 17.1) [16].
Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 classi-
fies proctitis (either acute or chronic) into five grades (Table 17.2) [17]:
17.6 Prevention 167

Table 17.1 RTOG/EORTC radiation toxicity grading for lower gastrointestinal tract
Definition
Grade 1 Increased frequency or change in quality of bowel habits not requiring medication/
rectal discomfort not requiring analgesics
Grade 2 Diarrhea requiring parasympatholytic drugs (e.g., Lomotil)/mucous discharge not
necessitating sanitary pads/rectal or abdominal pain requiring analgesics
Grade 3 Diarrhea requiring parenteral support/severe mucous or bloody discharge
necessitating sanitary pads/abdominal distention (flat plate radiograph demonstrates
distended bowel loops)
Grade 4 Acute or subacute obstruction, fistula, or perforation; GI bleeding requiring
transfusion; abdominal pain or tenesmus requiring tube decompression or bowel
diversion

Table 17.2 CTCAE v4 proctitis grading


Definition
Grade 1 Rectal discomfort, intervention not indicated
Grade 2 Symptoms (e.g., rectal discomfort, passing blood or mucus); medical intervention
indicated; limiting instrumental ADL
Grade 3 Severe symptoms; fecal urgency or stool incontinence; limiting self-care and ADLs
Grade 4 Life-threatening consequences; urgent intervention indicated
Grade 5 Death

17.6 Prevention

Intensity-modulated radiation therapy (IMRT) can reduce the irradiated volume of


the rectum and improve acute radiation-induced proctitis compared to 3D confor-
mal radiation therapy (3DCRT). However, dose-volume parameters do not signifi-
cantly change in rectal cancer treated with IMRT because of large volume of rectum
included in target volume of treatment fields [18–25].
Amifostine can reduce acute radiation-induced proctitis in patients receiving pel-
vic radiation therapy and is recommended at a dose of ≥340 mg/m2 intravenously to
prevent radiation proctitis [26, 27]. Intrarectal and subcutaneous application of ami-
fostine also have been reported to be effective alternative routes to the intravenous
amifostine in protection against acute radiation-induced rectal damage [28–30].
Investigational methods including injecting hyaluronic acid [31, 32] and human
collagen [33] in the perirectal fat as well as insertion of an inflatable balloon appli-
cator into the rectum [33, 34] have been proposed in prostate cancer radiation ther-
apy to increase the distance between rectum and prostate and consequently decrease
the radiation dose of the anterior rectal wall and proctitis.
Sucralfate is a nonabsorbable basic aluminum salt of sucrose sulfate that has
been proposed in prevention of radiation-induced proctitis due to its cytoprotective
168 17 Radiation Proctitis

properties, angiogenesis and epithelial proliferation improvement, protection of


denuded mucosa, and binding bile acids; however, several studies did not find sub-
stantial benefit of oral or rectal sucralfate in the reduction of acute radiation proctitis
symptoms [35, 36].
Rectal misoprostol, a prostaglandin E1 analog, is another agent that is evaluated
as a radioprotection of acute proctitis and provides mixed results in related studies
[37, 38]. It has been shown that rectal hyaluronic acid (HA), a major mucopolysac-
charide, can reduce epithelial cell death and provide a positive effect on radiation
proctitis [39].
Aminosalicylate compounds including prodrugs (sulfasalazine and balsalazide)
or active (mesalazine) agents have anti-inflammatory properties due to their ability
in inhibition of inflammatory mediators and cells. Balsalazide converts to active
form by colonic bacteria after oral administration with minimal systemic absorption
and has more efficacy in patients with more distal colon disease due to a high con-
centration of active drug to the distal compared to other oral agents like mesalazine
[40]. It has been suggested that oral balsalazide is able to prevent or reduce symp-
toms of radiation proctitis in patients undergoing irradiation for prostate cancer
[41]. Rectal mesalazine did not show any prophylactic effect on radiation-induced
proctitis during radiation therapy for prostatic carcinoma [42].
Nonsystemic glucocorticosteroids, such as beclomethasone dipropionate (BDP),
have been suggested as efficient agents for the prevention of radiation-induced
proctitis due to their anti-inflammatory properties and safer pharmacokinetic profile
compared with systemic agents. Further studies are needed to confirm these primary
promising results [43].
Clinical use of prophylactic sucralfate, misoprostol, HA, balsalazide, mesala-
mine, or BDP for radiation proctitis protection cannot be recommended based on
current results.
Butyrate enemas have not shown any benefit given for the prevention of acute
radiation proctitis [44].
Fecal calprotectin and lactoferrin are two indicative markers of intestinal inflam-
mation that are released by migrated intestinal neutrophils [45, 46]. It has been
shown that the fecal levels of calprotectin and lactoferrin increase during pelvic
radiation therapy and are associated with acute radiation proctitis in non-colorectal
cancers [47–49]. Increased fecal calprotectin and lactoferrin levels in colorectal
cancer limit their use as a diagnostic marker in this setting [50, 51]. Another marker
proposed in this regard is human DNA concentration that is excreted in feces due to
epithelial cell damage [51]. These factors are promising tools that allow monitoring
mucosal damage and treatment modification with the aim of acute injury
prevention.

17.7 Management

Acute proctitis can be treated with dietary modification, topical agents, and/or ces-
sation of treatment in severe cases.
References 169

Fecal urgency or incontinence can be improved by dietary modification. Patients


should be encouraged to have adequate soluble fiber (e.g., fruits without skins, oat,
bran, barley) in their regulatory diet. Dietary fiber increases stool bulk and enhances
stool consistency and viscosity [52]. Treatment of concomitant diarrhea may help to
control fecal urgency or incontinence. All dietary modification and medications that
are discussed for treatment of diarrhea in radiation-induced enteritis (Chap. 15)
could be effective in improvement of these patients’ symptoms. Loperamide is par-
ticularly useful in this setting due its positive effect on anal sphincter pressure that
helps in stool continence [3, 53, 54].
Current data support using of butyrate as an effective medical treatment of acute
radiation proctitis. Butyrate enema (80 mL once per day or enemas of 40 mL twice
daily) can improve patient symptoms. Butyrate, a distinct form of short-chain fatty
acid, is the major energy source for colonocytes and provides a trophic effect on
colonic mucosa by enhancing epithelial cell proliferation and differentiation
[55–57].
Other medical agents such as rectal sucralfate and rectal steroid enema plus oral
sulfasalazine may be effective [58], but there is no recommendation for using of
them in treatment of radiation proctitis. Due to high incidence of radiation proctitis,
there is substantial need for further studies to evaluate several topical agents alone
or in combination with oral medications as well as in combination with different
topical agents with different actions in treatment of radiation proctitis.
Patients with perianal skin reaction should be informed to have good hygiene and
use a sitz bath several times daily. Topical agents could be used based on severity of
reaction (see Chap. 1).

References
1. Counter SF, Froese DP, Hart MJ (1999) Prospective evaluation of formalin therapy for radia-
tion proctitis. Am J Surg 177(5):396–398
2. Grodsky MB, Sidani SM (2015) Radiation proctopathy. Clin Colon Rectal Surg
28(02):103–111
3. Yeoh E, Russo A, Botten R, Fraser R, Roos D, Penniment M et al (1998) Acute effects of
therapeutic irradiation for prostatic carcinoma on anorectal function. Gut 43(1):123–127
4. Cotti G, Seid V, Araujo S, Kiss DR, Habr-Gama A (2003) Conservative therapies for hemor-
rhagic radiation proctitis: a review. Rev Hosp Clin Fac Med Sao Paulo 58(5):284–292
5. Hovdenak N, Fajardo LF, Hauer-Jensen M (2000) Acute radiation proctitis: a sequential clini-
copathologic study during pelvic radiotherapy. Int J Radiat Oncol Biol Phys
48(4):1111–1117
6. Haboubi N, Rowland P, Schofield P (1988) The light and electron microscopic features of
early and late phase radiation-induced proctitis. Am J Gastroenterol 83(10):1140–1144
7. Sedgwick D, Howard G, Ferguson A (1994) Pathogenesis of acute radiation injury to the rec-
tum. Int J Color Dis 9(1):23–30
8. Cole A, Slater K, Sokal M, Hawkey C (1993) In vivo rectal inflammatory mediator changes
with radiotherapy to the pelvis. Gut 34(9):1210–1214
9. Yang TJ, JH O, Son CH, Apte A, Deasy JO, Wu A et al (2013) Predictors of acute gastrointes-
tinal toxicity during pelvic chemoradiotherapy in patients with rectal cancer. Gastrointest
Cancer Res 6(5–6):129–136
170 17 Radiation Proctitis

10. Valdagni R, Rancati T, Fiorino C, Fellin G, Magli A, Baccolini M et al (2008) Development of


a set of nomograms to predict acute lower gastrointestinal toxicity for prostate cancer
3D-CRT. Int J Radiat Oncol Biol Phys 71(4):1065–1073
11. Fuccio L, Frazzoni L, Guido A (2015) Prevention of pelvic radiation disease. World
J Gastrointest Pharmacol Ther 6(1):1–9
12. Peeters ST, Hoogeman MS, Heemsbergen WD, Slot A, Tabak H, Koper PC et al (2005)
Volume and hormonal effects for acute side effects of rectum and bladder during conformal
radiotherapy for prostate cancer. Int J Radiat Oncol Biol Phys 63(4):1142–1152
13. Gérard J-P, Conroy T, Bonnetain F, Bouché O, Chapet O, Closon-Dejardin M-T et al (2006)
Preoperative radiotherapy with or without concurrent fluorouracil and leucovorin in T3-4 rec-
tal cancers: results of FFCD 9203. J Clin Oncol 24(28):4620–4625
14. Sharma B, Pandey D, Chauhan V, Gupta D, Mokta J, Thakur S (2005) Radiation proctitis.
J Indian Acad Clin Med 6:146–151
15. Sanda MG, Dunn RL, Michalski J, Sandler HM, Northouse L, Hembroff L et al (2008) Quality
of life and satisfaction with outcome among prostate-cancer survivors. N Engl J Med
358(12):1250–1261
16. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
17. NCI, NIH, DHHS (2009) Common terminology criteria for adverse events v4. 0. National
Cancer Institute, Bethesda, MD
18. Zelefsky MJ, Fuks Z, Happersett L, Lee HJ, Ling CC, Burman CM et al (2000) Clinical experi-
ence with intensity modulated radiation therapy (IMRT) in prostate cancer. Radiother Oncol
55(3):241–249
19. Lukovic J, Patil N, D’souza D, Millman B, Yaremko BP, Leung E et al (2016) Intensity-­
modulated radiation therapy versus 3D conformal radiotherapy for postoperative gynecologic
cancer: are they covering the same planning target volume? Cureus 8(1):e467
20. Ahamad A, D’Souza W, Salehpour M, Iyer R, Tucker SL, Jhingran A et al (2005) Intensity-­
modulated radiation therapy after hysterectomy: comparison with conventional treatment and
sensitivity of the normal-tissue–sparing effect to margin size. Int J Radiat Oncol Biol Phys
62(4):1117–1124
21. Yang B, Zhu L, Cheng H, Li Q, Zhang Y, Zhao Y (2012) Dosimetric comparison of intensity
modulated radiotherapy and three-dimensional conformal radiotherapy in patients with gyne-
cologic malignancies: a systematic review and meta-analysis. Radiat Oncol 7(1):1
22. Jabbour SK, Patel S, Herman JM, Wild A, Nagda SN, Altoos T et al (2012) Intensity-modulated
radiation therapy for rectal carcinoma can reduce treatment breaks and emergency department
visits. Int J Surg Oncol 2012:891067
23. Sharma NK, Li T, Chen DY, Pollack A, Horwitz EM, Buyyounouski MK (2011) Intensity-­
modulated radiotherapy reduces gastrointestinal toxicity in patients treated with androgen
deprivation therapy for prostate cancer. Int J Radiat Oncol Biol Phys 80(2):437–444
24. Arbea L, Ramos LI, Martínez-Monge R, Moreno M, Aristu J (2010) Intensity-modulated radi-
ation therapy (IMRT) vs. 3D conformal radiotherapy (3DCRT) in locally advanced rectal can-
cer (LARC): dosimetric comparison and clinical implications. Radiat Oncol 5(1):1
25. Bazan JG, Hara W, Hsu A, Kunz PA, Ford J, Fisher GA et al (2011) Intensity-modulated radia-
tion therapy versus conventional radiation therapy for squamous cell carcinoma of the anal
canal. Cancer 117(15):3342–3351
26. Athanassiou H, Antonadou D, Coliarakis N, Kouveli A, Synodinou M, Paraskevaidis M et al
(2003) Protective effect of amifostine during fractionated radiotherapy in patients with pelvic
carcinomas: results of a randomized trial. Int J Radiat Oncol Biol Phys 56(4):1154–1160
27. Peterson D, Boers-Doets C, Bensadoun R, Herrstedt J (2015) Management of oral and gastro-
intestinal mucosal injury: ESMO clinical practice guidelines for diagnosis, treatment, and
follow-up. Ann Oncol 26(Suppl 5):v139–v151
28. Katsanos KH, Briasoulis E, Tsekeris P, Batistatou A, Bai M, Tolis C et al (2010) Randomized
phase II exploratory study of prophylactic amifostine in cancer patients who receive radical
radiotherapy to the pelvis. J Exp Clin Cancer Res 29(1):1
References 171

29. Singh AK, Ménard C, Guion P, Simone NL, Smith S, Crouse NS et al (2006) Intrarectal ami-
fostine suspension may protect against acute proctitis during radiation therapy for prostate
cancer: a pilot study. Int J Radiat Oncol Biol Phys 65(4):1008–1013
30. Kouloulias VE, Kouvaris JR, Pissakas G, Mallas E, Antypas C, Kokakis JD et al (2005) Phase
II multicenter randomized study of amifostine for prevention of acute radiation rectal toxicity:
topical intrarectal versus subcutaneous application. Int J Radiat Oncol Biol Phys
62(2):486–493
31. Wilder RB, Barme GA, Gilbert RF, Holevas RE, Kobashi LI, Reed RR et al (2010) Cross-­
linked hyaluronan gel reduces the acute rectal toxicity of radiotherapy for prostate cancer. Int
J Radiat Oncol Biol Phys 77(3):824–830
32. Prada PJ, Fernández J, Martinez AA, De La Rua A, Gonzalez JM, Fernandez JM et al (2007)
Transperineal injection of hyaluronic acid in anterior perirectal fat to decrease rectal toxicity
from radiation delivered with intensity modulated brachytherapy or EBRT for prostate cancer
patients. Int J Radiat Oncol Biol Phys 69(1):95–102
33. Noyes WR, Hosford CC, Schultz SE (2012) Human collagen injections to reduce rectal dose
during radiotherapy. Int J Radiat Oncol Biol Phys 82(5):1918–1922
34. van Lin ENT, Hoffmann AL, van Kollenburg P, Leer JW, Visser AG (2005) Rectal wall sparing
effect of three different endorectal balloons in 3D conformal and IMRT prostate radiotherapy.
Int J Radiat Oncol Biol Phys 63(2):565–576
35. O’Brien PC, Franklin CI, Dear KB, Hamilton CC, Poulsen M, Joseph DJ et al (1997) A phase
III double-blind randomised study of rectal sucralfate suspension in the prevention of acute
radiation proctitis. Radiother Oncol 45(2):117–123
36. Kneebone A, Mameghan H, Bolin T, Berry M, Turner S, Kearsley J et al (2001) The effect of
oral sucralfate on the acute proctitis associated with prostate radiotherapy: a double-blind,
randomized trial. Int J Radiat Oncol Biol Phys 51(3):628–635
37. Khan AM, Birk JW, Anderson JC, Georgsson M, Park TL, Smith CJ et al (2000) A prospective
randomized placebo-controlled double-blinded pilot study of misoprostol rectal suppositories
in the prevention of acute and chronic radiation proctitis symptoms in prostate cancer patients.
Am J Gastroenterol 95(8):1961–1966
38. Hille A, Schmidberger H, Hermann RM, Christiansen H, Saile B, Pradier O et al (2005) A
phase III randomized, placebo-controlled, double-blind study of misoprostol rectal supposito-
ries to prevent acute radiation proctitis in patients with prostate cancer. Int J Radiat Oncol Biol
Phys 63(5):1488–1493
39. Stefanelli A, Pascale G, Rainieri E, Ursino S, Colella M, Zini G et al (2012) Can we decrease
the acute proctitis in prostate cancer patients using hyaluronic acid during radiation therapy: a
prospective historically controlled clinical study. Eur Rev Med Pharmacol Sci 16(5):639–645
40. Pruitt R, Hanson J, Safdi M, Wruble L, Hardi R, Johanson J et al (2002) Balsalazide is superior
to mesalamine in the time to improvement of signs and symptoms of acute mild-to-moderate
ulcerative colitis. Am J Gastroenterol 97(12):3078–3086
41. Jahraus CD, Bettenhausen D, Malik U, Sellitti M, Clair WHS (2005) Prevention of acute
radiation-induced proctosigmoiditis by balsalazide: a randomized, double-blind, placebo con-
trolled trial in prostate cancer patients. Int J Radiat Oncol Biol Phys 63(5):1483–1487
42. Freund U, Schölmerich J, Siems H, Kluge F, Schäfer H, Wannenmacher M (1987) Unwanted
side-effects in using mesalazine (5-aminosalicylic acid) during radiotherapy. Strahlenther
Onkol 163(10):678–680
43. Fuccio L, Guido A, Laterza L, Eusebi L, Busutti L, Bunkheila F et al (2011) Randomised clini-
cal trial: preventive treatment with topical rectal beclomethasone dipropionate reduces post-­
radiation risk of bleeding in patients irradiated for prostate cancer. Aliment Pharmacol Ther
34(6):628–637
44. Maggio A, Magli A, Rancati T, Fiorino C, Valvo F, Fellin G et al (2014) Daily sodium butyrate
enema for the prevention of radiation proctitis in prostate cancer patients undergoing radical
radiation therapy: results of a multicenter randomized placebo-controlled dose-finding phase 2
study. Int J Radiat Oncol Biol Phys 89(3):518–524
172 17 Radiation Proctitis

45. Kane SV, Sandborn WJ, Rufo PA, Zholudev A, Boone J, Lyerly D et al (2003) Fecal lactoferrin
is a sensitive and specific marker in identifying intestinal inflammation. Am J Gastroenterol
98(6):1309–1314
46. Erbayrak M, Turkay C, Eraslan E, Cetinkaya H, Kasapoglu B, Bektas M (2009) The role of
fecal calprotectin in investigating inflammatory bowel diseases. Clinics 64(5):421–425
47. Larsen A, Hovdenak N, Karlsdottir A, Wentzel-Larsen T, Dahl O, Fagerhol M (2004) Faecal
calprotectin and lactoferrin as markers of acute radiation proctitis: a pilot study of eight stool
markers. Scand J Gastroenterol 39(11):1113–1118
48. Hille A, Schmidt-Giese E, Hermann RM, Herrmann MK, Rave-Fränk M, Schirmer M et al
(2008) A prospective study of faecal calprotectin and lactoferrin in the monitoring of acute
radiation proctitis in prostate cancer treatment. Scand J Gastroenterol 43(1):52–58
49. Hille A, Rave-Fränk M, Christiansen H, Herrmann MK, Kertesz T, Hermann RM et al (2009)
Faecal calprotectin and lactoferrin values during irradiation of prostate cancer correlate with
chronic radiation proctitis: results of a prospective study. Scand J Gastroenterol
44(8):939–946
50. Hirata I, Hoshimoto M, Saito O, Kayazawa M, Nishikawa T, Murano M et al (2007) Usefulness
of fecal lactoferrin and hemoglobin in diagnosis of colorectal diseases. World J Gastroenterol
13(10):1569–1574
51. Varela E, Antolín M, Guarner F, Verges R, Giralt J, Malagelada JR (2009) Faecal DNA and
calprotectin as biomarkers of acute intestinal toxicity in patients undergoing pelvic radiother-
apy. Aliment Pharmacol Ther 30(2):175–185
52. Read M, Read N, Barber D, Duthie H (1982) Effects of loperamide on anal sphincter function
in patients complaining of chronic diarrhea with fecal incontinence and urgency. Dig Dis Sci
27(9):807–814
53. Halverson AL (2005) Nonoperative management of fecal incontinence. Clin Colon Rectal
Surg 18(1):17–21
54. Hallgren T, Fasth S, Delbro D, Nordgren S, Öresland T, Hulten L (1994) Loperamide improves
anal sphincter function and continence after restorative proctocolectomy. Dig Dis Sci
39(12):2612–2618
55. Weiner JP, Wong AT, Schwartz D, Martinez M, Aytaman A, Schreiber D (2016) Endoscopic
and non-endoscopic approaches for the management of radiation-induced rectal bleeding.
World J Gastroenterol 22(31):6972–6986
56. Hille PDA, Herrmann MK, Kertesz T, Christiansen H, Hermann RM, Pradier O et al (2008)
Sodium butyrate enemas in the treatment of acute radiation-induced proctitis in patients with
prostate cancer and the impact on late proctitis. Strahlenther Onkol 184(12):686–692
57. Vernia P, Fracasso P, Casale V, Villotti G, Marcheggiano A, Stigliano V et al (2000) Topical
butyrate for acute radiation proctitis: randomised, crossover trial. Lancet
356(9237):1232–1235
58. Kochhar R, Patel F, Sharma S, Ayyagari S, Aggarwal R, Goenka M et al (1991) Radiation-­
induced proctosigmoiditis. Dig Dis Sci 36(1):103–107
Fatigue
18

Radiotherapy-induced fatigue is a common early and long-lasting side effect of


radiation therapy, which, despite its negative effect on patient quality of life, is often
underestimated and undertreated in daily practice. Patients usually do not consider
fatigue as a treatment side effect. Indeed only in one-fourth of cases are any inter-
vention proposed to the patient, and only about 50% of patients discuss it with a
physician [1].
Hickok et al. reported on the radiation-induced fatigue incidence in 372 patients
with various types of cancer who had received radiation therapy without concurrent
chemotherapy. They found that of the 160 patients who did not have any fatigue at
the start of irradiation, 70% (112 patients) developed the symptoms during radiation
therapy, and approximately 84% of patients that reported fatigue at the initiation of
therapy also reported fatigue over the course of treatment [2].

18.1 Mechanism

Several conditions can induce fatigue in cancer patients including anemia, mood,
and sleep disorders; patient’s symptoms such as pain, nausea and vomiting, diarrhea
and electrolyte disturbances, cardiopulmonary, hepatic renal or endocrine dysfunc-
tion, infection, and poor nutritional status; and the side effects of drugs such as
opioid analgesics or anticonvulsants [3].
Some of these variables may be produced by the tumor itself or treatment with
radiation or chemotherapy.
It has been proposed that anemia induced by radiation can cause fatigue due to
decreased oxygen delivery to tissue and a negative energy balance [3–5]. Irradiation
of different parts of the body may induce fatigue by different mechanisms. Severe
diarrhea during pelvic radiation therapy [6], hypothyroidism as a consequence of
neck radiation therapy [7], psychological effect in women receiving radiation ther-
apy for early breast cancer [8], direct effects on normal brain parenchyma related to
cranial radiation therapy [9], or a decline in neuromuscular efficiency in prostate

© Springer International Publishing AG 2017 173


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_18
174 18 Fatigue

cancer radiation therapy [10] may be related to fatigue etiology in patients treated
with radiation therapy.
Available data suggest that activation of the pro-inflammatory cytokine network
and inflammatory response may be responsible for radiation-induced fatigue. In
particular, increased levels of the interleukin-6 (IL-6), IL-1 receptor antagonist
(IL-1ra), and C-reactive protein (CRP) are associated with a higher frequency and
severity of fatigue [11–15]. Excessive nuclear factor (NF)-κB pathway activation
has been seen in fatigued patients who have a key role in controlling expression of
pro-inflammatory genes [16, 17].
Pro-inflammatory cytokines can also be produced in the central nervous system
in response to radiation and generate fatigue [11, 12, 18].
Persistent posttreatment fatigue is associated with elevated markers of pro-­
inflammatory cytokine activity and alterations in the cellular immune system along
with subtle dysregulation in hypothalamic-pituitary-adrenal axis function [11,
19–21].
Some investigators have not validated these relationships, so further evaluation is
needed [22].
Other mechanism proposed for radiation-induced fatigue is mitochondrial dys-
function. A defect in mitochondrial oxidative phosphorylation is induced by radia-
tion and causes genetic instability and cellular damage. Mitochondrial synthesis of
ATP is disrupted and fatigue develops [23].

18.2 Timing

Fatigue may be developed or increased from baseline levels during radiation ther-
apy. Fatigue usually begins during the second or third week of radiation therapy.
Symptoms of fatigue become more severe over the course of treatment (cumulative
fatigue) [24]. More than three-fourths of fatigue occurs by the third to fifth weeks of
treatment. Fatigue is usually reduced gradually after treatment completion to the
pretreatment levels within 4–8 weeks following the completion of treatment [25]
but in some patients is protracted over many weeks (may last from 3–4 weeks to
2–3 months after treatment stops) [20, 26–31].
It has been observed that the radiation therapy-free weekends are associated with
a lesser fatigue [32].

18.3 Risk Factors

There are inconsistent data across studies evaluating fatigue-related factors, and
additional research is warranted to determinate the predictor factors of radiation-­
induced fatigue. We discuss some of these factors here.
One of the uniformly reported predictive factors for fatigue during radiation
therapy is higher than the baseline fatigue level. The presence of fatigue at the
18.4 Symptoms 175

initiation of irradiation is associated with more fatigue experienced by patients dur-


ing radiation therapy [33, 34].
Both fatigue and psychological disorders (like anxiety and mood disorders) are
prevalent in cancer patients and have overlapping symptoms that produce some
ambiguity for conclusion. There are conflicting data regarding to relationship
between fatigue severity and psychological distress [7, 22, 35, 36]. Some have
reported no significant relationship [37], and others have shown predictability of
mood disorders for fatigue [36].
Some researchers have found differences in fatigue levels by radiation therapy
sites. High frequency of radiation-induced fatigue was seen in breast cancer patients
receiving radiation therapy rather than other common cancers including lung or
prostate cancer [38–44], although some reported that patients with lung, gastroin-
testinal, and head and neck cancers experience more severe fatigue and that their
fatigue increases more intensely over the course of radiation than does that of
patients with breast cancer [45].
There are sparse data about the treatment parameter for radiation-induced fatigue.
Radiation field sizes seem to be positively associated with maximum radiation-­
induced fatigue [46, 47]. Total dose might also be expected to influence the severity
of fatigue although linear increase in fatigue with cumulative radiation dose over
time is not seen [48].
Correlation between immune serum markers and fatigued patients has been pro-
posed [49]. An analysis of different variables showed that higher baseline neutrophil
counts have more consistent relation with fatigue than circulating cytokines, coagu-
lation factors, peripheral blood indices, and biochemical factors [33]; however,
more studies are needed to assess the relation of different immune markers and
fatigue level.
Data about the role of chemotherapy as a predictor of radiation therapy-induced
fatigue is also inconclusive. Some studies have shown that pretreatment with che-
motherapy may increase fatigue severity, although others have reported that fatigue
severity scores are not associated with chemotherapy [50, 51].
Other clinical variables including disease stage, previous surgery, weight [51,
52], and demographic variables such as race, gender, age, marital status, personality
characteristics, and employment status [2, 3, 48, 53, 54] have not been consistently
reported to be related with fatigue induced during radiation therapy.

18.4 Symptoms

Cancer-related fatigue is defined as a sensation of tiredness or lack of energy that is


associated with functional limitations and impaired quality of life [34, 53, 55–60].
Unlike simple tiredness, it is more debilitating and not relieved by sleep or rest. It
interferes with activities of daily living like preparing food or cleaning the house
and leads to withdrawal from enjoyable activities like social activities with friends
and family and even discontinuation of cancer treatment [26, 61]. Objective
176 18 Fatigue

physical function (performance status) [62–64] and psychological status (mental/


emotional aspects) [65–67] are also affected by fatigue.
There is a diurnal variation in radiation-induced fatigue, with increasing levels in the
evening, which is consistent with the diurnal pattern of fatigue in the general population
[68–71]. The severity of both evening and morning fatigue increases during radiation
therapy and then decreases following the completion of radiation therapy [72].

18.5 Diagnosis and Scoring

Fatigue is a sensation with multidimensional components including sensory, cogni-


tive, affective, behavioral, and physiologic aspects.
Based on NCCN guidelines “Cancer-related fatigue is defined as a persistent,
subjective sense of physical, emotional and/or cognitive tiredness or exhaustion
related to cancer or cancer treatment that is not proportional to recent activity and
that significantly interferes with usual functioning” [73].
Numerous assessment measures (including unidimensional and multidimen-
sional scales) have been developed for screening and diagnosis of cancer-related
fatigue. Most of them are based on patient self-report; however, there is no consen-
sus about the best method for assessment.
Multidimensional measures examine several domains of sensory, cognitive,
affective, behavioral, and physiologic function that is affected by cancer-related
fatigue. Therefore, a multidimensional measurement may seem to be a more spe-
cific and refined diagnostic tool but may be too long to complete and not be suitable
for screening or scoring in practice [9, 74].
Unidimensional measures (single-item or multi-item) are often single-question
scales that are rapid and sensitive and can be applied efficiently for screening of the
occurrence and measuring the severity of cancer-related fatigue but may lose the
multiple dimensions of fatigue [3, 75]. Moreover, some argue that the extent of
overlap between fatigue and depression is reduced with using a single item than a
multi-item measure for fatigue [76].
Consideration of the measurement properties, strengths, and limitations of these
instruments, including reliability, validity, specificity, sensitivity, recall period,
respondent burden, translation in multiple languages, and the availability of normed
values to aid interpretation, should be used to guide decisions about the utility of a
measure for specific clinical or research purposes [3].
Specific diagnostic criteria have been proposed for defining cancer-related
fatigue as an independent entity in the International Classification of Diseases, 10th
revision (ICD-10) [77–79].
Some of the other multidimensional scales for cancer-related fatigue have been
proposed including the Multidimensional Fatigue Inventory [80, 81], the Functional
Assessment of Cancer Therapy-Fatigue Scale [82], the Piper Fatigue Scale [83], the
Fatigue Symptom Inventory [84, 85], the Lee Fatigue Scale [86, 87], the revised
Schwartz Cancer Fatigue Scale [88], and the Cancer Fatigue Scale [89].
The Lee Fatigue Scale has established internal consistency reliability in a variety
of populations including cancer patients [86, 87, 90, 91]. In this scale, fatigue
18.5 Diagnosis and Scoring 177

severity is measured using 13 items. Each item has a 0–10 numeric rating scale with
higher scores indicating higher levels of fatigue severity. Respondents are asked to
rate each item based on how they felt “right now,” within 30 min of awakening (i.e.,
morning fatigue), and before going to bed (i.e., evening fatigue) for two consecutive
days and nights. The fatigue scale score is calculated as the mean of the 13 fatigue
items. It has established that cutoff scores for clinically significant levels of fatigue
(i.e., ≥3.2 for morning fatigue, ≥5.6 for evening fatigue) [86, 91].
There are several unidimensional single-item scales such as the Symptom
Distress Scale [92], the Rotterdam Symptoms Checklist [93], the European
Organization for Research and Treatment of Cancer (EORTC) QLQ-C30 quality-­
of-­life measure [94], the Medical Outcomes Study 36-Item Short-Form Health
Survey (SF-36) [95], the MD Anderson Symptoms Inventory [96], the Zung Self-­
Rating Depression Scale [97], the Visual Analog Fatigue Scale [98], or multi-item
scales such as the Brief Fatigue Inventory [99].
Based on National Comprehensive Cancer Network (NCCN) guidelines, fatigue
should be assessed quantitatively on a 0–l0 scale (where 0 means no fatigue and 10
means the worst fatigue imaginable, how would you rate your fatigue at its worst
over the past 7 days?); A score of 0 indicates an absence of fatigue, a score of 1–3
indicates the presence of mild fatigue that does not require clinical intervention, and
scores of 4–6 and 7–10 indicate moderate and severe fatigue, respectively, which
require further evaluation and clinical intervention [73].
Proposed (1998 draft) ICD-10 criteria for cancer-related fatigue is shown in
Table 18.1.

Table 18.1 Proposed (1998 draft) ICD-10 criteria for cancer-related fatigue
Six (or more) of the following symptoms have been present every day or nearly every day
during the same 2-week period in the past month, and at least one of the symptoms is (A1)
significant fatigue (A1)
A1. Significant fatigue, diminished energy, or increased need to rest, disproportionate to any
recent change in activity level
A2. Complaints of generalized weakness or limb heaviness
A3. Diminished concentration or attention
A4. Decreased motivation or interest to engage in usual activities
A5. Insomnia or hypersomnia
A6. Experience of sleep as unrefreshing or nonrestorative
A7. Perceived need to struggle to overcome inactivity
A8. Marked emotional reactivity (e.g., sadness, frustration, or irritability) to feeling fatigued
A9. Difficulty completing daily tasks attributed to feeling fatigued
A10. Perceived problems with short-term memory
A11. Post-exertional malaise lasting several hours
B. The symptoms cause clinically significant distress or impairment in social, occupational, or
other important areas of functioning
C. There is evidence from the history, physical examination, or laboratory findings that the
symptoms are a consequence of cancer or cancer therapy
D. The symptoms are not primarily a consequence of comorbid psychiatric disorders such as
major depression, somatization disorder, somatoform disorder, or delirium
178 18 Fatigue

18.6 Management

All cancer patients should be screened by a certain fatigue scale at their initial and
follow-up visits.
Patients with significant fatigue scale should be further evaluated by history and
physical examination. Cutoff for significant fatigue varies based on different fatigue
scale tools (see Sect. 18.5).
Based on NCCN guidelines, primary patient evaluation includes:

History and physical examination (disease status and treatment, review of systems,
social support status/availability of caregivers, economic status, onset, pattern,
duration change over time) [73]
Assessment of: Treatable contributing factors (pain, anemia, nutritional deficits/
imbalance, sleep disturbance/poor sleep hygiene, decreased functional status,
emotional distress, drug induced sedation comorbidities (alcohol/substance
abuse, cardiac dysfunction, hot flashes, hypothyroidism, hypogonadism, adrenal
insufficiency, gastrointestinal dysfunction, hepatic dysfunction, infection, neuro-
logic dysfunction, pulmonary dysfunction, renal dysfunction) [73]

In context with medical assessment of treatable factors, laboratory tests may be


ordered as clinically indicated including complete blood count (CBC), electrolyte
profile (sodium, potassium, chloride, bicarbonate), chemistry panel (creatinine,
blood urea nitrogen, glucose, magnesium, calcium, phosphorus, bilirubin, serum
transaminases, alkaline phosphatase, lactate dehydrogenase, albumin, total protein),
transferrin, total iron-binding capacity, ferritin, iron levels, folic acid, B12 level, and
thyroid function tests [100].
After primary patient evaluation, if there are any treatable contributing factors, it
should be managed, and if fatigue persists with clinical management of these condi-
tions, additional therapeutic modalities may be required [101].
There are both pharmacologic interventions and integrative non-pharmacologic
behavioral interventions to alleviate fatigue symptoms that physicians can consider
in the setting of no specific causal factors. Patients should be managed by multidis-
ciplinary teams (oncologist, psychologist, physical therapist, exercise specialist,
oncologist nurse, and nutritionist) for the best treatment approach.

18.6.1 Non-pharmacologic Interventions

There are various non-pharmacologic interventions including exercises (e.g., home-­


based exercise, supervised exercise), education and counseling, sleep therapy, and
complementary therapy. These non-pharmacologic interventions alone or in combi-
nation with pharmacologic approaches may be implemented for the effective man-
agement of cancer-related fatigue [102].
18.6 Management 179

18.6.2 Exercise

Exercise can be helpful for individuals with cancer-related fatigue during and post-­
cancer therapy [103–108]. Exercise improves patient function during radiation ther-
apy, which is attributed to reduce radiation fatigue and the improvement of quality
of life [109–111]. The proposed mechanism of exercise in reducing cancer-related
fatigue is balancing in energy resources, attenuation of the progressive muscle wast-
ing, and disruptions in muscle metabolism that occur with cancer and prescribed
treatments [112].
Both home-based exercise and supervised exercise can improve fatigue. Home-­
based exercise program is a potentially effective, low-cost, and safe intervention
[108], and supervised exercise allows individualizing the exercise regimen to the
specific condition of the patient and type of cancer by professional therapists and
offers greater motivation. Therefore, supervised exercise might be more effective
than home-based exercise in improvement in physical and psychological function-
ing especially in patients that have cancer or treatment-related morbidity [110,
113–116].
Exercise contraindications (such as extensive lytic bone metastases, extreme
thrombocytopenia) should be regarded. Neutropenic patients must avoid environ-
ments where the risk of exposure to infectious agents is high (e.g., public swimming
pools) [117]).
Home-based physical activity interventions usually consist of at least 10–30 min
per day for at least 2–5 days per week [102].
Supervised exercise interventions consist of combined aerobic, resistance, and
stretching exercises [118].

18.6.3 Counseling and Education

Education and counseling about cancer-related fatigue, its adverse effects, and strat-
egies to deal with it by telephone or online support programs, comprehensive cop-
ing strategy programs or other programs, or organizations have been reported to
greatly benefit cancer-related fatigue management [119, 120].
Efforts to educate should be directed at patient’s educational level. Open com-
munication between patient, family, and caregiving team can facilitate discussions
about the experience of fatigue and its effects on daily life.

18.6.4 Optimize Sleep Quality

One approach of non-pharmacologic supportive interventions to optimize sleep


quality and also decrease cancer-related fatigue is cognitive behavioral therapy,
which is a form of psychotherapy that treats problems and boosts happiness by
modifying dysfunctional emotions, behaviors, and thoughts.
180 18 Fatigue

Cognitive behavioral therapy for insomnia (stimulus control instructions, sleep


restriction therapy, and sleep hygiene counseling) seems to improve cancer-related
fatigue [120–122].
Due to some inconsistent results, additional research is needed to explore the
impact of this intervention on patients with different degrees of insomnia and
fatigue.
The aim of stimulus control is to limit the amount of time that patients spend
awake in bed. The recommendations consist of going to bed at the same time each
night or when sleepy, waking up at the same time every morning, and leaving the
bed after 20 min if unable to fall sleep [102].
The aim of sleep restriction is to limit the amount of time that patients spend in
bed to the amount of actual total sleep time to increase sleep consolidation. The
protocol includes avoiding afternoon naps and limiting total time in bed [102].
The sleep hygiene principle consists of a variety of behaviors and environmental
factors; it can be implemented by educating patients to avoid heavy meal closest to
bed time, avoiding caffeine in the afternoon, and establishing an environment that
can promote sleep, such as preparing a dark, quiet, and comfortable bedroom [102].

18.6.5 Complementary Therapies

Energy conservation such as tai chi (energy arts from China) [122, 123], polarity
therapy (energy therapy) [124], pranayama (control of breath) [125], and yoga [126,
127] may be effective interventions for managing cancer-related fatigue. Further
confirmatory studies are warranted.
The general consideration of energy conservation should be provided to patients
like balancing activities with rest, pacing slowly and steadily, selecting the tasks
based on priority and eliminating unnecessary tasks, and avoiding poor body pos-
ture [128].
There are some positive results about use of back massage [100] and acupuncture
[129] in cancer-related fatigue. Further research is required to investigate their
mechanism and efficacy in this era.

18.6.6 Other Psychosocial Interventions

Cognitive behavior therapy has a clinically relevant effect in reducing fatigue and
functional impairments in cancer survivors [130]. Although a variety of cognitive
behavioral and psychosocial interventions can be beneficial, discovering what patients
benefit from what type of psychosocial intervention is an unresolved issue [131].
Whether all patients require formal cognitive behavioral therapy by a psycholo-
gist or psychiatrist in addition of general counseling is unclear [131].
18.6 Management 181

Group therapy (approximately six patients per group) may be effective in both
emotional and physical symptoms and enhance quality of life for cancer patients
undergoing radiation therapy [117].
The results suggest that relaxation training [132] and hypnosis [133] may
improve several psychological parameters such as fatigue in ambulatory patients
who are undergoing radiation therapy [132].
Mindfulness-based stress reduction may be a useful therapeutic intervention for
improving cancer-related fatigue, although the evidence is preliminary [134].

18.6.7 Nutrition Counseling

Patients should be encouraged to have adequate intake of fluid* (e.g., 8–12 cups of
fluid throughout the day) and adequate nutrition (e.g., high-protein diet) [128].
Caution should be taken in patients with comorbidities that affect fluid balance
(e.g., congestive heart failure).

18.6.8 Pharmacologic Intervention

Methylphenidate (Ritalin®) is a central nervous system (CNS) stimulant that is


structurally related to amphetamines with a short half-life and a rapid onset of
action.
The results suggest that methylphenidate could be considered in patients with
severe cancer-related fatigue [135–138].
Prescribing strategy: start methylphenidate at 5 mg in the morning and 5 mg at
noon, titrating as necessary. The maximal dose with the potential for benefit in
cancer-­related fatigue is 40 mg daily [117].
Contraindications: hypersensitivity, glaucoma, family history of Tourette’s syn-
drome or motor tics, marked anxiety, tension, agitation, taking MAOIs within
2 weeks, and risk of severe hypertensive reaction.
Modafinil, a non-amphetamine wake-promoting agent, is used for the treatment
of narcolepsy. As compared with other psychostimulants such as methylphenidate,
it has a relatively selective site of action in the brain, with resultant fewer adverse
effects and lower potential for abuse.
There is insufficient evidence to prescribe modafinil for patients with cancer-­
related fatigue outside of a clinical trial context [139, 140].
Steroids (e.g., dexamethasone [141], methylprednisolone [142], megestrol ace-
tate [143]) may be effective in cancer-related fatigue in patients with advanced can-
cer [144, 145]. The possibility of steroid-induced secondary fatigue in terminally ill
cancer patients should be taken into consideration [146].
Antidepressants (e.g., paroxetine) have failed to demonstrate any improvements
in fatigue [145, 147]. There are some positive results about bupropion sustained-­
release efficacy in cancer-related fatigue. Further studies would be necessary to
establish the efficacy of this intervention [147, 148].
182 18 Fatigue

Guarana (Paullinia cupana) is a plant native to the Amazon basin. Guarana was
shown to be effective for fatigue in breast cancer patients receiving systemic chemo-
therapy. Further studies are needed to confirm its efficacy and its use in radiation
therapy-related fatigue [149].
High doses of American ginseng (Panax quinquefolius) have been shown to be
an effective and safe natural supplement for helping manage the fatigue associated
with cancer treatment. While data seem promising, additional studies are needed to
confirm these findings before ginseng can be recommended as a treatment for
cancer-­related fatigue [150].
Multivitamins do not improve radiation-related fatigue [151].
IV vitamin C administration appears to reduce cancer-related fatigue. Additional
well-designed placebo-controlled studies investigating the effects of IV vitamin C
on cancer-related fatigue appear warranted [152].
Donepezil is an inhibitor of acetylcholinesterase used for Alzheimer’s, and
dementia was not significantly superior to placebo in the treatment of cancer-related
fatigue [153].

References
1. Jereczek-Fossa BA, Marsiglia HR, Orecchia R (2002) Radiotherapy-related fatigue. Crit Rev
Oncol Hematol 41(3):317–325
2. Hickok JT, Roscoe JA, Morrow GR, Mustian K, Okunieff P, Bole CW (2005) Frequency,
severity, clinical course, and correlates of fatigue in 372 patients during 5 weeks of radio-
therapy for cancer. Cancer 104(8):1772–1778
3. Mitchell A, Berger A (2013) Fatigue. In: Devita, Hellman, and Rosenberg’s cancer principles
& practice of oncology, 10th edn. Lippincott Williams & Wilkins, Philadelphia, PA
4. Morrow GR, Andrews PL, Hickok JT, Roscoe JA, Matteson S (2002) Fatigue associated with
cancer and its treatment. Support Care Cancer 10(5):389–398
5. Ahlberg K, Ekman T, Gaston-Johansson F (2004) Levels of fatigue compared to levels of
cytokines and hemoglobin during pelvic radiotherapy: a pilot study. Biol Res Nurs
5(3):203–210
6. Wang XS, Janjan NA, Guo H et al (2001) Fatigue during preoperative chemoradiation for
resectable rectal cancer. Cancer 92:1725–1732
7. Jereczek-Fossa BA, Santoro L, Alterio D et al (2007) Fatigue during head-and-neck radio-
therapy: prospective study on 117 consecutive patients. Int J Radiat Oncol Biol Phys
68:403–415
8. Dagnelie PC, Pijls-Johannesma MC, Lambin P et al (2007) Impact of fatigue on overall qual-
ity of life in lung and breast cancer patients selected for high-dose radiotherapy. Ann Oncol
18:940–944
9. Escalante C. Cancer-related fatigue: prevalence, screening and clinical assessment. www.
uptodate.com
10. Monga U, Kerrigan AJ, Thornby J, Monga TN (1999) Prospective study of fatigue in local-
ized prostate cancer patients undergoing radiotherapy. Radiat Oncol Investig 7(3):178–185
11. Nelson R (2009) Radiation therapy-induced fatigue linked to inflammation. Clin Cancer Res
15(17):OF1–7
12. Greenberg DB, Gray JL, Mannix CM, Eisenthal S, Carey M (1993) Treatment-related fatigue
and serum interleukin-1 levels in patients during external beam irradiation for prostate can-
cer. J Pain Symptom Manag 8(4):196–200
References 183

13. Bower JE, Ganz PA, Tao ML et al (2009) Inflammatory biomarkers and fatigue during radia-
tion therapy for breast and prostate cancer. Clin Cancer Res 15(17):5534–5540
14. Courtier N, Gambling T, Enright S, Barrett-Lee P, Abraham J, Mason MD (2013)
Psychological and immunological characteristics of fatigued women undergoing radiother-
apy for early-stage breast cancer. Support Care Cancer 21(1):173–181
15. Greenberg DB, Gray JL, Mannix CM, Eisenthal S, Carey M (1993) Treatment-related fatigue
and serum interleukin-1 levels in patients during external beam irradiation for prostate can-
cer. J Pain Symptom Manag 8:196–200
16. Bower JE, Ganz PA, Irwin MR, Arevalo JM, Cole SW (2011) Fatigue and gene expression in
human leukocytes: increased NF-kappa B and decreased glucocorticoid signaling in breast
cancer survivors with persistent fatigue. Brain Behav Immun 25(1):147–150
17. Brach MA, Hass R, Sherman ML, Gunji H, Weichselbaum R, Kufe D (1991) Ionizing radia-
tion induces expression binding activity of the nuclear factor kappa B. J Clin Invest
88:691–695
18. Konsman JP, Parnet P, Dantzer R (2002) Cytokine-induced sickness behaviour: mechanisms
and implications. Trends Neurosci 25:154–159
19. Bower JE, Ganz PA, Aziz N, Fahey JL (2002) Fatigue and proinflammatory cytokine activity
in breast cancer survivors. Psychosom Med 64:604–611
20. Bower JE, Ganz PA, Aziz N, Fahey JL, Cole SW (2003) T-cell homeostasis in breast cancer
survivors with persistent fatigue. J Natl Cancer Inst 95:1165–1168
21. Bower JE, Ganz PA, Dickerson SS, Petersen L, Aziz N, Fahey JL (2005) Diurnal cortisol
rhythm and fatigue in breast cancer survivors. Psychoneuroendocrinology 30:92–100
22. Geinitz H, Zimmermann FB, Stoll P et al (2001) Fatigue, serum cytokine levels, and blood
cell counts during radiotherapy of patients with breast cancer. Int J Radiat Oncol Biol Phys
51(3):691–698
23. Hsiao C, Daly B, Saligan L (2016) The Etiology and management of radiotherapy-induced
fatigue. Expert Rev Qual Life Cancer Care 1(4):323–328
24. Schwartz AL, Nail LM, Chen S et al (2000) Fatigue patterns observed in patients receiving
chemotherapy and radiotherapy. Cancer Investig 18:11–19
25. Dhruva A, Dodd M, Paul S, Cooper B, Lee K, West C et al (2010) Trajectories of fatigue in
patients with breast cancer before, during, and after radiation therapy. Cancer Nurs
33(3):201–212
26. Hofman M, Ryan J, Figueroa-Moseley C, Jean-Pierr P, Morrow G (2007) Cancer-related
fatigue: the scale of the problem. Oncologist 12(Suppl 1):4–10
27. Fatigue and cancer fatigue. Available from http://chemocare.com/chemotherapy/side-effects/
fatigue-and-cancer.aspx
28. Side effects of radiation therapy. Canadian Cancer Society. Available from http://www.can-
cer.ca/en/cancer-information/diagnosis-andtreatment/radiation-therapy/side-effects-of-radiation-
therapy/?region=on
29. Janaki MG, Kadam A, Mukesh S, Nirmala S, PonniA RBS, Rajeev AG (2010) Magnitude of
fatigue in cancer patients receiving radiotherapy and its short term effect on quality of life.
J Cancer Res Ther 6(1):22–26
30. Bower JE, Ganz PA, Desmond KA et al (2000) Fatigue in breast cancer survivors: occur-
rence, correlates, and impact on quality of life. J Clin Oncol 18:743–753
31. Bower JE, Ganz PA, Desmond KA et al (2006) Fatigue in long-term breast carcinoma survi-
vors: a longitudinal investigation. Cancer 106:751–758
32. Jereczek-Fossa BA, Marsiglia HR, Orecchia R (2002) Radiotherapy-related fatigue. Crit Rev
Oncol Hematol 41:317–325
33. Wratten C, Kilmurray J, Nash S et al (2004) Fatigue during breast radiotherapy and its rela-
tionship to biological factors. Int J Radiat Oncol Biol Phys 59:160–167
34. Smets EM, Visser MR, Willems-Groot AF, Garssen B, Schuster-Uitterhoeve AL, Haes JC
(1998) Fatigue and radiotherapy: (B) experience in patients 9 months following treatment. Br
J Cancer 78:907–912
184 18 Fatigue

35. Bower JE, Ganz PA, Desmond KA, Rowland JH, Meyerowitz BE, Belin TR (2000) Fatigue
in breast cancer survivors: occurrence, correlates, and impact on quality of life. J Clin Oncol
18:743–753
36. Irvine D, Vincent L, Graydon JE, Bubela N, Thompson L (1994) The prevalence and corre-
lates of fatigue in patients receiving treatment with chemotherapy and radiotherapy. A com-
parison with the fatigue experienced by healthy individuals. Cancer Nurs 17:367–378
37. Celik G, Tuna A, Samancioglu S, Korkma M (2016) The fatigue, anxiety and depression
levels of patients with breast cancer during radiotherapy. Int J Clin Exp Med
9(2):4053–4058
38. Hurny C, Bernhard J, Joss R et al (1993) “Fatigue and malaise” as a quality-of-life indicator
in small-cell lung cancer patients. The Swiss Group for Clinical Cancer Research (SAKK).
Support Care Cancer 1:316–320
39. Hickok JT, Morrow GR, McDonald S et al (1996) Frequency and correlates of fatigue in lung
cancer patients receiving radiation therapy: implications for management. J Pain Symptom
Manag 11:370–377
40. Monga U, Kerrigan AJ, Thornby J et al (1999) Prospective study of fatigue in localized pros-
tate cancer patients undergoing radiotherapy. Radiat Oncol Investig 7:178–185
41. Stone P, Hardy J, Huddart R et al (2000) Fatigue in patients with prostate cancer receiving
hormone therapy. Eur J Cancer 36:1134–1141
42. Forlenza MJ, Hall P, Lichtenstein P et al (2005) Epidemiology of cancer-related fatigue in the
Swedish Twin Registry. Cancer 104:2022–2031
43. Sadler IJ, Jacobsen PB (2001) Progress in understanding fatigue associated with breast can-
cer treatment. Cancer Investig 19:723–731
44. Noal S, Levy C, Hardouin A et al (2011) One-year longitudinal study of fatigue, cognitive
functions, and quality of life after adjuvant radiotherapy for breast cancer. Int J Radiat Oncol
Biol Phys 81(3):795–803
45. Hickok JT, Morrow GR, Roscoe JA et al (2005) Occurrence, severity, and longitudinal course
of twelve common symptoms in 1129 consecutive patients during radiotherapy for cancer.
J Pain Symptom Manag 30:433–442
46. Taunk NK, Haffty BG, Chen S, Khan AJ, Nelson C, Pierce D, Goyal S (2011) Comparison of
radiation-induced fatigue across 3 different radiotherapeutic methods for early stage breast
cancer. Cancer 117(18):4116–4124
47. Beard CJ, Propert KJ, Rieker PP et al (1997) Complications after treatment with external-­
beam irradiation in early-stage prostate cancer patients: a prospective multiinstitutional out-
comes study. J Clin Oncol 15:223–229
48. Greenberg DB, Sawicka J, Eisenthal S, Ross D (1992) Fatigue syndrome due to localized
radiation. J Pain Symptom Manag 7(1):38–45
49. Ahlberg K, Ekman T, Gaston-Johansson F (2004) Levels of fatigue compared to levels of
cytokines and hemoglobin during pelvic radiotherapy: a pilot study. Biol Res Nurs
5:203–210
50. Donovan KA, Jacobsen PB, Andrykowski MA et al (2004) Course of fatigue in women
receiving chemotherapy and/or radiotherapy for early stage breast cancer. J Pain Symptom
Manag 28(4):373–380
51. Lavdaniti M, Patiraki E, Dafni U, Katapodi M, Papathanasoglou E, Sotiropoulou A (2006)
Prospective assessment of fatigue and health status in Greek patients with breast cancer
undergoing adjuvant radiotherapy. Oncol Nurs Forum 33(3):603–610
52. Irvine DM, Vincent L, Graydon JE, Bubela N (1998) Fatigue in women with breast cancer
receiving radiation therapy. Cancer Nurs 21(2):127–135
53. Smets EM, Visser MR, Willems-Groot AF et al (1998) Fatigue and radiotherapy: (A) experi-
ence in patients undergoing treatment. Br J Cancer 78(7):899–906
54. Poirier P (2006) The relationship of sick leave benefits, employment patterns, and individual
characteristics to radiation therapy-related fatigue. Oncol Nurs Forum 33(3):593–601
55. Cella D (1998) Factors influencing quality of life in cancer patients: anemia and fatigue.
Semin Oncol 25:43–46
References 185

56. Wagner LI, Cella D (2004) Fatigue and cancer: causes, prevalence and treatment approaches.
Br J Cancer 91:822–828
57. Irvine D, Vincent L, Graydon JE, Bubela N, Thompson L (1994) The prevalence and corre-
lates of fatigue in patients receiving treatment with chemotherapy and radiotherapy. A com-
parison with the fatigue experienced by healthy individuals. Can Nurse 17:367–378
58. Curt GA (2001) Fatigue in cancer. BMJ 322:1560
59. Miaskowski C, Lee KA (1999) Pain, fatigue, and sleep disturbances in oncology outpatients
receiving radiation therapy for bone metastasis: a pilot study. J Pain Symptom Manag
17(5):320–332
60. Lovely MP, Miaskowski C, Dodd M (1999) Relationship between fatigue and quality of life
in patients with glioblastoma multiforme. Oncol Nurs Forum 26:921–925
61. Hwang SS, Chang VT, Cogswell J, Kasmis BS (2002) Clinical relevance of fatigue levels in
cancer patients at a Veterans Administration Medical Center. Cancer 94(9):2481
62. Ahlberg K, Ekman T, Gaston-Johansson F (2005) Fatigue, psychological distress, coping
resources, and functional status during radiotherapy for uterine cancer. Oncol Nurs Forum
32:633–640
63. Mallinson T, Cella D, Cashy J et al (2006) Giving meaning to measure: linking self-reported
fatigue and function to performance of everyday activities. J Pain Symptom Manag
31:229–241
64. Brown DJ, McMillan DC, Milroy R (2005) The correlation between fatigue, physical func-
tion, the systemic inflammatory response, and psychological distress in patients with
advanced lung cancer. Cancer 103:377–382
65. Curt GA, Breitbart W, Cella D, Groopman JE, Horning SJ, Itri LM et al (2000) Impact of
cancer-related fatigue on the lives of patients: new findings from the fatigue coalition.
Oncologist 5:353–360
66. Tchekmedyian NS, Kallich J, McDermott A et al (2003) The relationship between psycho-
logic distress and cancer-related fatigue. Cancer 98:198–203
67. Stone P, Richards M, A’Hern R et al (2000) A study to investigate the prevalence, severity and
correlates of fatigue among patients with cancer in comparison with a control group of vol-
unteers without cancer. Ann Oncol 11:561–567
68. Curran SL, Beacham AO, Andrykowski MA (2004) Ecological momentary assessment of
fatigue following breast cancer treatment. J Behav Med 27(5):425–444
69. Dimsdale JE, Ancoli-Israel S, Ayalon L, Elsmore TF, Gruen W (2007) Taking fatigue seri-
ously, II: variability in fatigue levels in cancer patients. Psychosomatics 48(3):247–252
70. Fletcher BA, Schumacher KL, Dodd M et al (2009) Trajectories of fatigue in family caregiv-
ers of patients undergoing radiation therapy for prostate cancer. Res Nurs Health
32:125–139
71. Miaskowski C, Lee KA (1999) Pain, fatigue, and sleep disturbances in oncology outpatients
receiving radiation therapy for bone metastasis: a pilot study. J Pain Symptom Manag
17(5):320–332
72. Miaskowski C, Paul SM, Cooper BA et al (2008) Trajectories of fatigue in men with prostate
cancer before, during, and after radiation therapy. J Pain Symptom Manag 35(6):632–643
73. National Comprehensive Cancer Network (NCCN) guidelines. www.nccn.org. Accessed
2015
74. Yeh ET, Lau SSC, Su WJ, Tsai DJ, Tu YY, Lai YL (2011) An examination of cancer-related
fatigue through proposed diagnostic criteria in a sample of cancer patients in Taiwan. BMC
Cancer 11:387
75. Van Belle S, Paridaens R, Evers G, Kerger J, Bron D, Foubert J, Ponnet G, Vander Steichel
D, Heremans C, Rosillon D (2005) Comparison of proposed diagnostic criteria with FACT-F
and VAS for cancer-related fatigue: proposal for use as a screening tool. Support Care Cancer
13(4):246–254
76. Jean-Pierre P, Figueroa-Moseley C, Kohli S, Fiscella K, Palesh O, Morrow G (2007)
Assessment of cancer-related fatigue: implications for clinical diagnosis and treatment.
Oncologist 12(1):11–21
186 18 Fatigue

77. Cella D, Peterman A, Passik S et al (1998) Progress toward guidelines for the management of
fatigue. Oncology (Williston Park) 12:369–377
78. Ovcharov VK (1997) [International classification of diseases (tenth revision)]. Problemy
sotsial’noi gigieny i istoriia meditsiny/NII sotsial’noi gigieny, ekonomiki i upravleniia zdra-
vookhraneniem im. NA Semashko RAMN; AO “Assotsiatsiia ‘Meditsinskaia literatura’.”
(4):23–27
79. Cella D, Davis K, Breitbart W, Curt G, Fatigue Coalition (2001) Cancer-related fatigue: prev-
alence of proposed diagnostic criteria in a United States sample of cancer survivors. J Clin
Oncol 19(14):3385–3391
80. Smets EMA, Garssen B, Bonke B et al (1995) The Multidimensional Fatigue Inventory
(MFI): psychometric qualities of an instrument to assess fatigue. J Psychosom Res
39:315–325
81. Smets EM, Garssen B, Cull A, de Haes JC (1996) Application of the multidimensional fatigue
inventory (MFI-20) in cancer patients receiving radiotherapy. Br J Cancer 73:241–245
82. Yellen SB, Cella DF, Webster K et al (1997) Measuring fatigue and other anemia-related
symptoms with the Functional Assessment of Cancer Therapy (FACT) measurement system.
J Pain Symptom Manag 13:63–74
83. Piper BF, Dibble SL, Dodd MJ et al (1998) The revised Piper Fatigue Scale: psychometric
evaluation in women with breast cancer. Oncol Nurs Forum 25:677–684
84. Hann DM, Jacobsen PB, Azzarello LM et al (1998) Measurement of fatigue in cancer
patients: development and validation of the Fatigue Symptom Inventory. Qual Life Res
7:301–310
85. Hann DM, Denniston MM, Baker F (2000) Measurement of fatigue in cancer patients: fur-
ther validation of the Fatigue Symptom Inventory. Qual Life Res 9:847–854
86. Lerdal A, Kottorp A, Gay C, Lee K (2013) Lee Fatigue and Energy Scales: exploring aspects
of validity in a sample of women with HIV using an application of a Rasch model. Psychiatry
Res 205(3):241–246
87. Lee KA, Hicks G, Nino-Murcia G (1991) Validity and reliability of a scale to assess fatigue.
Psychiatry Res 36:291–298
88. Schwartz A, Meek P (1999) Additional construct validity of the Schwartz Cancer Fatigue
Scale. J Nurs Meas 7:35–45
89. Okuyama T, Akechi T, Kugaya A et al (2000) Development and validation of the Cancer
Fatigue Scale: a brief, three-dimensional, self-rating scale for assessment of fatigue in cancer
patients. J Pain Symptom Manag 19:5–14
90. Lee KA, Portillo CJ, Miramontes H (1999) The fatigue experience for women with human
immunodeficiency virus. J Obstet Gynecol Neonatal Nurs 28(2):193–200
91. Lee KA, DeJoseph JF (1992) Sleep disturbances, vitality, and fatigue among a select group
of employed childbearing women. Birth 19(4):208–213
92. McCorkle R, Quint-Benoliel J (1983) Symptom distress, current concerns and mood distur-
bance after diagnosis of life-threatening disease. Soc Sci Med 17:431–438
93. de Haes JC, van Knippenborg FC, Neijt JP (1990) Measuring psychological and physical
distress in cancer patients: structure and application of the Rotterdam Symptom Checklist. Br
J Cancer 62:1034–1038
94. Aaronson NK, Ahmedzai S, Bergman B et al (1993) The European Organization for Research
and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clini-
cal trials in oncology. J Natl Cancer Inst 85:365–376
95. McHorney CA, Ware JE Jr, Raczek AE (1993) The MOS 36-Item Short-Form Health Survey
(SF-36): II. Psychometric and clinical tests of validity in measuring physical and mental
health constructs. Med Care 31:247–263
96. Cleeland CS, Mendoza TR, Wang XS et al (2000) Assessing symptom distress in cancer
patients: the M.D. Anderson Symptom Inventory. Cancer 89:1634–1646
97. Kirsh KL, Passik S, Holtsclaw E et al (2001) I get tired for no reason: a single item screening
for cancer-related fatigue. J Pain Symptom Manag 22:931–937
References 187

98. Glaus A (1993) Assessment of fatigue in cancer and non-cancer patients and in healthy indi-
viduals. Support Care Cancer 1:305–315
99. Mendoza TR, Wang XS, Cleeland CS et al (1999) The rapid assessment of fatigue severity in
cancer patients: use of the Brief Fatigue Inventory. Cancer 85:1186–1196
100. Symptom Management Guidelines: cancer—related fatigue and anemia. www.bccancer.
bc.ca
101. Mustian K, Morrow G, Carroll J, Figueroa-Moseley C, Jean-Pierre P, Williams G (2007)
Integrative nonpharmacologic behavioral interventions for the management of cancer-related
fatigue. Oncologist 12(1):52–67
102. Wanchai A, Armer JM, Stewart BR (2011) Nonpharmacologic supportive strategies to pro-
mote quality of life in patients experiencing cancer-related fatigue: a systematic review. Clin
J Oncol Nurs 15(2):203–214
103. Graydon JE, Bubela N, Irvine D, Vincent L (1995) Fatigue-reducing strategies used by
patients receiving treatment for cancer. Cancer Nurs 18:23–28
104. Stevinson C, Lawlor DA, Fox KR (2004) Exercise interventions for cancer patients: system-
atic review of controlled trials. Cancer Causes Control 15(10):1035–1056
105. Cramp F, Byron-Daniel J (2012) Exercise for the management of cancer-related fatigue in
adults. Cochrane Database Syst Rev 11:CD006145
106. Milne HM, Wallman KE, Gordon S, Courneya KS (2008) Effects of a combined aerobic and
resistance exercise program in breast cancer survivors: a randomized controlled trial. Breast
Cancer Res Treat 108(2):279–288
107. Heim ME, v d Malsburg ML, Niklas A (2007) Randomized controlled trial of a structured
training program in breast cancer patients with tumor-related chronic fatigue. Onkologie
30(8–9):429–434
108. Mock V, Pickett M, Ropka ME, Muscari Lin E, Stewart KJ, Rhodes VA, McDaniel R, Grimm
PM, Krumm S, McCorkle R (2001) Fatigue and quality of life outcomes of exercise during
cancer treatment. Cancer Pract 9(3):119–127
109. Mock V, Dow KH, Meares CJ et al (1997) Effects of exercise on fatigue, physical function-
ing, and emotional distress during radiation therapy for breast cancer. Oncol Nurs Forum
24:991–1000
110. Hwang JH, Chang HJ, Shim YH, Park WH, Park W, Huh SJ, Yang JH (2008) Effects of
supervised exercise therapy in patients receiving radiotherapy for breast cancer. Yonsei Med
J 49(3):443–450
111. Windsor PM, Nicol KF, Potter J (2004) A randomized, controlled trial of aerobic exercise for
treatment-related fatigue in men receiving radical external beam radiotherapy for localized
prostate carcinoma. Cancer 101:550–555
112. Ryan JL, Carroll JK, Ryan EP, Mustian KM, Fiscella K, Morrow GR (2007) Mechanisms of
cancer-related fatigue. Oncologist 12(Suppl 1):22–34
113. Mutrie N, Campbell AM, Whyte F, McConnachie A, Emslie C, Lee L et al (2007) Benefits of
supervised group exercise programme for women being treated for early stage breast cancer:
pragmatic randomised controlled trial. BMJ 334:517
114. Lin KY, Shun SC, Lai YH, Liang JT, Tsauo JY (2014) Comparison of the effects of a super-
vised exercise program and usual care in patients with colorectal cancer undergoing chemo-
therapy. Cancer Nurs 37(2):21–29
115. Sadeeka A-M (2009) A biobehavioral model for the study of exercise interventions in cancer-­
related fatigue. Biol Res Nurs 10(4):381–391
116. Brown JC, Huedo-Medina TB, Pescatello LS, Pescatello SM, Ferrer RA, Johnson BT (2011)
Efficacy of exercise interventions in modulating cancer-related fatigue among adult cancer
survivors: a meta-analysis. Cancer Epidemiol Biomark Prev 20(1):123–133
117. Escalante C, Hesketh PJ (2010) Cancer-related fatigue: treatment. UpToDate, Inc., Watham,
MA. Available from https://www.uptodate.com/contents/cancer-related-fatigue-treatment#H7
118. Meneses-Echávez JF, González-Jiménez E, Ramírez-Vélez R (2015) Effects of supervised
multimodal exercise interventions on cancer-related fatigue: systematic review and meta-­
analysis of randomized controlled trials. Biomed Res Int. doi:10.1155/2015/328636
188 18 Fatigue

119. Yates P, Aranda S, Hargraves M, Mirolo B, Clavarino A, McLachlan S, Skerman H (2005)


Randomized controlled trial of an educational intervention for managing fatigue in women
receiving adjuvant chemotherapy for early-stage breast cancer. J Clin Oncol
23(25):6027–6036
120. Dirksen SR, Epstein DR (2008) Efficacy of an insomnia intervention on fatigue, mood and
quality of life in breast cancer survivors. J Adv Nurs 61(6):664–675
121. Savard J, Simard S, Ivers H, Morin CM (2005) Randomized study on the efficacy of cognitive-­
behavioral therapy for insomnia secondary to breast cancer, part II: immunologic effects.
J Clin Oncol 23(25):6097–6106
122. Berger AM, Kuhn BR, Farr LA, Von Essen SG, Chamberlain J, Lynch JC, Agrawal S (2009)
One-year outcomes of a behavioral therapy intervention trial on sleep quality and cancer-­
related fatigue. J Clin Oncol 27(35):6033–6040
123. Larkey LK, Roe DJ, Weihs KL, Jahnke R, Lopez AM, Rogers CE, Oh B, Guillen-Rodriguez
J (2015) Randomized controlled trial of Qigong/Tai Chi Easy on cancer-related fatigue in
breast cancer survivors. Ann Behav Med 49(2):165–176
124. Mustian KM, Roscoe JA, Palesh OG, Sprod LK, Heckler CE, Peppone LJ, Usuki KY, Ling
MN, Brasacchio RA, Morrow GR (2011) Polarity Therapy for cancer-related fatigue in
patients with breast cancer receiving radiation therapy: a randomized controlled pilot study.
Integr Cancer Ther 10(1):27–37
125. Chakrabarty J, Vidyasagar M, Fernandes D, Joisa G, Varghese P, Mayya S (2015) Effectiveness
of pranayama on cancer-related fatigue in breast cancer patients undergoing radiation ther-
apy: a randomized controlled trial. Int J Yoga 8(1):47–53
126. Sprod LK, Fernandez ID, Janelsins MC, Peppone LJ, Atkins JN, Giguere J, Block R, Mustian
KM (2015) Effects of yoga on cancer-related fatigue and global side-effect burden in older
cancer survivors. J Geriatr Oncol 6(1):8–14
127. Wang G, Wang S, Jiang P, Zeng C (2014) Effect of Yoga on cancer related fatigue in breast
cancer patients with chemotherapy. Zhong Nan Da Xue Xue Bao Yi Xue Ban
39(10):1077–1082
128. Karagozoglu S, Kahve E (2013) Effects of back massage on chemotherapy-related fatigue
and anxiety: supportive care and therapeutic touch in cancer nursing. Appl Nurs Res
26(4):210–217
129. Posadzki P, Moon TW, Choi TY, Park TY, Lee MS, Ernst E (2013) Acupuncture for cancer-­
related fatigue: a systematic review of randomized clinical trials. Support Care Cancer
21(7):2067–2073
130. Gielissen M, Verhagen S, Witjes F, Bleijenberg G (2006) Effects of cognitive behavior ther-
apy in severely fatigued disease-free cancer patients compared with patients waiting for cog-
nitive behavior therapy: a randomized controlled trial. J Clin Oncol 24(30):4882–4887
131. Forester B, Kornfeld DS, Fleiss JL, Thompson S (1993) Group psychotherapy during radio-
therapy: effects on emotional and physical distress. Am J Psychiatry 150:1700–1706
132. Decker TW, Cline-Elsen J, Gallagher M (1992) Relaxation therapy as an adjunct in radiation
oncology. J Clin Psychol 48:388–393
133. Montgomery G, David D, Kangas M, Green S, Sucala M, Bovbjerg D et al (2014) Randomized
controlled trial of a cognitive-behavioral therapy plus hypnosis intervention to control fatigue
in patients undergoing radiotherapy for breast cancer. J Clin Oncol 32(6):557–563
134. Kabat-Zinn J (1982) An outpatient program in behavioral medicine for chronic pain patients
based on the practice of mindfulness meditation: theoretical considerations and preliminary
results. Gen Hosp Psychiatry 4:33–47
135. Lower E, Fleishman S, Cooper A et al (2005) A phase III, randomized placebo-controlled
trial of the safety and efficacy of d-MPH as new treatment of fatigue and “chemobrain” in
adult cancer patients. J Clin Oncol. ASCO Meeting Abstract 8000
136. Sarhill N, Walsh D, Nelson KA et al (2001) Methylphenidate for fatigue in advanced cancer:
a prospective open-label pilot study. Am J Hosp Palliat Care 18:187–192
137. Homsi J, Nelson KA, Sarhill N et al (2001) A phase II study of methylphenidate for depres-
sion in advanced cancer. Am J Hosp Palliat Care 18:403–407
References 189

138. Hanna A, Sledge G, Mayer ML et al (2006) A phase II study of methylphenidate for the treat-
ment of fatigue. Support Care Cancer 14(3):210–215
139. Jean-Pierre P, Morrow GR, Roscoe JA, Heckler C, Mohile S, Janelsins M, Peppone L,
Hemstad A, Esparaz BT, Hopkins JO (2010) A phase 3 randomized, placebo-controlled,
double-blind, clinical trial of the effect of modafinil on cancer-related fatigue among 631
patients receiving chemotherapy: a University of Rochester Cancer Center Community
Clinical Oncology Program Research base study. Cancer 116(14):3513–3520
140. Spathis A, Fife K, Blackhall F, Dutton S, Bahadori R, Wharton R et al (2010) Modafinil for
the treatment of fatigue in lung cancer: results of a placebo-controlled, double-blind, random-
ized trial. Cancer 116(14):3513–3520
141. Yennurajalingam S, Frisbee-Hume S, Delgado-Guay MO et al (2012) Dexamethasone (DM)
for cancer-related fatigue: a double-blinded, randomized, placebo-controlled tiral. J Clin
Oncol 30, (suppl; abstract 9002). Abstract available online at https://www.asco.org\ASCOv2\
Meetings\Abstracts?&vmview=abst_detail_view&confID=114&abstractID=99080.
Accessed 18 Jun 2012
142. Bruera E, Roca E, Cedaro L et al (1985) Action of oral methylprednisolone in terminal cancer
patients: a prospective randomized double-blind study. Cancer Treat Rep 69:751–754
143. Bruera E, Ernst S, Hagen N, Spachynski K, Belzile M, Hanson J, Summers N, Brown B,
Dulude H, Gallant G (1998) Effectiveness of megestrol acetate in patients with advanced
cancer: a randomized, double-blind, crossover study. Cancer Prev Control 2(2):74–78
144. Morrow GR, Hickok JT, Roscoe JA et al (2003) Differential effects of paroxetine on fatigue
and depression: a randomized, double-blind trial from the University of Rochester Cancer
Center Community Clinical Oncology Program. J Clin Oncol 21:4635–4641
145. Roscoe JA, Morrow GR, Hickok JT et al (2005) Effect of paroxetine hydrochloride (Paxil) on
fatigue and depression in breast cancer patients receiving chemotherapy. Breast Cancer Res
Treat 89:243–249
146. Matsuo N, Yomiya K (2014) Aggravation of fatigue by steroid therapy in terminally ill
patients with cancer. Am J Hosp Palliat Care 31(3):341–344
147. Cullum JL, Wojciechowski AE, Pelletier G et al (2004) Bupropion sustained release treat-
ment reduces fatigue in cancer patients. Can J Psychiatry 49:139–144
148. Moss EL, Simpson JS, Pelletier G et al (2006) An open-label study of the effects of bupro-
pion SR on fatigue, depression and quality of life of mixed-site cancer patients and their
partners. Psychooncology 15:259–267
149. de Oliveira Campos MP, Riechelmann R, Martins LC, Hassan BJ, Casa FB, Del Giglio A
(2011) Guarana (Paullinia cupana) improves fatigue in breast cancer patients undergoing
systemic chemotherapy. J Altern Complement Med 17(6):505–512
150. Barton DL, Liu H, Dakhil SR et al (2012) Phase III evaluation of American ginseng (panax
quinquefolius) to improve cancer-related fatigue: NCCTG trial N07C2. J Clin Oncol 30,
(suppl, abstract 9001). Abstract available online at https://www.asco.org\ASCOv2\Meetings
151. de Souza Fêde AB, Bensi CG, Trufelli DC et al (2007) Multivitamins do not improve radia-
tion therapy-related fatigue: results of a double-blind randomized crossover trial. Am J Clin
Oncol 30(4):432–436
152. Carr AC, Vissers MC, Cook JS (2014) The effect of intravenous vitamin C on cancer- and
chemotherapy-related fatigue and quality of life. Front Oncol 4:283
153. Bruera E, El Osta B, Valero V, Driver LC, Pei BL, Shen L, Poulter VA, Palmer JL (2007)
Donepezil for cancer fatigue: a double-blind, randomized, placebo-controlled trial. J Clin
Oncol 25(23):3475–3481
Hematological Side Effects
19

Profound hematologic toxicity, prominently leukopenia, frequently develops fol-


lowing radiation therapy that includes a large volume of bone marrow in the radia-
tion field [1–8]. The percentage of patients with radiation-induced leukopenia has
been reported in up to 50% and 90% of patients treated with pelvic field irradiation
alone and with concurrent chemotherapy, respectively [1, 6]. These values are lower
for thrombocytopenia (1% and 30% of patients treated with pelvic field irradiation
alone and with concurrent chemotherapy, respectively) and anemia (30% and 50%
of patients treated with pelvic field irradiation alone and with concurrent chemo-
therapy, respectively) [1, 10].

19.1 Mechanism

The bone marrow is a cellular stroma consisting of the vascular system with nutri-
tive vessels and a very complex sinusoidal system [11]. At birth, virtually all mar-
row is hematopoietic. Conversion of red marrow to yellow marrow with advancing
age begins in the distal bones, and the distribution of active red marrow in the adult
is in the skull (13.1%), clavicle (1.5%), scapula (4.8%), sternum (2.3%), ribs (7.9%),
vertebrae (28.4%), pelvis (36.2%), and proximal extremities (5.7%). There is a
gradual reduction in the percentage of cellularity of red marrow with age and its
replacement by fatty yellow marrow [12].
The cellular stroma of bone marrow houses active blood-cell-forming stem cells
[13]. The peripheral blood also contains hematopoietic stem and progenitor cells.
Due to the dynamic equilibrium between the hemopoietic cell populations in the
bone marrow and peripheral blood, the peripheral blood cell populations could be
sensitive indicators of radiation-induced damage to hematopoietic tissues [14].
The hematopoietic stem and progenitor cells are highly sensitive to radiation
[15]; however, nondividing mature cells (e.g., granulocytes, red cells, or platelets)
tolerate higher radiation doses. After exposures at doses as low as 4 Gy, the cellular-
ity of the irradiated bone marrow progressively declines [16–18]. It has been found

© Springer International Publishing AG 2017 191


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_19
192 19 Hematological Side Effects

that an exposure dose of 30–47 Gy delivered in a period of 3–4 weeks to the sternal
area results in severe depression of the sternal marrow [19].
Breakdown of the sinusoidal system results in erythrocytes spreading throughout
the parenchyma, and a hemorrhagic bone marrow develops with radiation [16].
The most sensitive indication of acute radiation effect on peripheral blood counts
is a reduction in number of circulating lymphocytes, together with the appearance
of abnormal lymphocyte figures [17]. If the limited volume of marrow receives
radiation (less than 3% of the total active adult marrow), the peripheral blood counts
may be within normal limits despite marrow hypoplasia [17].
It has been demonstrated that irradiated bone marrow sites can be recovered
early by the stem cell migration of the protected bone marrow sites or peripheral
blood [13, 20, 21]. Proliferation and release of stem cells increase in non-irradiated
bone marrow after radiation therapy. The depletion of stem cells in the protected
bone marrow at the beginning of radiotherapy seems to be due to such migration
[16, 22]. Another reason for this diminution could be the result of an increased rate
of differentiation.
After stem cell transplantation, the times to “rebuild” for the granulocytic, erythro-
poietic, and megakaryocytic cell lines are 10–12, 5–7, and 10 days, respectively [11].
Although blood count recovery occurs a few months after completion of therapy
in 90% of patients, bone marrow regeneration and recovery require a much longer
time. The potential of regeneration appears to depend on the dose received. It has
been shown that a dosage of 30 Gy or more of bone marrow radiation needs an
extended time to recover and may permanently depress the marrow [19, 23]. This
could be due to permanent damage to the precursor cells of the marrow elements or
to a physiological alteration of the blood vessels and supporting syncytium. These
factors could be responsible for the prevention of growth of the regenerating cells
and/or the repopulation of cells from marrow in other parts of the body [17].
Permanent loss of hematopoietic activity in the irradiated areas is compensated by
increased activity in the non-irradiated areas [24, 25].

19.2 Timing

In patients undergoing conventional fractionated radiation therapy, occurrence,


degree, and timing of blood count suppression are dependent on the proportion of
active marrow volume in the radiation fields. The field size is the most important
determining factor for the adverse hematologic effects on severity and timing. For
example, it is estimated that the proportion of active marrow included in the periaor-
tic, mantle, and pelvic fields is about 10%, 25%, and 40%, respectively, and nadir
levels of the cell counts occur at different times for these fields [26].
The number of peripheral blood cells decreases according to their radiation sen-
sitivity and life expectancy. The total white cell count declines during radiation
therapy. Lymphocytes are the most sensitive, and monocytes are the most refractory
leukocytes to change. The lymphocyte count drops first and by the greatest amount,
19.4 Symptoms 193

the neutrophils less, and the monocytes only exhibit a transient, early, modest dip.
After the nadir, counts for white cells levels maintain a plateau [27].
The platelet count declines gradually with radiation therapy to a nadir and there-
after is maintained its level [27].
The hemoglobin very gradually decreases during radiation therapy and reaches
its lowest value at the end of radiation therapy, which could be related to the long
red blood cell life expectancy compared to leukocytes and platelets [27].
Complete peripheral blood count recovery is observed at 1–3 months following
therapy, although it may still be below the pre-therapy levels, and lymphocyte count
recovers fastest [27]. Peripheral blood counts are an unreliable indicator for assess-
ing the true status of bone marrow reserve, and bone marrow suppression may sus-
tain for months to years despite normalized peripheral blood counts [28, 29].
Patients may have normal peripheral blood counts with a marked suppression of
bone marrow secondary to increasing proliferation of unexposed bone marrow [28].

19.3 Risk Factors

Radiation-related factors such as larger radiation field and higher radiation dose are
the most important factors affecting bone marrow injury and degree of depression
in the peripheral blood counts [19, 30].
Chemotherapy is another factor affecting hematologic toxicity of radiation ther-
apy. Patients with previous and concomitant chemotherapy have increased risk for
radiation-induced neutropenia [31].

19.4 Symptoms

Depression of each hematopoietic cell lineage translates into potential problems for
the patient. The Radiation Therapy Oncology Group has defined a scoring system
for acute hematologic toxicity of radiotherapy (Table 19.1) [9].

Table 19.1 RTOG acute radiation hematologic effects scoring


Grade 1 Grade 2 Grade 3 Grade 4
White blood cells/ml 3000 to <4000 2000 to 1000 to <2000 <1000
<3000
Platelet/ml 75,000 to 50,000 to 25,000 to <50,000 <25,000 or
<100,000 <75,000 spontaneous
bleeding
Neutrophils/ml 1500 to <1900 1000 to 500 to <1000 <500 or sepsis
<1500
Hgb (g/dl)/Hct(%) 9.5 to <11/28 7.5 to <9.5/ 5 to <7.5 (pack cell –
to <32 <28 transfusion
required)
194 19 Hematological Side Effects

Patients with isolated neutropenia may not have any specific symptoms.
Neutropenia may place the patient at substantial risk for infection, and a fever may
be the first sign.
Thrombocytopenia is generally not associated with any symptoms and is self-­
limited, but manifestations of bruising, petechiae, and mucosal bleeding infre-
quently occur. Intracranial hemorrhage is the most dangerous manifestation of
thrombocytopenia, though it is particularly rare, and most episodes occur at platelet
counts less than 5000/mL.
Symptoms related to significant anemia are easy fatigue, low compliance for
activity, rapid heart rate, and shortness of breath.

19.5 Prevention

There is correlation between radiation technique parameters regarding bone marrow


and hematologic toxicity. These parameters could be modifiable with intensity-­
modulated radiation therapy (IMRT) and more advanced techniques. New tech-
niques can reduce radiation dose to bone marrow and decrease acute hematologic
toxicity [32–35].
One problem with standard IMRT planning is the large volume of bone marrow
that needs to be avoided. By considering only red (active) marrow in marrow con-
touring, the planning process could be improved [36]. The distribution of marrow
containing red (active) and yellow (inactive) marrow is different among individuals
[37]. Active red marrow can be visualized poorly in computed tomography (CT)
scanning. Functional bone marrow imaging like 2-deoxy-2-[F-18]fluoro-D-glucose
(FDG)-positron emission tomography (FDG-PET) [38] or single-photon emission
computed tomography (SPECT) [18] can provide information about functional nor-
mal bone marrow and its physiologic distribution. Incorporation of these modalities
into the IMRT planning process avoids the unnecessary contouring of inactive yel-
low marrow for calculation of dose constraints. Structural imaging techniques such
as magnetic resonance imaging (MRI) in place of functional imaging may be useful
in this regard [18, 37, 38].
The potential for cytokines in prevention of the hematopoietic system radiation
injury has been studied in lethally irradiated animals, and observations suggest that
some cytokines such as G-CSF [39, 40], interleukin-1 (IL-1) [41, 42], tumor necro-
sis factor (TNF)-alpha [43], and Fms-related tyrosine kinase 3 ligand (FLT3LG)
[44–46] may mediate radioprotective effects to the hematopoietic progenitor cell
and stem cell compartments. It has also been shown that meloxicam, a selective
inhibitor of cyclooxygenase 2, can modulate hematopoiesis and elevate numbers of
granulocytic precursor cells in bone marrow and granulocyte counts in peripheral
blood by increasing endogenous G-CSF production [47]. Currently, there is no rec-
ommendation for the prophylactic use of these factors.
19.6 Treatment 195

19.6 Treatment

19.6.1 Neutropenia

The proliferation and differentiation of hematopoietic cells are regulated by a fam-


ily of cytokines including colony-stimulating factors (CSFs) and interleukins, which
are specific to each cell lineage [48]. Granulocyte colony-stimulating factor (G-CSF)
controls the production, differentiation, and function of neutrophilic granulocytes.
Single classes of high-affinity receptor for G-CSF are present on the precursor and
mature cells of neutrophilic granulocytes [48].
The mature human G-CSF is a 19.6 kDa glycoprotein containing 174 amino
acids [49], and its molecular cloning has been well described [50].
It is well known that E. coli is a useful host for the production of recombinant
human G-CSF [51], but there are also various products that are derived from differ-
ent tissues (e.g., yeast, transgenic plants, and mammalian cells) [52].
There are lots of controversies about using of colony-stimulating factors during
radiation therapy regarding acute and late side effects.
G-CSF treatment is well tolerated during continuous fractionated radiation ther-
apy and can be used clinically to alleviate severe and threatening neutropenia caused
by radiation therapy or by combined radio-chemotherapy [53, 54].
The number of G-CSF receptors on bone marrow cells increases with radiation
[55], and administration of recombinant human G-CSF can accelerate the recovery
of radiation-induced neutropenia [56–58] and reduce its severity and duration
[59–62].
After administration of recombinant human G-CSF, the level of circulating neu-
trophils increases by accelerating production, inhibition of neutrophil apoptosis,
reduction of transit time from stem cell to mature neutrophil, and accelerating neu-
trophil entry into the blood, and neutrophil function is enhanced. An increase in
neutrophil progenitors in the marrow and hematopoietic stem cell mobilization can
be seen in G-CSF administration [63–65].
Clinical application of recombinant human G-CSF is hampered in some situa-
tions by various side effects. G-CSFs should not be included in patients receiving
concomitant chemotherapy and radiation therapy, particularly involving the medias-
tinum based on the results of GM-CSF or G-CSF morbidity in mediastinal chemo-
radiotherapy. More severe thrombocytopenia was seen in patients with CSF
administration [66–68]. Further studies will be required to determine if chemora-
diotherapy for other sites of disease will provoke similar problems.
Possible reason for thrombocytopenia observed in this site of irradiation is that
the CSF mobilizes progenitor cells into the peripheral blood and new precursor cells
are destroyed during migration into the irradiation volume because large numbers of
these cells may be irradiated as they pass through the heart [67].
Outside of clinical trials, in patients receiving concomitant chemotherapy and
radiation therapy, treatment is suspended for severe neutropenia and is resumed
when the toxicity decreases to lower grade.
196 19 Hematological Side Effects

There is a concern about the reduction in the capacity of bone marrow recovery
with simultaneous treatment of G-CSF during radiation therapy in the postradiation
phase. Mechanisms like enhanced production of inhibitory cytokines, a decrease in
endogenous production of the growth factors due to the exogenously induced phar-
macological levels, downregulation of growth factor receptors on target cells [69],
and decreased sensitivity of bone marrow to radiation during G-CSF application are
proposed for this phenomenon [70]. Further investigations are needed to determine
the late bone marrow effect of G-CSF administration during radiation therapy and
the timing, duration, and dosage of G-CSF application for successful treatment and
a favorable outcome.
The American Society of Clinical Oncology (ASCO) guidelines recommend that
“CSFs should be avoided in patients receiving concomitant chemotherapy and radi-
ation therapy, particularly involving the mediastinum. In the absence of chemo-
therapy, therapeutic use of CSFs may be considered in patients receiving radiation
therapy alone if prolonged delays secondary to neutropenia are expected.”
European Society of Clinical Oncology (ESMO) guidelines also recommend that
“primary prophylaxis with G-CSF is not indicated during chemoradiotherapy to the
chest due to increased rate of bone marrow suppression associated with an increased
risk of complications and death” [71].
Three clinically available forms of G-CSF are [72]:

*A glycosylated form (lenograstim), which is produced by using the expression in


mammalian cells, and a *non-glycosylated form (filgrastim), which is produced
by using expression in E. coli [73]
*A pegylated form of non-glycosylated Hu G-CSF (pegfilgrastim)

Pegfilgrastim is developed to produce a long-acting filgrastim requiring less fre-


quent dosing than its parent drug [74]. A single dose of PEG-rhG-CSF has a similar
clinical safety to rhG-CSF and significant advantageous effects [75].
The recommended dose for G-CSF is 5 g/kg/day and for Pegfilgrastim is as a
single dose of either 100 mcg/kg (individualized) or of a total dose of 6 mg (general
approach) [71, 76]. Both are injected subcutaneously. A single subcutaneous injec-
tion of rhG-CSF results in an increase in circulating granulocytes within 2 h; the
granulocyte concentration peaks by 12 h and remains elevated for the next 36 h
before decreasing slowly back to baseline [77]. The median time to reach the peak
concentration is 2.5–5 days for pegfilgrastim, and neutrophilic level remains above
baseline for around 9–10 days [74].
Patients should be monitored with serial CBCs. Administration should be contin-
ued until the absolute neutrophil count reaches to upper level or normal [78].
The G-CSF receptors are expressed not only on myeloid cells but also on other
hematopoietic cells including monocytes, platelets, and possibly certain lympho-
cyte subsets [79]. No significant changes in lymphocyte or monocyte counts have
been observed during the course of rhG-CSF treatment.
Data on the effects of G-CSF on platelet function are limited. It seems that
G-CSF enhances platelet aggregation and activation in humans [80].
19.6 Treatment 197

Radiation-induced thrombocytopenia may be worsened by G-CSF administra-


tion, possibly due to a decrease in the number of megakaryocytes in the bone mar-
row and an increase in the trapping of megakaryocytes in the spleen [72, 81].
G-CSF-induced thrombocytopenia seems to be a transient event and may improve
spontaneously despite continual G-CSF treatment [82].
Common side effects observed during G-CSF treatment consist of mild muscu-
loskeletal pain, papular skin rash, and some laboratory abnormalities like elevated
alkaline phosphates [54, 61, 82].
The exact effects of G-CSF administration on cancer cells remain controversial.
G-CSF administration increases the production of neutrophils, and cytotoxic media-
tors produced by neutrophils could kill the cancer cells [83, 84]. On the other hand,
G-CSF has been shown to promote tumor angiogenesis by upregulating VEGF [85],
which is released by neutrophils and reduces radiation-induced vascular damage [86].
It has been shown that prophylactic G-CSF administration for treatment of
advanced unresectable head and neck cancers resulted in an unexpected reduced
local control [87, 88]. The effects of G-CSF on tumor growth, especially during
radiation therapy, are controversial and require further investigation [84].
Furthermore G-CSF can stimulate the clonal growth of some non-hematopoietic
tumor cells like bladder [89] or colon [90] cancers, and the remote effect of G-CSF
on the growth of tumor cells lying outside radiation treatment portals is proposed by
some [91].
It has been suggested that the simultaneous or sequential administration of two
or more complementary cytokines has resulted in better patterns of hematologic
recovery. The capacity of multiple cytokine combinations (like GM-CSF and IL-3
[92–94], TPO with IL-4 or IL-11 [95], IL-3 receptor agonist and rhG-CSF [96], and
rhG-CSF and pegylated megakaryocyte growth and development factor [97]) to
accelerate hematologic recovery and ensure multi-lineage protection following
severe radiation-induced myelosuppression has been assessed in animal studies
with promising results [92–98]. Further investigations in this field will clearly be
necessary before drawing any conclusions.

19.6.2 Thrombocytopenia

There is no threshold for platelet counts that for counts below which prophylactic
transfusion should be prescribed to all patients with radiation-induced thrombocy-
topenia. Several studies support a threshold of 10,000 platelets/mL or less for initi-
ating transfusions to patients with solid tumors and a higher threshold; perhaps
20,000/mL has been proposed for patients with tumor with the presence of necrotic
sites (e.g., gynecologic, colorectal, melanoma, or bladder tumors) [23, 99].
Thrombopoietin, a lineage-specific growth factor specific for platelets, regulates
platelet production via the stimulation of its receptor on megakaryocytes and plate-
lets leading to proliferation and differentiation. Two variant forms of human throm-
bopoietin have been developed including first-generation agents, recombinant
198 19 Hematological Side Effects

human thrombopoietin (rHuTPO) and pegylated recombinant human megakaryo-


cyte growth and development factor (PEG rHuMGDF), and second-generation
agents—the TPO peptide mimetics, TPO non-peptide mimetics, and TPO agonist
antibodies [100–102].
Clinical trials with recombinant TPO [100] and recently romiplostim (a TPO
peptide mimetic) [103] have shown that these agents can improve platelet counts in
patients receiving chemotherapy. Further studies need to be performed for use of
these agents in radiation-induced thrombocytopenia.
Platelet factor 4 (PF4) is released from radiation-damaged megakaryocytes and
inversely affects platelet count recovery after radiation. Anti-PF4 strategy can improve
outcomes of radiation-induced thrombocytopenia and needs to be explored [104].
Interleukin 11 (IL-11) is a stromal cell-derived cytokine that enhances the growth
of early progenitors and promotes megakaryocytopoiesis and erythropoiesis [105].
Given recombinant human interleukin-11 (rHuIL-11) activity in chemotherapy-­
induced thrombocytopenia, it needs to be studied as one possible therapeutic option
in radiation-induced thrombocytopenia [106–108].

19.6.3 Anemia

Radiation is more effective on tumor cells with higher oxygenation. Anemia is asso-
ciated with poor tumor oxygenation, relative tumor radioresistance, and decreases
the effectiveness of radiation therapy, with subsequent reductions in treatment out-
comes including locoregional control and overall survival of cancer patients [10,
109]. Correcting the radiation-induced anemia and maintaining normal hemoglobin
levels in patients undergoing radiation therapy have a significant importance due to
decrease in tumor hypoxia, improve treatment outcomes, and have a determinant
impact on quality of life [109–112]. Both blood transfusion and use of erythropoiesis-­
stimulating agents (e.g., darbepoetin, erythropoietin) can increase the hemoglobin
levels during radiation therapy, but the effects of medications used for correction of
radiation-induced anemia on locoregional control and overall survival are question-
able [113–117].
Many radiation oncologists have not given ample attention to treating mild to
moderate anemia during radiation therapy unless symptoms of severe anemia are
present or the hemoglobin falls below a threshold of 9–10 g/dL. However, managing
mild to moderate anemia is important for radiation therapy outcome improvement
and quality of life preservation [109, 118].
Radiation-induced anemia is usually mild and readily correctable. It has been
reported that for most patients with uterine and cervical cancer, the severity of ane-
mia during radiation therapy was modest (59% of anemic patients with hemoglobin
levels between 10 g/dL and 11.9 g/dL and 11% with hemoglobin levels between
9 g/dL and 9.9 g/dL). Only a few patients had levels below 8.9 g/dL. Data on
colorectal, prostate, lung, and breast cancers yielded similar results [109].
References 199

Blood transfusion and recombinant human erythropoietin (rHuEPO) administra-


tion can significantly improve radiation-induced anemia [111, 113, 119–122]. There
are no randomized studies comparing blood transfusion and rHuEPO efficacy and
safety in patients undergoing radiation therapy.
A number of studies have shown an effect of perioperative transfusion on cancer
recurrence rates [123–128]. The mechanism of how blood transfusion affects tumor
viability is related to some types of immunosuppression [129]. Therefore, some
have proposed that rHuEPO administration in patients undergoing radiation therapy
may have some benefits over transfusion due to its potential to correct anemia with-
out the risks associated with transfusion [130].
Several studies using transfusion to correct radiation-induced anemia have shown
improvement in local control, but there is no study comparing transfusion with
rHuEPO, and no definitive conclusion can be drawn about transfusion or rHuEPO
safety during radiation therapy.
It has been demonstrated that erythropoietin receptors are expressed in several
human cancer cells and may modulate the cellular effects of recombinant human
erythropoietin on cancer cells, leading to increased cell survival and growth [131,
132]. Belenkov et al. reported that the addition of exogenous recombinant human
erythropoietin induces cancer cells to become more resistant to ionizing radiation
and to cisplatin [133]. These data indicate that the administration of rHuEPO to
cancer patients undergoing radiation therapy should proceed with caution, espe-
cially when the cancer type has been shown to be positive for erythropoietin
receptor.
There is no guideline for erythropoietin use during radiation therapy. Based on
these findings, we recommend that erythropoietin should not be administered dur-
ing radiation therapy outside of the experimental setting.

References
1. Peters WA, Liu P, Barrett RJ, Stock RJ, Monk BJ, Berek JS et al (2000) Concurrent chemo-
therapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adju-
vant therapy after radical surgery in high-risk early-stage cancer of the cervix. J Clin Oncol
18(8):1606–1613
2. Keys HM, Bundy BN, Stehman FB, Muderspach LI, Chafe WE, Suggs CL et al (1999)
Cisplatin, radiation, and adjuvant hysterectomy compared with radiation and adjuvant hyster-
ectomy for bulky stage IB cervical carcinoma. N Engl J Med 340(15):1154–1161
3. Bartelink H, Roelofsen F, Eschwege F, Rougier P, Bosset J, Gonzalez DG et al (1997)
Concomitant radiotherapy and chemotherapy is superior to radiotherapy alone in the treat-
ment of locally advanced anal cancer: results of a phase III randomized trial of the European
Organization for Research and Treatment of Cancer Radiotherapy and Gastrointestinal
Cooperative Groups. J Clin Oncol 15(5):2040–2049
4. Pearcey R, Brundage M, Drouin P, Jeffrey J, Johnston D, Lukka H et al (2002) Phase III trial
comparing radical radiotherapy with and without cisplatin chemotherapy in patients with
advanced squamous cell cancer of the cervix. J Clin Oncol 20(4):966–972
5. Small W Jr, Winter K, Levenback C, Iyer R, Hymes SR, Jhingran A et al (2011) Extended-­
field irradiation and intracavitary brachytherapy combined with cisplatin and amifostine for
cervical cancer with positive para-aortic or high common iliac lymph nodes: results of arm II
200 19 Hematological Side Effects

of Radiation Therapy Oncology Group (RTOG) 0116. Int J Gynecol Cancer


21(7):1266–1275
6. Wan J, Liu K, Li K, Li G, Zhang Z (2015) Can dosimetric parameters predict acute hemato-
logic toxicity in rectal cancer patients treated with intensity-modulated pelvic radiotherapy?
Radiat Oncol 10(1):152
7. Jefferies S, Rajan B, Ashley S, Traish D, Brada M (1998) Haematological toxicity of cranio-­
spinal irradiation. Radiother Oncol 48(1):23–27
8. Farah R, Ultmann J, Griem M, Golomb H, Kalokhe U, Desser R et al (1988) Extended mantle
radiation therapy for pathologic stage I and II Hodgkin’s disease. J Clin Oncol
6(6):1047–1052
9. Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the radiation therapy oncology group
(RTOG) and the European organization for research and treatment of cancer (EORTC). Int
J Radiat Oncol Biol Phys 31(5):1341–1346
10. Harrison L, Shasha D, Shiaova L, White C, Ramdeen B, Portenoy R (eds) (2001) Prevalence
of anemia in cancer patients undergoing radiation therapy. Semin Oncol 28(2 Suppl
8):54–59
11. Fliedner T, Graessle D, Paulsen C, Reimers K (2002) Structure and function of bone marrow
hemopoiesis: mechanisms of response to ionizing radiation exposure. Cancer Biother
Radiopharm 17(4):405–426
12. Ellis R (1961) The distribution of active bone marrow in the adult. Phys Med Biol
5(3):255–258
13. Fliedner TM (1998) The role of blood stem cells in hematopoietic cell renewal. Stem Cells
16(6):361–374
14. Nothdurft W, Kreja L (1998) Hemopoietic progenitor cells in the blood as indicators of the
functional status of the bone marrow after total-body and partial-body irradiation: experi-
ences from studies in dogs. Stem Cells 16(S2):97–111
15. Rozgaj R, Kašuba V, Šentija K, Prlić I (1999) Radiation-induced chromosomal aberrations
and haematological alterations in hospital workers. Occup Med 49(6):353–360
16. Haas R, Bohne F, Fliedner T (1971) Cytokinetic analysis of slowly proliferating bone marrow
cells during recovery from radiation injury. Cell Prolif 4(1):31–45
17. Sykes MP, Chu FC, Wilkerson WG (1960) Local bone-marrow changes secondary to thera-
peutic irradiation 1. Radiology 75(6):919–924
18. Roeske JC, Lujan A, Reba RC, Penney BC, Yamada SD, Mundt AJ (2005) Incorporation of
SPECT bone marrow imaging into intensity modulated whole-pelvic radiation therapy treat-
ment planning for gynecologic malignancies. Radiother Oncol 77(1):11–17
19. Sykes MP, Chu FC, Savel H, Bonadonna G, Mathis H (1964) The effects of varying dosages
of irradiation upon sternal-marrow regeneration 1. Radiology 83(6):1084–1088
20. Hanks GE (1964) In vivo migration of colony-forming units from shielded bone marrow in
the irradiated mouse. Nature 203:1393–1395
21. Croizat H, Frindel E, Tubiana M (1980) The effect of partial body irradiation on haemopoi-
etic stem cell migration. Cell Prolif 13(3):319–325
22. Croizat H, Frindel E, Tubiana M (1970) Proliferative activity of the stem cells in the bone-­
marrow of mice after single and multiple irradiations (total-or partial-body exposure). Int
J Radiat Biol Relat Stud Phys Chem Med 18(4):347–358
23. Mauch P, Constine L, Greenberger J, Knospe W, Sullivan J, Liesveld JL et al (1995)
Hematopoietic stem cell compartment: acute and late effects of radiation therapy and chemo-
therapy. Int J Radiat Oncol Biol Phys 31(5):1319–1339
24. Parmentier C, Morardet N, Tubiana M (1983) Late effects on human bone marrow after
extended field radiotherapy. Int J Radiat Oncol Biol Phys 9(9):1303–1311
25. Tubiana M, Carde P, Frindel E (1993) Ways of minimising hematopoietic damage induced by
radiation and cytostatic drugs—the possible role of inhibitors. Radiother Oncol 29(1):1–17
26. Salzman JR, Kaplan HS (1971) Effect of prior splenectomy on hematologic tolerance during
total lymphoid radiotherapy of patients with Hodgkin’s disease. Cancer 27(2):471–478
27. Plowman P (1983) The effects of conventionally fractionated, extended portal radiotherapy
on the human peripheral blood count. Int J Radiat Oncol Biol Phys 9(6):829–839
References 201

28. Scarantino CW, Rubin P, Constine LS (1984) The paradoxes in patterns and mechanism of
bone marrow regeneration after irradiation: 1. Different volumes and doses. Radiother Oncol
2(3):215–225
29. Rubin P, Landman S, Mayer E, Keller B, Ciccio S (1973) Bone marrow regeneration and
extension after extended field irradiation in Hodgkin’s disease. Cancer 32(3):699–711
30. Zachariah B (1992) Case report: role of granulocyte colony stimulating factor in radiother-
apy. Am J Med Sci 304(4):252–253
31. Mac Manus M, Lamborn K, Khan W, Varghese A, Graef L, Knox S (1997) Radiotherapy-­
associated neutropenia and thrombocytopenia: analysis of risk factors and development of a
predictive model. Blood 89(7):2303–2310
32. Lujan AE, Mundt AJ, Yamada SD, Rotmensch J, Roeske JC (2003) Intensity-modulated
radiotherapy as a means of reducing dose to bone marrow in gynecologic patients receiving
whole pelvic radiotherapy. Int J Radiat Oncol Biol Phys 57(2):516–521
33. Hui B, Zhang Y, Shi F, Wang J, Wang T, Wang J et al (2014) Association between bone mar-
row dosimetric parameters and acute hematologic toxicity in cervical cancer patients under-
going concurrent chemoradiotherapy: comparison of three-dimensional conformal
radiotherapy and intensity-modulated radiation therapy. Int J Gynecol Cancer
24(9):1648–1652
34. Albuquerque K, Giangreco D, Morrison C, Siddiqui M, Sinacore J, Potkul R et al (2011)
Radiation-related predictors of hematologic toxicity after concurrent chemoradiation for cer-
vical cancer and implications for bone marrow–sparing pelvic IMRT. Int J Radiat Oncol Biol
Phys 79(4):1043–1047
35. Brixey CJ, Roeske JC, Lujan AE, Yamada SD, Rotmensch J, Mundt AJ (2002) Impact of
intensity-modulated radiotherapy on acute hematologic toxicity in women with gynecologic
malignancies. Int J Radiat Oncol Biol Phys 54(5):1388–1396
36. Liang Y, Bydder M, Yashar CM, Rose BS, Cornell M, Hoh CK et al (2013) Prospective study
of functional bone marrow-sparing intensity modulated radiation therapy with concurrent
chemotherapy for pelvic malignancies. Int J Radiat Oncol Biol Phys 85(2):406–414
37. Berg BCV, Malghem J, Lecouvet FE, Maldague B (1998) Magnetic resonance imaging of the
normal bone marrow. Skeletal Radiol 27(9):471–483
38. Basu S, Houseni M, Bural G, Chamroonat W, Udupa J, Mishra S et al (2007) Magnetic reso-
nance imaging based bone marrow segmentation for quantitative calculation of pure red mar-
row metabolism using 2-Deoxy-2-[F-18] fluoro-D-glucose-positron emission tomography: a
novel application with significant implications for combined structure–function approach.
Mol Imaging Biol 9(6):361–365
39. Uckun FM, Souza L, Waddick KG, Wick M, Song CW (1990) In vivo radioprotective effects
of recombinant human granulocyte colony-stimulating factor in lethally irradiated mice.
Blood 75(3):638–645
40. Waddick K, Song C, Souza L, Uckun F (1991) Comparative analysis of the in vivo radiopro-
tective effects of recombinant granulocyte colony-stimulating factor (G-CSF), recombinant
granulocyte-macrophage CSF, and their combination. Blood 77(11):2364–2371
41. Zucali J, Moreb J, Gibbons W, Alderman J, Suresh A, Zhang Y et al (1994) Radioprotection
of hematopoietic stem cells by interleukin-1. Exp Hematol 22(2):130–135
42. Neta R, Douches S, Oppenheim J (1986) Interleukin 1 is a radioprotector. J Immunol
136(7):2483–2485
43. Slørdal L, Muench MO, Warren DJ, Moore MA (1989) Radioprotection by murine and
human tumor-necrosis factor: dose-dependent effects on hematopoiesis in the mouse. Eur
J Haematol 43(5):428–434
44. Du N, Feng K, Luo C, Li L, Bai C, Pei X (2003) Radioprotective effect of FLT3 ligand
expression regulated by Egr-1 regulated element on radiation injury of SCID mice. Exp
Hematol 31(3):191–196
45. Hudak S, Leach M, Xu Y, Menon S, Rennick D (1998) Radioprotective effects of flk2/flt3
ligand. Exp Hematol 26(6):515–522
46. Streeter PR, Dudley LZ, Fleming WH (2003) Activation of the G-CSF and Flt-3 receptors
protects hematopoietic stem cells from lethal irradiation. Exp Hematol 31(11):1119–1125
202 19 Hematological Side Effects

47. Hofer M, Pospíšil M, Holá J, Vacek A, Štreitová D, Znojil V (2008) Inhibition of cyclooxy-
genase 2 in mice increases production of G-CSF and induces radioprotection. Radiat Res
170(5):566–571
48. Fukunaga R, Ishizaka-Ikeda E, Pan C, Seto Y, Nagata S (1991) Functional domains of the
granulocyte colony-stimulating factor receptor. EMBO J 10(10):2855–2865
49. Souza LM, Boone TC, Gabrilove J, Lai PH, Zsebo KM, Murdock DC et al (1986)
Recombinant human granulocyte colony-stimulating factor: effects on normal and leukemic
myeloid cells. Science 232(4746):61–65
50. Nagata S, Tsuchiya M, Asano S, Kaziro Y, Yamazaki T, Yamamoto O et al (1986) Molecular
cloning and expression of cDNA for human granulocyte colony-stimulating factor. Nature
319(6092):415–418
51. Babaeipour V, Khanchezar S, Mofid MR, Abbas MPH (2015) Efficient process development
of recombinant human granulocyte colony-stimulating factor (rh-GCSF) production in
Escherichia coli. Iran Biomed J 19(2):102–110
52. Sharifi TM, Habashi A, Rajabi MH (2013) Human granulocyte colony-stimulating factor
(hG-CSF) expression in plastids of Lactuca sativa. Iran Biomed J 17(3):158–164
53. Schmidberger H, Hess CF, Hoffmann W, Reuss-Borst MA, Bamberg M (1993) Granulocyte
colony-stimulating factor treatment of leucopenia during fractionated radiotherapy. Eur
J Cancer 29(14):1927–1931
54. Gava A, Bertossi L, Ferrarese F, Coghetto F, Marazzato G, Andrulli A et al (1998) Use of
filgrastim, granulocyte colony stimulating factor (G-CSF), in radiotherapy to reduce drop-­
outs because of radiogenic leukopenia. Radiol Med 95(3):232–236
55. Zhang H (1993) Stimulation of low dose radiation on hematopoietic system. Zhonghua Yi
Xue Za Zhi 73(2):99–100, 27–28
56. Fushiki M, Ono K, Sasai K, Shibamoto Y, Tsutsui K, Nishidai T et al (1990) Effect of recom-
binant human granulocyte colony-stimulating factor on granulocytopenia in mice induced by
irradiation. Int J Radiat Oncol Biol Phys 18(2):353–357
57. Schuening FG, Storb R, Goehle S, Graham TC, Appelbaum FR, Hackman R et al (1989)
Effect of recombinant human granulocyte colony-stimulating factor on hematopoiesis of nor-
mal dogs and on hematopoietic recovery after otherwise lethal total body irradiation. Blood
74(4):1308–1313
58. Marks LB, Friedman HS, Kurtzberg J, Oakes WJ, Hockenberger BM (1992) Reversal of
radiation-induced neutropenia by granulocyte colony-stimulating factor. Med Pediatr Oncol
20(3):240–242
59. MacVittie T, Monroy R, Patchen M, Souza L (1990) Therapeutic use of recombinant human
G-CSF (rhG-CSF) in a canine model of sublethal and lethal whole-body irradiation. Int
J Radiat Biol 57(4):723–736
60. Knox SJ, Fowler S, Marquez C, Hoppe RT (1994) Effect of filgrastim (G-CSF) in Hodgkin’s
disease patients treated with radiation therapy. Int J Radiat Oncol Biol Phys 28(2):445–450
61. Fyles AW, Manchul L, Levin W, Robertson JM, Sturgeon J, Tsuji D (1998) Effect of filgras-
tim (G-CSF) during chemotherapy and abdomino-pelvic radiation therapy in patients with
ovarian carcinoma. Int J Radiat Oncol Biol Phys 41(4):843–847
62. Kolotas C, Zamboglou N, Schnabel T, Bojar H, Wintzer A, Vogt H-G et al (1996) Effect of
recombinant human granulocyte colony stimulating factor (R-metHuG-CSF) as an adjunct to
large-field radiotherapy: a phase I study. Int J Radiat Oncol Biol Phys 35(1):137–142
63. Root RK, Dale DC (1999) Granulocyte colony-stimulating factor and granulocyte-­
macrophage colony-stimulating factor: comparisons and potential for use in the treatment of
infections in nonneutropenic patients. J Infect Dis 179(Suppl 2):S342–S352
64. Price T, Chatta G, Dale D (1996) Effect of recombinant granulocyte colony-stimulating fac-
tor on neutrophil kinetics in normal young and elderly humans. Blood 88(1):335–340
65. Lord B, Bronchud M, Owens S, Chang J, Howell A, Souza L et al (1989) The kinetics of
human granulopoiesis following treatment with granulocyte colony-stimulating factor
in vivo. Proc Natl Acad Sci 86(23):9499–9503
References 203

66. Bunn PA, Crowley J, Kelly K, Hazuka MB, Beasley K, Upchurch C et al (1995)
Chemoradiotherapy with or without granulocyte-macrophage colony-stimulating factor in
the treatment of limited-stage small-cell lung cancer: a prospective phase III randomized
study of the Southwest Oncology Group. J Clin Oncol 13(7):1632–1641
67. American Society of Clinical Oncology (1994) American Society of Clinical Oncology.
Recommendations for the use of hematopoietic colony-stimulating factors: evidence-based,
clinical practice guidelines. J Clin Oncol 12(11):2471–2508
68. Sheikh H, Colaco R, Lorigan P, Blackhall F, Califano R, Ashcroft L et al (2011) Use of
G-CSF during concurrent chemotherapy and thoracic radiotherapy in patients with limited-­
stage small-cell lung cancer safety data from a phase II trial. Lung Cancer 74(1):75–79
69. Pospíšil M, Hofer M, Netíková J, Holá J, Znojil V, Vácha J et al (1999) Pretreatment with
granulocyte colony-stimulating factor reduces myelopoiesis in irradiated mice. Radiat Res
151(3):363–367
70. Pape H, Orth K, Heese A, Heyll A, Kobbe G, Schmitt G et al (2006) G-CSF during large field
radiotherapy reduces bone marrow recovery capacity. Eur J Med Res 11(8):322–328
71. Crawford J, Caserta C, Roila F, ESMO Guidelines Working Group (2009) Hematopoietic
growth factors: ESMO recommendations for the applications. Ann Oncol 20(Suppl
4):iv162–iv165
72. Son Y, Bae MJ, Lee CG, Jo WS, Kim SD, Yang K et al (2014) Treatment with granulocyte
colony-stimulating factor aggravates thrombocytopenia in irradiated mice. Mol Cell Toxicol
10(3):311–317
73. Vanz AL, Renard G, Palma MS, Chies JM, Dalmora SL, Basso LA et al (2008) Human granu-
locyte colony stimulating factor (hG-CSF): cloning, overexpression, purification and charac-
terization. Microb Cell Fact 7(1):13
74. Molineux G (2003) Pegfilgrastim: using pegylation technology to improve neutropenia sup-
port in cancer patients. Anticancer Drugs 14(4):259–264
75. FP W, Wang J, Wang H, Li N, Guo Y, Cheng YJ et al (2015) Clinical observation of the thera-
peutic effects of pegylated recombinant human granulocyte colony-stimulating factor in
patients with concurrent chemoradiotherapy-induced grade IV neutropenia. Exp Ther Med
9(3):761–765
76. Smith TJ, Khatcheressian J, Lyman GH, Ozer H, Armitage JO, Balducci L et al (2006) 2006
update of recommendations for the use of white blood cell growth factors: an evidence-based
clinical practice guideline. J Clin Oncol 24(19):3187–3205
77. Cohen AM, Zsebo KM, Inoue H, Hines D, Boone TC, Chazin VR et al (1987) In vivo stimu-
lation of granulopoiesis by recombinant human granulocyte colony-stimulating factor. Proc
Natl Acad Sci 84(8):2484–2488
78. Adamietz I, Rosskopf B, Dapper F, Von Lieven H, Boettcher H (1996) Comparison of two
strategies for the treatment of radiogenic leukopenia using granulocyte colony stimulating
factor. Int J Radiat Oncol Biol Phys 35(1):61–67
79. Demetri GD, Griffin JD (1991) Granulocyte colony-stimulating factor and its receptor. Blood
78(11):2791–2808
80. Spiel AO, Bartko J, Schwameis M, Firbas C, Siller-Matula J, Schuetz M et al (2011) Increased
platelet aggregation and in vivo platelet activation after granulocyte colony-stimulating factor
administration. A randomised controlled trial. Thromb Haemost 105(4):655–662
81. Akizuki S, Mizorogi F, Inoue T, Sudo K, Ohnishi A (2000) Pharmacokinetics and adverse
events following 5-day repeated administration of lenograstim, a recombinant human granu-
locyte colony-stimulating factor, in healthy subjects. Bone Marrow Transplant
26(9):939–946
82. Takamatsu Y, Jimi S, Sato T, Hara S, Suzumiya J, Tamura K (2007) Thrombocytopenia in
association with splenomegaly during granulocyte–colony-stimulating factor treatment in
mice is not caused by hypersplenism and is resolved spontaneously. Transfusion
47(1):41–49
204 19 Hematological Side Effects

83. Di Carlo E, Forni G, Lollini P, Colombo MP, Modesti A, Musiani P (2001) The intriguing
role of polymorphonuclear neutrophils in antitumor reactions. Blood 97(2):339–345
84. Kim JS, Son Y, Bae MJ, Lee M, Lee CG, Jo WS et al (2015) Administration of granulocyte
colony-stimulating factor with radiotherapy promotes tumor growth by stimulating vascular-
ization in tumor-bearing mice. Oncol Rep 34(1):147–154
85. Natori T, Sata M, Washida M, Hirata Y, Nagai R, Makuuchi M (2002) G-CSF stimulates
angiogenesis and promotes tumor growth: potential contribution of bone marrow-derived
endothelial progenitor cells. Biochem Biophys Res Commun 297(4):1058–1061
86. Garcia-Barros M, Paris F, Cordon-Cardo C, Lyden D, Rafii S, Haimovitz-Friedman A et al
(2003) Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science
300(5622):1155–1159
87. Vokes EE, Haraf DJ, Mick R, McEvilly J-M, Weichselbaum RR (1994) Intensified concomi-
tant chemoradiotherapy with and without filgrastim for poor-prognosis head and neck cancer.
J Clin Oncol 12(11):2351–2359
88. Staar S, Rudat V, Stuetzer H, Dietz A, Volling P, Schroeder M et al (2001) Intensified hyper-
fractionated accelerated radiotherapy limits the additional benefit of simultaneous chemo-
therapy—results of a multicentric randomized German trial in advanced head-and-neck
cancer. Int J Radiat Oncol Biol Phys 50(5):1161–1171
89. Ohigashi T, Tachibana M, Tazaki H, Nakamura K (1992) Bladder cancer cells express func-
tional receptors for granulocyte-colony stimulating factor. J Urol 147(1):283–286
90. Berdel WE, Danhauser-Riedl S, Steinhauser G, Winton EF (1989) Various human hemato-
poietic growth factors (interleukin-3, GM-CSF, G-CSF) stimulate clonal growth of nonhema-
topoietic tumor cells [see comments]. Blood 73(1):80–83
91. Mac Manus M, McCormick D, Trimble A, Abram W (1995) Value of granulocyte colony
stimulating factor in radiotherapy induced neutropenia: clinical and laboratory studies. Eur
J Cancer 31(3):302–307
92. Farese AM, Williams DE, Seiler FR, Macvittie TJ (1993) Combination protocols of cytokine
therapy with interleukin-3 and granulocyte-macrophage colony-stimulating factor in a pri-
mate model of radiation-induced marrow aplasia. Blood 82(10):3012–3018
93. Williams D, Dunn J, Park L, Frieden E, Seiler F, Farese A et al (1992) A GM-CSF/IL-3 fusion
protein promotes neutrophil and platelet recovery in sublethally irradiated rhesus monkeys.
Biotechnol Ther 4(1–2):17–29
94. Vadhan-Raj S (1994) PIXY321 (GM-CSF/IL-3 fusion protein): biology and early clinical
development. Stem Cells 12(3):253–261
95. Van der Meeren A, Mouthon M-A, Vandamme M, Squiban C, Aigueperse J (2004)
Combinations of cytokines promote survival of mice and limit acute radiation damage in
concert with amelioration of vascular damage. Radiat Res 161(5):549–559
96. MacVittie TJ, Farese AM, Herodin F, Grab LB, Baum CM, McKearn JP (1996) Combination
therapy for radiation-induced bone marrow aplasia in nonhuman primates using synthokine
SC-55494 and recombinant human granulocyte colony-stimulating factor. Blood
87(10):4129–4135
97. Farese AM, Hunt P, Grab LB, MacVittie TJ (1996) Combined administration of recombinant
human megakaryocyte growth and development factor and granulocyte colony-stimulating
factor enhances multilineage hematopoietic reconstitution in nonhuman primates after
radiation-­induced marrow aplasia. J Clin Investig 97(9):2145
98. Hérodin F, Bourin P, Mayol J-F, Lataillade J-J, Drouet M (2003) Short-term injection of
antiapoptotic cytokine combinations soon after lethal γ-irradiation promotes survival. Blood
101(7):2609–2616
99. Schiffer CA, Anderson KC, Bennett CL, Bernstein S, Elting LS, Goldsmith M et al (2001)
Platelet transfusion for patients with cancer: clinical practice guidelines of the American
Society of Clinical Oncology. J Clin Oncol 19(5):1519–1538
100. Vadhan-Raj S, Murray LJ, Bueso-Ramos C, Patel S, Reddy SP, Hoots WK et al (1997)
Stimulation of megakaryocyte and platelet production by a single dose of recombinant human
thrombopoietin in patients with cancer. Ann Intern Med 126(9):673–681
References 205

101. Kuter DJ (2009) Thrombopoietin and thrombopoietin mimetics in the treatment of thrombo-
cytopenia. Annu Rev Med 60:193–206
102. Kuter DJ (2008) New drugs for familiar therapeutic targets: thrombopoietin receptor agonists
and immune thrombocytopenic purpura. Eur J Haematol 80(Suppl 69):9–18
103. Parameswaran R, Lunning M, Mantha S, Devlin S, Hamilton A, Schwartz G et al (2014)
Romiplostim for management of chemotherapy-induced thrombocytopenia. Support Care
Cancer 22(5):1217–1222
104. Lambert MP, Xiao L, Nguyen Y, Kowalska MA, Poncz M (2011) The role of platelet factor
4 in radiation-induced thrombocytopenia. Int J Radiat Oncol Biol Phys 80(5):1533–1540
105. Teramura M, Kobayashi S, Yoshinaga K, Iwabe K, Mizoguchi H (1996) Effect of interleukin
11 on normal and pathological thrombopoiesis. Cancer Chemother Pharmacol
38(1):S99–S102
106. Gordon M, McCaskill-Stevens W, Battiato L, Loewy J, Loesch D, Breeden E et al (1996) A
phase I trial of recombinant human interleukin-11 (neumega rhIL-11 growth factor) in
women with breast cancer receiving chemotherapy. Blood 87(9):3615–3624
107. Isaacs C, Robert NJ, Bailey FA, Schuster MW, Overmoyer B, Graham M et al (1997)
Randomized placebo-controlled study of recombinant human interleukin-11 to prevent
chemotherapy-­induced thrombocytopenia in patients with breast cancer receiving dose-­
intensive cyclophosphamide and doxorubicin. J Clin Oncol 15(11):3368–3377
108. Tepler I, Elias L, Hussein M, Rosen G, Chang A, Moore J et al (1996) A randomized placebo-­
controlled trial of recombinant human interleukin-11 in cancer patients with severe thrombo-
cytopenia due to chemotherapy. Blood 87(9):3607–3614
109. Harrison LB, Shasha D, White C, Ramdeen B (2000) Radiotherapy-associated anemia: the
scope of the problem. Oncologist 5(Suppl 2):1–7
110. Dunst J (2004) Low hemoglobin levels: influence on tumor biology and radiotherapy treat-
ment outcome. Eur J Cancer 2(Suppl 2):3–10
111. Grogan M, Thomas GM, Melamed I, Wong FL, Pearcey RG, Joseph PK et al (1999) The
importance of hemoglobin levels during radiotherapy for carcinoma of the cervix. Cancer
86(8):1528–1536
112. Hofheinz R-D, Raab B, Mai S, Wenz F, Willeke F, Emig M et al (2004) Impact of
chemoradiotherapy-­induced anemia on survival in uniformly staged patients with locally
advanced squamous cell carcinoma of the esophagus. Oncol Res Treat 27(5):462–466
113. Lambin P, Ramaekers BL, van Mastrigt GA, Van den Ende P, de Jong J, De Ruysscher DK
et al (2009) Erythropoietin as an adjuvant treatment with (chemo) radiation therapy for head
and neck cancer. Cochrane Libr. doi:10.1002/14651858.CD006158.pub2
114. Shenouda G, Zhang Q, Ang KK, Machtay M, Parliament MB, Hershock D et al (2015) Long-­
term results of radiation therapy oncology group 9903: a randomized phase 3 trial to assess
the effect of erythropoietin on local-regional control in anemic patients treated with radiation
therapy for squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys
91(5):907–915
115. Machtay M, Pajak TF, Suntharalingam M, Shenouda G, Hershock D, Stripp DC et al (2007)
Radiotherapy with or without erythropoietin for anemic patients with head and neck cancer:
a randomized trial of the Radiation Therapy Oncology Group (RTOG 99-03). Int J Radiat
Oncol Biol Phys 69(4):1008–1017
116. Henke M, Laszig R, Rübe C, Schäfer U, Haase K-D, Schilcher B et al (2003) Erythropoietin
to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised,
double-blind, placebo-controlled trial. Lancet 362(9392):1255–1260
117. Overgaard J (2013) SP-033: randomized study of darbepoetin alfa as modifier of radiotherapy
in patients SCCHN. Final outcome of the dahanca 10 trial. Radiother Oncol 106:S13
118. Harrison LB, Chadha M, Hill RJ, Hu K, Shasha D (2002) Impact of tumor hypoxia and ane-
mia on radiation therapy outcomes. Oncologist 7(6):492–508
119. Glaser CM, Millesi W, Kornek GV, Lang S, Schüll B, Watzinger F et al (2001) Impact of
hemoglobin level and use of recombinant erythropoietin on efficacy of preoperative
206 19 Hematological Side Effects

chemoradiation therapy for squamous cell carcinoma of the oral cavity and oropharynx. Int
J Radiat Oncol Biol Phys 50(3):705–715
120. Vijayakumar S, Roach M, Wara W, Chan SK, Ewing C, Rubin S et al (1993) Effect of subcu-
taneous recombinant human erythropoietin in cancer patients receiving radiotherapy: pre-
liminary results of a randomized, open-labeled, phase II trial. Int J Radiat Oncol Biol Phys
26(4):721–729
121. Lavey RS, Dempsey WH (1993) Erythropoietin increases hemoglobin in cancer patients dur-
ing radiation therapy. Int J Radiat Oncol Biol Phys 27(5):1147–1152
122. Dusenbery KE, McGuire WA, Holt PJ, Carson LF, Fowler JM, Twiggs LB et al (1994)
Erythropoietin increases hemoglobin during radiation therapy for cervical cancer. Int J Radiat
Oncol Biol Phys 29(5):1079–1084
123. Pastorino U, Valente M, Cataldo I, Lequaglie C, Ravasi G (1986) Perioperative blood transfu-
sion and prognosis of resected stage Ia lung cancer. Eur J Cancer Clin Oncol
22(11):1375–1378
124. Moffat L, Sunderland G, Lamont D (1987) Blood transfusion and survival following nephrec-
tomy for carcinoma of kidney. Br J Urol 60(4):316–319
125. Rosenberg SA, Seipp CA, White DE, Wesley R (1985) Perioperative blood transfusions are
associated with increased rates of recurrence and decreased survival in patients with high-­
grade soft-tissue sarcomas of the extremities. J Clin Oncol 3(5):698–709
126. Kaneda M, Horimi T, Ninomiya M, Nagae S, Mukai K, Takeda I et al (1987) Adverse affect
of blood transfusions on survival of patients with gastric cancer. Transfusion 27(5):375–377
127. Creasy T, Veitch P, Bell P (1987) A relationship between perioperative blood transfusion and
recurrence of carcinoma of the sigmoid colon following potentially curative surgery. Ann R
Coll Surg Engl 69(3):100–103
128. Arnoux R, Corman J, Peloquin A, Smeesters C, St-Louis G (1988) Adverse effect of blood
transfusions on patient survival after resection of rectal cancer. Can J Surg 31(2):121–126
129. Jones KR, Weissler MC (1990) Blood transfusion and other risk factors for recurrence of
cancer of the head and neck. Arch Otolaryngol Head Neck Surg 116(3):304–309
130. Bush RS (1986) The significance of anemia in clinical radiation therapy. Int J Radiat Oncol
Biol Phys 12(11):2047–2050
131. Arcasoy MO, Jiang X, Haroon ZA (2003) Expression of erythropoietin receptor splice vari-
ants in human cancer. Biochem Biophys Res Commun 307(4):999–1007
132. Westenfelder C, Baranowski RL (2000) Erythropoietin stimulates proliferation of human
renal carcinoma cells. Kidney Int 58(2):647–657
133. Belenkov AI, Shenouda G, Rizhevskaya E, Cournoyer D, Belzile J-P, Souhami L et al (2004)
Erythropoietin induces cancer cell resistance to ionizing radiation and to cisplatin. Mol
Cancer Ther 3(12):1525–1532
Radiation-Induced Nausea and Vomiting
(RINV) 20

Radiation-induced nausea and vomiting (RINV) is generally less frequent and less
severe than the nausea and vomiting encountered in patients receiving chemother-
apy. This problem is often underestimated and undertreated by radiation oncologists
in clinical practice; however, it can be very distressing and may cause delay or even
interruption of radiotherapy [1–3]. The incidence of RINV was studied in two pro-
spective observational studies. The Italian Group for Antiemetic Research in
Radiotherapy (IGARR) analyzed the incidence of RINV in 1020 patients receiving
various kinds of radiation therapy without concurrent chemotherapy. Vomiting and
nausea occurred in 11% and 27.1% of patients, respectively, and 27.9% of patients
had both vomiting and nausea [1]. Nausea was twice as frequent as vomiting (27.1%
vs. 11%) and lasted longer (median duration 10 vs. 3 days). The RINV incidence
rates based on anatomical sites were 28% for breast, 39% for pelvis, 40.4% for head
and neck, 48.8% for thorax, 40.4% for brain, and 71.4% for upper abdominal sites.
In a second study of 368 patients receiving radiation therapy again without con-
current chemotherapy, the overall incidence rates for nausea and vomiting were
39% and 7%, respectively, with 63% in abdominal or pelvic fields and by 48% in
head/neck/brain fields [4].

20.1 Mechanism

The mechanism of RINV differs depending on radiation site. RINV is primarily due
to a serotonin (5-hydroxytryptamine)-mediated pathway [5]. Damage to the entero-
chromaffin cells of the gastrointestinal mucosa by radiation leads to release of sero-
tonin, which may initiate the emetogenic response through activation of serotonin
receptors, visceral afferent fibers, and the chemoreceptor trigger zone (CTZ) in the
brain stem [6].
If the gastrointestinal tract is included in the field of radiation, direct effects are
likely with stimulation of afferent pathways in the upper gastrointestinal tract.

© Springer International Publishing AG 2017 207


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6_20
208 20 Radiation-Induced Nausea and Vomiting (RINV)

RINV also can be mediated by activation of CTZ through release of humoral factors
from tissue injury [7].
Increased intracranial pressure due to radiation-induced edema and inflamma-
tory processes associated with radiation injury may also play a role in RINV [7].

20.2 Timing

Acute RINV is seen most frequently with radiation therapy. The latent period ranges
from 30 min to 4 h in single-fraction study [6], and it generally begins 3–4 days after
fractionated radiation therapy [1, 4, 8]. Delayed emesis is not observed with radia-
tion therapy, unlike with chemotherapy, and anticipatory emesis is extremely rare
with radiation therapy [6].

20.3 Risk Factors

The risk of developing RINV depends on several patient and treatment-related risk
factors.
The most significant radiotherapy-related risk factors include the site (upper
abdomen) and radiation field size (>400 cm) [1, 9]. Other factors like single and total
dose, fractionation schedule, and treatment techniques are also important [6]. A sin-
gle high dose and larger dose per fraction have a greater risk of inducing RINV [10].
Gagnon and Kuettel demonstrated diurnal variation in the gastrointestinal tract
with more sensitivity to radiation-induced damage in the late morning than in the
afternoon. This study was limited to prostate cancer patients [11].
Patient-related risk factors include age, gender, general health status, alcohol
consumption, concurrent or previous chemotherapy, and previous experience of
emesis [6]. The risk of RINV is higher in female patients, those younger than
50 years, and patients with a previous history of poorly controlled emesis.
Conversely, the risk is decreased in those with a high alcohol intake [6].
It has been reported that the incidence of RINV may have some correlation with
ABO blood group and it seems that patients with type A blood groups are the most
vulnerable individuals to these side effects [12].

20.4 Symptoms

Patients with RINV experience lower well-being and quality of life, lower satisfac-
tion with aspects of daily living, and more frequent fatigue, anxiety, and depression
[4]. Prolonged vomiting can cause dehydration, electrolyte imbalances, and
malnutrition.
20.5 Prevention and Management 209

20.5 Prevention and Management

Radiation therapy sites are classified base on their potential risk for developing
RINV in ASCO and MASCC/ESMO guidelines (Table 20.1) [13, 14]. The National
Comprehensive Cancer Network (NCCN) [15], Multinational Association for
Supportive Care in Cancer (MASCC)/European Society for Medical Oncology
(ESMO) [14], and American Society of Clinical Oncology (ASCO) [13] have
released guidelines for the management of RINV based on risk category (Table 20.2).
The NCCN guidelines stratify patients into two categories of total-body irradia-
tion and upper abdomen/localized sites [15]. Localized sites have not been deter-
mined, but recommendations for both are nearly the same (Table 20.2).
In the setting of concurrent chemoradiotherapy, antiemetic strategies consistent
with chemotherapy agents being used should be offered based on each of the three
guidelines.
Neurokinin-1 (NK-1) receptor inhibitors in combination with a 5-HT3 receptor
antagonist and dexamethasone improve control of acute and delayed chemotherapy-­
induced nausea and vomiting after highly and moderately emetogenic chemother-
apy. The role of these agents in the management of RINV is not well studied. Dennis
et al. have suggested that the combination of aprepitant and granisetron may have a
protective effect against RINV after both single- and multiple-fraction moderately
emetogenic radiation therapy for thoracolumbar irradiation, which is more than his-
torical control data from patients receiving prophylaxis with a 5-HT3 receptor
antagonists alone [16]. Further evaluation of this combination in patients receiving
concurrent chemoradiotherapy is ongoing.
The optimal duration of antiemetic therapies for the prevention of RINV is not
clear. The MASCC/ESMO guidelines have no recommendation for the duration of
5-HT3 antagonist treatment, and the ASCO and NCCN guidelines recommend
5-HT3 antagonist administration prior to each radiation fraction for high- and
moderate-­risk radiation therapies. Some argued that RINV episodes occur signifi-
cantly during the first and second week of treatment, and prophylactic antiemetics
may not be necessary for a full course of radiation therapy. This issue needs to be
further evaluated in future studies to demonstrate the optimal duration of adminis-
tration [8, 17].

Table 20.1 Risk classification for RINV


Definition
High risk Total body irradiation and total nodal irradiation
Moderate risk Upper abdomen, half-body irradiation, upper-body
irradiation
Low risk Cranium, craniospinal, head and neck, lower thorax
region, pelvis
Minimal risk Breast, extremity
210 20 Radiation-Induced Nausea and Vomiting (RINV)

Table 20.2 Recommendations for RINV


MASCC/ESMO ASCO NCCN
High risk A 5-HT3 receptor A 5-HT3 receptor Total-body irradiation:
antagonist + dexamethasone antagonist before Granisetron 2 mg PO daily
each fraction and or ondansetron 8 mg PO
for at least 24 h twice daily ± dexamethasone
after completion 4 mg PO daily prior to each
or radiotherapy + a day of radiotherapy
5-day course of
dexamethasone
during fraction
1–5
Moderate A 5-HT3 receptor antagonist; a A 5-HT3 receptor
risk short course of dexamethasone is antagonist before
optional each fraction and
for at least 24 h
after completion
or radiotherapy + a
short course of
dexamethasone
during fraction
1–5 may be
offered
Low risk A 5-HT3 receptor antagonist as A 5-HT3 receptor
either prophylaxis or rescue antagonist alone
as either
prophylaxis or
rescue. For
patients suffering
from RINV while
receiving rescue
therapy only,
prophylactic
treatment should
continue until
radiotherapy is
completed
Minimal A dopamine receptor antagonist or A dopamine
risk a 5-HT3 receptor antagonist rescue receptor
antagonist or a
5-HT3 receptor
antagonist rescue.
Antiemetic
prophylaxis
should continue
throughout
radiation
treatment if a
patient
experiences RINV
while receiving
rescue therapy
References 211

There are five different 5-HT3 receptor antagonist agents. Granisetron and
ondansetron are commonly employed for the treatment of RINV. In general, these
agents are considered to be equally effective; however, pharmacologic differences
do exist between these two agents [17–19].
Granisetron binds irreversibly to 5-HT3 receptors with antiemetic activity up to
48 h following administration, and ondansetron binds reversibly to the receptor, losing
antagonist activity by 24 h following administration of commonly employed doses.
Higher doses or multiple-daily dosing of ondansetron may be required to maintain
antiemetic efficacy; however, granisetron is effective as a once-daily dose [19]:

Granisetron: 2 mg orally, 1 mg or 0.01 mg/kg IV


Ondansetron: 8 mg orally 2–3 times daily, 8 mg or 0.15 mg/kg IV

Side effects include headache, constipation, diarrhea, asthenia, and dizziness.


5-HT3 receptor antagonists have been reported to produce some dose-dependent,
nonclinically asymptomatic changes in electrocardiographic parameters like
increased PR interval, QT interval, or QRS complex duration. Intravenous granise-
tron has been shown to be associated with fewer effects on the electrocardiogram
than intravenous ondansetron [20]. In patients at risk for QT prolongation, 5-HT3
receptor antagonists should be used with caution (e.g., patients with electrolyte
abnormalities, congestive heart failure, brady-arrhythmias, in combination with
other medications that cause QT prolongation) [21].
Two dopamine agonist receptors are more commonly used [18]:

Metoclopramide: 20 mg orally
Prochlorperazine: 10 mg orally or IV

References
1. Maranzano E, De Angelis V, Pergolizzi S, Lupattelli M, Frata P, Spagnesi S, Frisio ML,
Mandoliti G, Malinverni G, Trippa F, Fabbietti L, Parisi S, Di Palma A, De Vecchi P, De Renzis
C, Giorgetti C, Bergami T, Orecchia R, Portaluri M, Signor M, Di Gennaro D, Italian Group
for Antiemetic Research in Radiotherapy (IGARR) (2010) A prospective observational trial on
emesis in radiotherapy: analysis of 1020 patients recruited in 45 Italian radiation oncology
centres. Radiother Oncol 94(1):36–41
2. Maranzano E, Feyer PC, Molassiotis A, Rossi R, Clark-Snow RA, Olver I, Warr D, Schiavone
C, Roila F, Participants in the Perugia Consensus Conference 2004 (2005) Evidence-based
recommendations for the use of antiemetics in radiotherapy. Radiother Oncol 76(3):227–233
3. Tonini G, Vincenzi B, Santini D, La Cesa A, Finolezzi E, Onori N, D’Angelillo R, Baldi A,
Trodella L (2003) Prevention of radiotherapy-induced emesis. J Exp Clin Cancer Res
22(1):17–22
4. Enblom A, Axelsson BB, Steineck G, Hammar M, Börjeson S (2009) One third of patients
with radiotherapy-induced nausea consider their antiemetic treatment insufficient. Support
Care Cancer 17(1):23–32
212 20 Radiation-Induced Nausea and Vomiting (RINV)

5. Scarantino CW, Ornitz RD, Hoffman LG, Anderson RF Jr (1994) On the mechanism of
radiation-­induced emesis: the role of serotonin. Int J Radiat Oncol Biol Phys 30(4):825–830
6. Feyer PC, Stewart AL, Titlbach OJ (1998) Aetiology and prevention of emesis induced by
radiotherapy. Support Care Cancer 6(3):253–260
7. Grahame-Smith DG (1968) Nausea and vomiting: mechanisms and treatment. Springer-­
Verlag, Berlin
8. Poon M, Dennis K, DeAngelis C, Chung H, Stinson J, Zhang L, Bedard G, Popovic M, Lao N,
Pulenzas N, Wong S, Chow E (2014) A prospective study of gastrointestinal radiation therapy-­
induced nausea and vomiting. Support Care Cancer 22(6):1493–1507
9. IGARR (the Italian Group for Antiemetic Research in Radiotherapy) (1999) Radiation-
induced emesis: a prospective observational multicenter Italian trial. Int J Radiat Oncol Biol
Phys 44(3):619–625
10. Scarantino CW, Ornitz RD, Hoffman LG, Anderson RF Jr (1992) Radiation-induced emesis:
effects of ondansetron. Semin Oncol 19(6 Suppl 15):38–43
11. Gagnon GJ, Kuettel M (1994) Diurnal variation in acute GI toxicity from prostate cancer
radiotherapy. Int J Radiat Oncol Biol Phys 30(1):253
12. Habibi M, Namimoghadam A, Korouni R, Fashiri P, Borzoueisileh S, Elahimanesh F, Amiri F,
Moradi G (2016) Radiation-induced nausea and vomiting: is ABO blood group as important as
radiation and patient-related factors? An observational study. Medicine (Baltimore)
95(31):e4334
13. Basch E, Prestrud AA, Hesketh PJ, Kris MG, Feyer PC, Somerfield MR, Chesney M, Clark-­
Snow RA, Flaherty AM, Freundlich B, Morrow G, Rao KV, Schwartz RN, Lyman GH,
American Society of Clinical Oncology (2011) Antiemetics: American Society of Clinical
Oncology clinical practice guideline update. J Clin Oncol 29(31):4189–4198
14. Gralla RJ, Roila F, Tonato M, Herrstedt J. MASCC/ESMO Antiemetic Guideline 2013.
Multinational Association of Supportive Care in Cancer. http://ekstern.infonet.regionsyddan-
mark.dk/files/Formularer/Upload/2013/09/mascc_guidelines_english_2013.pdf. Accessed 30
Mar 2016
15. NCCN Clinical Practice Guidelines in Oncology. Antiemesis. Version 1.2016. National
Comprehensive Cancer Network. Accessed 30 Mar 2016
16. Dennis K, De Angelis C, Jon F, Lauzon N, Pasetka M, Holden L, Barnes E, Danjoux C, Sahgal
A, Tsao M, Chow E (2014) Aprepitant and granisetron for the prophylaxis of radiotherapy-­
induced nausea and vomiting after moderately emetogenic radiotherapy for bone metastases:
a prospective pilot study. Curr Oncol 21(6):e760–e767
17. Dennis K, Makhani L, Maranzano E, Feyer P, Zeng L, De Angelis C, Holden L, Wong CS,
Chow E (2013) Timing and duration of 5-HT3 receptor antagonist therapy for the prophylaxis
of radiotherapy-induced nausea and vomiting: a systematic review of randomized and non-­
randomized studies. J Radiat Oncol 2(3):271–284
18. Feyer P, Jahn F, Jordan K (2015) Prophylactic management of radiation-induced nausea and
vomiting. Biomed Res Int 2015:893013
19. Horiot JC (2004) Prophylaxis versus treatment: is there a better way to manage radiotherapy-­
induced nausea and vomiting? Int J Radiat Oncol Biol Phys 60(4):1018–1025
20. de Wit R, Aapro M, Blower PR (2005) Is there a pharmacological basis for differences in
5-HT3-receptor antagonist efficacy in refractory patients? Cancer Chemother Pharmacol
56(3):231–238
21. Alidoosti A, Taslimi F, Ameri A, Khosro MS, Miri R, Yavari P (2009) Electrocardiographic
changes after granisetron administration for chemotherapy induced nausea and vomiting. Acta
Med Iran 47(4):289–292
Index

A Doxepin, 64
Acetylcysteine, 66 Dry mouth. See Xerostomia
Acute radiation dermatitis, 6
Amifostine, 8, 61, 84, 85–88, 113, 128, 141,
149, 167 E
Antifungals, 68 Ear toxicity
Ascorbic acid, 8 diagnosis, 49
management, 50
mechanism, 47–48
B risk factors, 48–49
Barrier film, 9 scoring, 49
Benzydamine mouth wash, 64 symptoms, 49
Brain injury. See Radiation brain injury timing, 48
Breast irradiation, 4 Eczema, 7
Enteritis
diagnosis, 147
C management
CAM2028, 66 antidiarrheals, 150
Caphosol (Cytogen Corp), 62–63 antispasmodics, 150
Carmellose sodium paste, 67 dietary modification, 150
Cellulitis, 6 mechanism, 145–146
Chlorhexidine, 62, 89 prevention, 148–149
Common Terminology Criteria for Adverse risk factors, 146–147
Events (CTCAE) v3, 139 scoring, 147–148
Common Terminology Criteria for Adverse symptoms, 147
Events (CTCAE) v4, 23, 147, timing, 146
167, 168 EORTC. See European Organization for
Common Toxicity Criteria for Adverse Effect Research and Treatment of Cancer
(CTCAE) v4.03, 100, 112, 120, 139 (EORTC)
Corticosteroids, 67 Epidermal growth factor receptor (EGFR), 5
Cystitis. See Radiation cystitis Esophagitis
diagnosis, 127
management, 128–129
D mechanism, 125
Deodorant, 11 prevention, 127–128
Dermatitis. See Radiation dermatitis risk factors, 126–127
Dexpanthenol, 10 scoring, 127
Dimensional conformal radiotherapy symptoms, 127
(3D–CRT), 60, 148 timing, 126

© Springer International Publishing AG 2017 213


A. Sourati et al., Acute Side Effects of Radiation Therapy,
DOI 10.1007/978-3-319-55950-6
214 Index

European Organization for Research and I


Treatment of Cancer (EORTC), 31, Intensity-modulated radiation therapy (IMRT),
49, 58, 72, 83–84, 121, 127, 8, 23, 42, 56, 60, 79, 86, 121, 128,
147–148, 166–167 141, 147, 158, 167, 194

F J
Fatigue Juvenile rheumatoid arthritis (JRA), 6
diagnosis, 176–177
management
complementary therapies, 180 K
counseling and education, 179 Keratinocyte growth factor (KGF), 61
exercise, 179
non-pharmacologic interventions, 178
nutrition counseling, 181 L
optimize sleep quality, 179–180 Laryngeal cancer, 3
pharmacologic intervention, 181–182 Laryngeal edema
psychosocial interventions, 180–181 management, 107
mechanism, 173–174 mechanism, 105
risk factors, 174–175 prevention, 107
scoring, 176–177 risk factors, 106
symptoms, 175–176 scoring, 106
timing, 174 symptoms, 106
Free-radical production, 1–2 timing, 105
Laughter therapy, 11
Loss of taste
G diagnosis, 99
Gabapentin, 67 management, 100–101
Gastritis. See Radiation gastritis mechanism, 97–98
Gelclair, 66 prevention, 100
Glutamine, 60, 128, 149 risk factors, 99
Granulocyte-macrophage colony-stimulating scoring, 100
factor (GM-CSF), 61 symptoms, 99
timing, 98
Low-level laser therapy, 8
H
Hair loss
management, 24 M
mechanism, 22 Magic mouthwash, 65
prevention, 23 Mammalian target of rapamycin (mTOR), 63
scoring, 23 Marigold, 60
timing MAS065D, 9
risk factors, 22 Mothers against decapentaplegic homolog 7
symptoms, 22–23 (SMAD7), 63
Hematological side effects
mechanism, 191–192
prevention, 194 N
risk factors, 193 Nasopharyngeal cancer, 3
symptoms, 193–194 National Cancer Institute Common
timing, 192–193 Terminology Criteria for
treatment Adverse Events (NCI-CTCAE
anemia, 198–199 grading), 4, 58
neutropenia, 195–197 Natural honey, 60–61
thrombocytopenia, 197–198 Nonsteroidal anti-inflammatory drugs
Hyaluronic acid, 9 (NSAIDs), 44
Index 215

O timing, 118
Opiates, 66–67 treatment, 122
Oral mucositis Phenytoin mouthwash, 68
management Pilocarpine, 62, 87, 88, 90, 91
antivirals, 68–69 Proctitis. See Radiation proctitis
feeding tube/nutritional support, 69 Prostaglandins, 62
mouth care, 64 Provitamin B5, 10
oral pain, 64–68
patient assessment, 63–64
mechanism, 53–54 R
prevention Radiation brain injury
prophylactic interventions, 59–60 dexamethasone prescription, 32–33
prophylactic intervention under diagnosis, 30–31
evaluation, 60–63 mechanism, 28
risk factors prevention and management, 32–33
concomitant systemic therapy, 55–56 risk factors
patient-related factors, 56–57 patient-related factor, 29
radiation fractionation schedule, 56 treatment-related factors, 28–29
tumor site, 55 scoring, 31
scoring, 58–59 symptoms, 29–30
symptoms, 57–58 timing, 28
timing, 54 Radiation cystitis
Oral pain management diagnosis, 157
acetylcysteine, 66 mechanism, 155–156
antifungals, 68 prevention
benzydamine mouth wash, 64 alpha-1 blocker, 159
CAM2028, 66 analgesics, 160
carmellose sodium paste, 67 anticholinergics, 159
corticosteroids, 67 bladder sparing, 158
doxepin, 64 intravesical GAG, 160
gabapentin, 67 intravesical instillation, 158–159
gelclair, 66 phenazopyridine, 160
low-level laser therapy, 68 risk factors, 156–157
magic mouthwash, 65 scoring, 157
MF 5232, 66 symptoms, 157
opiates, 66–67 timing, 156
phenytoin mouthwash, 68 Radiation dermatitis
sucralfate, 68 diagnosis, 6–7
Oral zinc, 8 management
Orbital radiation therapy, 39. See also grade 1 dermatitis, 11–12
Radiation orbital toxicity grade 2-3 dermatitis, 12–13
Orbital toxicity. See Radiation orbital toxicity grade 4 dermatitis, 13
Oxygen nebulization, 63 mechanism, 1–2
prevention
Aloe vera, 10
P amifostine, 8
Payayor, 60 ascorbic acid, 8
Pentoxifylline, 10 barrier film, 9
Pericarditis Calendula, 10
diagnosis, 119–120 deodorant, 11
mechanism, 118 dexpanthenol (provitamin B5), 10
prevention, 121 general measures, 7–8
risk factors, 118 hyaluronic acid, 9
scoring, 120–121 intensity-modulated radiation therapy
symptoms, 119 (IMRT), 8
216 Index

Radiation dermatitis (cont.) Radiation pneumonitis (RP)


laughter therapy, 11 diagnosis, 112
low-level laser therapy, 8 mechanism, 109
MAS065D, 9 prevention, 113–114
oral zinc, 8 risk factors
pentoxifylline, 10 chemotherapy/hormone therapy, 111
silver leaf dressing, 9 fractionation schedule, 110–111
soft dressing, 9 smoking, 111
theta cream, 10 volume and dose parameters, 110
topical sucralfate, 10 scoring, 112–113
trolamine, 9 symptoms, 111
risk factors timing, 110
patient-related factors, 5–6 Radiation proctitis
treatment-related factors, 4–5 management, 168–169
scoring, 4–5 mechanism, 165
sign and symptoms, 3–4 prevention, 167–168
timing, 2–3 scoring, 166–167
Radiation gastritis symptoms, 166
diagnosis, 135 timing, 166
management, 135–136 Radiation Therapy Oncology Group (RTOG),
mechanism, 133–134 4, 31, 49, 58, 83, 106, 112, 113,
prevention, 135 120, 121, 125, 127, 147, 148, 157,
risk factors, 134 166–167
symptoms, 135 Radiotherapy-induced fatigue. See Fatigue
timing, 134–135 Rectum. See Radiation proctitis
Radiation-induced liver disease (RILD) RILD. See Radiation-induced liver disease
diagnosis, 140 (RILD)
management, 141–142 RINV. See Radiation-induced nausea and
mechanism, 137–138 vomiting (RINV)
prevention, 140–141 RK-0202 (RxKinetix), 62
risk factors, 138 Royal jelly, 61
scoring, 139 RP. See Radiation pneumonitis (RP)
symptoms, 139 RTOG. See Radiation Therapy Oncology
timing, 138 Group (RTOG)
Radiation-induced nausea and vomiting
(RINV)
management, 209–211 S
mechanism, 207–208 Sensorineural hearing loss (SNHL), 47–48.
prevention, 209–211 See also Ear toxicity
risk factors, 208 Silver leaf dressing, 9
symptoms, 208 Soft dressing, 9
timing, 208 Steroids, 62
Radiation orbital toxicity Sucralfate, 68
blepharitis, 40 Systemic lupus erythematosus (SLE), 6
conjunctivitis, 40–41 Systemic sclerosis, 6
corneal toxicity
analgesics, 44
ophthalmologist consult, 44 T
topical lubricants, 43–44 Taste buds, 97. See also Loss of taste
treatment, 43–44 Taste sensitivity, 97, 99
eyelash loss, 40 Theta cream, 10
eyelid dermatitis, 40 Topical sucralfate, 10
iris toxicity, 44 Trolamine, 9
xerophthalmia, 41–43 Two-dimensional radiotherapy (2DRT), 60
Index 217

V dental care, 89
Vasoconstrictor, 62 drink adequate fluids, 89
drugs, 90–91
dry lips, protection
W from, 89
Wobe-Mugos, 63 nocturnal symptoms, 89
oral hygiene, 89
saliva supplementation, 90
X supportive care, 88–89
Xerostomia mechanism, 79–81
amifostine, 85 prevention, 84–85
contraindications risk factors, 81–82
amifostine, adverse effects of, 85–88 scoring, 82–83
pilocarpine, 88 symptoms, 82
diagnosis, 82 timing, 81
management
acupuncture, 90
adequate nutrition, 89 Z
chewing, 89 Zinc, 60

You might also like