Imaging in Geriatrics 2023
Imaging in Geriatrics 2023
Giuseppe Guglielmi
Mario Maas Editors
Imaging in
Geriatrics
Practical Issues in Geriatrics
Series Editor
Stefania Maggi, Aging Branch
CNR-Neuroscience Institute, Padua, Italy
AL GRAWANY
This practically oriented series presents state of the art knowledge on the principal
diseases encountered in older persons and addresses all aspects of management,
including current multidisciplinary diagnostic and therapeutic approaches. It is
intended as an educational tool that will enhance the everyday clinical practice of
both young geriatricians and residents and also assist other specialists who deal
with aged patients. Each volume is designed to provide comprehensive information
on the topic that it covers, and whenever appropriate the text is complemented by
additional material of high educational and practical value, including informative
video-clips, standardized diagnostic flow charts and descriptive clinical cases.
Practical Issues in Geriatrics will be of value to the scientific and professional
community worldwide, improving understanding of the many clinical and social
issues in Geriatrics and assisting in the delivery of optimal clinical care.
Giuseppe Guglielmi • Mario Maas
Editors
Imaging in Geriatrics
AL GRAWANY
Editors
Giuseppe Guglielmi Mario Maas
Clinical and Experimental Medicine Department of Radiology and Nuclear
University of Foggia Medicine
Foggia, Italy University of Amsterdam
Amsterdam, Noord-Holland
The Netherlands
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Switzerland AG 2023
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and trans-
mission or information 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, expressed 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.
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface
Demographic changes, due to the longer lifespan and the improvement of the qual-
ity of life, led to an aging society, where developing high-quality healthcare for
older people becomes increasingly important.
Nowadays the elderly population is more informed and more demanding of bet-
ter care through cutting-edge technology and treatment. Specifically, radiologists
play an increasingly important role and occupy a frontline position in the evaluation
of this cohort of patients, who necessitate definitive imaging.
In this perspective, the complex relationship between geriatrics and radiology
must be redefined. Imaging in Geriatrics is the result of a teamwork of radiologists,
experts in the field, with the aim to provide guidance for the appropriate use of
imaging in modern geriatric care, describing how to recognize pathology from para-
physiological findings in the elderly population.
The study of diagnostic imaging in geriatrics, enriched by traditional and modern
imaging methods, is primarily oriented to the clinical and radiological analysis most
frequently encountered in these patients and, above all, to the not easy distinction
between “normal” and “pathological,” especially in situations in which para-physi-
ological changes resulting from aging processes are associated with alterations
related to comorbidity and chronicity.
This volume includes a multidisciplinary approach, and it covers all major medi-
cal issues related to aging, divided by apparatus. In particular, it encloses the main
pathologies in the neurological, cardiovascular, pulmonary, gastrointestinal, uro-
genital, hematologic, and musculoskeletal field.
This book considers all imaging techniques that have been the cornerstones of
radiology, but also modern innovations. Conventional radiography is still the first
approach in the diagnosis of a frequent variety of pathological conditions of elderly
patients, such as fractures, often due to osteoporosis, pneumothorax, or heart fail-
ure. On the other hand, Computer Tomography (CT), with its intrinsic resolution
power, allows radiologists to detect a possible ischemic or hemorrhagic cerebral
focus, as well as neoplastic metastases for the staging of the primary pathology.
Spinal cord injuries are best identified with Magnetic Resonance Imaging (MRI).
But nowadays, and therefore modern “tailored” medicine, is changing, going
towards Artificial Intelligence (AI), a technological evolution that brings with it the
limits related to the training and updating of the healthcare personnel involved. For
AL GRAWANY
vi Preface
this reason, a section about the role of AI in the management of geriatric patients
could not be missed at the end of this volume.
Finally, the aim of this book is to simplify the approach and the diagnostic imag-
ing process of geriatric diseases, bringing out the potential and limitations of each
imaging technique.
We recommend the reading of this book not only to radiologists for their daily
clinical practice but also to all physicians who require a basic knowledge of imaging
concerning the main geriatric pathologies, because of their complex clinical
presentations.
1
Imaging Techniques in Geriatric Patients ���������������������������������������������� 1
Caterina Bernetti, Carlo Augusto Mallio, Rosario Francesco Grasso,
and Bruno Beomonte Zobel
2
Neurodegenerative Diseases in Geriatric Patients���������������������������������� 11
Camilla Russo, Rossana Senese, and Mario Muto
3
Neurovascular Emergencies in Geriatric Patients���������������������������������� 37
Giuseppe Maria Di Lella, Luca Ausili Cefaro,
and Cesare Colosimo
4
Head and Neck in Geriatric Patients ������������������������������������������������������ 73
T. Popolizio, L. Cassano, A. Pennelli, R. Izzo, G. Fascia,
M. Masciavè, and Giuseppe Guglielmi
5
Heart Diseases in Geriatric Patients�������������������������������������������������������� 109
Anna Palmisano, Raffaele Ascione, Francesco De Cobelli,
and Antonio Esposito
6
Vascular Diseases in Geriatric Patients��������������������������������������������������� 137
Gloria Caredda, Giuseppe Guglielmi, and Luca Saba
7
Airway Diseases in Geriatric Patients������������������������������������������������������ 151
Maurizio Balbi, Roberta Eufrasia Ledda, Silvia Pamparino,
Gianluca Milanese, Mario Silva, and Nicola Sverzellati
8
Neoplastic Diseases of the Respiratory System in Geriatric Patients �� 171
Zeno Falaschi, Francesco Filippone, Sergio Pansini, Stefano Tricca,
Paola Basile, Sara Cesano, and Alessandro Carriero
9
The Gastrointestinal System in Geriatric Patients �������������������������������� 217
Damiano Caruso, Domenico De Santis, Francesco Pucciarelli,
and Andrea Laghi
10
The Male Urogenital System in Geriatric Patients �������������������������������� 235
Emilio Quaia and Filippo Crimí
vii
AL GRAWANY
viii Contents
11
The Female Urogenital System in Geriatric Patients ���������������������������� 271
Maria Assunta Cova, Lorella Bottaro, Cristina Marrocchio,
and Alessandro Marco Bozzato
12
Osteoarthritis in Axial Skeleton in Geriatric Patients���������������������������� 319
Francesca Serpi, Salvatore Gitto, and Luca Maria Sconfienza
13
Osteoarthritis in Appendicular Skeleton in Geriatric Patients ������������ 345
Antonio Barile, Riccardo Monti, Federico Bruno, Julia Daffinà,
Francesco Arrigoni, and Carlo Masciocchi
14
Metabolic Bone Disease in Geriatric Patients ���������������������������������������� 367
Maria Pilar Aparisi Gómez, Francisco Aparisi, Giuseppe Guglielmi,
and Alberto Bazzocchi
15
Body Composition in Geriatric Patients�������������������������������������������������� 397
Maria Pilar Aparisi Gómez, Francisco Aparisi, Giuseppe Guglielmi,
and Alberto Bazzocchi
16
Myeloid and Lymphoid Disorders in Geriatric Patients������������������������ 427
Patrizia Toia, Massimo Galia, Giuseppe Filorizzo,
Ludovico La Grutta, Federico Midiri, Pierpaolo Alongi,
Emanuele Grassedonio, and Massimo Midiri
17 The Role of Artificial Intelligence (AI) in the Management
of Geriatric Patients���������������������������������������������������������������������������������� 445
Salvatore Claudio Fanni, Sherif Mohsen Shalaby,
and Emanuele Neri
Imaging Techniques in Geriatric Patients
1
Caterina Bernetti, Carlo Augusto Mallio,
Rosario Francesco Grasso, and Bruno Beomonte Zobel
In the last decades, there has been a rising trend toward global population aging,
especially in developed nations. This longevity revolution is happening faster than
historical precedents and a further increase of median age is expected in subsequent
years [1].
The improvement of living conditions and the advent of modern medicine carry-
ing innovations in treatment and prevention are some of the main reasons of
increased life expectancy.
This demographic change is determining a profound impact on society, in par-
ticular with regard to the healthcare system, that now has to face more age-related
diseases [2, 3].
In particular, people over 65 years, defined by convention as elderly, are more
prone to develop multiple chronic conditions, such as cardiovascular diseases,
dementia, cancer, and also osteoporosis that could lead to fragility fractures after
minor traumas [4].
In this population, there is an intrinsic difficulty in distinguishing the boundary
between para-physiologic modifications determined by aging and real pathologic
conditions. Furthermore, given the possibility of an incomplete medical history, the
complexity of symptoms and signs determined by the coexistence of multiple
comorbidities, and the reduced sensitivity of laboratory tests, the diagnosis is often
difficult to achieve. In this context, radiological imaging plays an important role in
the diagnostic workup of elderly patients [2, 5].
AL GRAWANY
2 C. Bernetti et al.
In the elderly, even more than the rest of the population, the first medical contact
is often represented by the general practitioner, who necessarily needs to be
equipped with adequate knowledge in order to suggest the appropriate specialist
referral or radiological examination [6]. Whereas, in a hospital setting, the two main
figures who will have to interface are the geriatrician and the radiologist, in associa-
tion with all the other specialists who may be involved. In order to provide high-
quality care, they must cooperate in the choice of the appropriate examination and
in the correct management of the patient, with medical and paramedical staff trained
with age-specific skills [7, 8]. Empathy is one of the main features that the health-
care personnel caring for the elderly patient should own [9].
The imaging examination must be chosen wisely, must be of clinical utility, and
must help modify patients management, without exposing them to unnecessary
stress or risks; in fact patients’ safety must be considered as the priority [10].
The goals of the exam are an improvement in the quality of life, the preservation
of functionality of these frail patients, and the prevention of progression from dis-
ease to disability [3].
In the decision process to find the appropriate image technique, the clinicians
must integrate several information, such as functional and mental status, and also
comorbidities, for instance chronic renal impairment, cardiac disfunction, poor
peripheral venous access, and dementia. Immobility and respiratory problems that
impair breath holds must be taken into consideration too, because could alter image
quality [3].
Handling elderly patients in the radiology department is burden by many diffi-
culties; this population need to be treated with caution and special care as their
conditions easily deteriorate. Logistical issues are one of the main challenges that
need to be addressed, in fact these patients often have mobility problems, need
supervision while being transferred to the radiology department and assistance
while waiting to perform the exam [2].
Proper positioning is more time-consuming and the maneuvering of the patient
needs more qualified trained staff [2]. Elders usually have decreased agility and
strength; hence, movements could be impaired, sense of balance could be altered,
and some patient could be unable to maintain required positions. In order to move
them safely and position them adequately, their cooperation and compliance need to
be elicited if possible. Specific movers and support devices are useful to position
and immobilize them. Attention need to be paid while having more independent
patients move or even walk, because they could misjudge distances [3]. The unfa-
miliar and cold environment of radiology rooms and equipment could frighten this
fragile patients; blankets are used to offer adequate warmth, privacy, and dignity.
Healthcare providers must never leave the patient alone; moreover, they need to
be aware of any sign of pain or discomfort, because in the elderly, sensation of pain
could be altered, and the patients could also have trouble expressing themselves [3].
Communication abilities of geriatric patients are often impaired because of many
conditions, such as reduced acuity of vision and hearing, depression, or dementia
[4]. Elders can have difficulties in understanding the procedures and in complying
with instructions to remain still. Speaking clearly, using gestures, beepers, and
1 Imaging Techniques in Geriatric Patients 3
Thanks to its availability and low costs, plain radiography often represents the ini-
tial tool for the diagnosis of many pathologic conditions, including pulmonary, car-
diovascular, and musculoskeletal disorders in older adults. This technique offers as
advantages low costs, celerity in execution, and with modern equipment low radia-
tion exposure.
Correct positioning of the patient is essential to obtain quality images; however,
it could require particular devices, to assure adequate posture and immobilization.
Due to mental impairment or hearing loss, elderly patients often have difficulties
hearing instruction and are not able to collaborate [9]. Short times reduce risk of
involuntary and voluntary motion, more common in the geriatric patients, during
the acquisition [12].
In order to avoid transportation from one department to another, mobile X-ray
equipment with digital panels and radiolucent beds and gurneys can be used in the
hospital setting.
Chest radiography (CXR) is the initial test for the diagnosis of pulmonary, pleu-
ral, and cardiovascular diseases [12]. Radiographs are usually performed in a
posterior-anterior and a perpendicular lateral projection; however, due to mobility
issues of elderly patients, standard projections often cannot be performed. Hence, a
supine or semi-supine position radiograph has to be taken with some limitations,
such as projective magnification. For lung diseases, such as pneumonia, fibrosis and
lung cancer, sensitivity, and specificity of CXR are quite high. As previously said,
in the elderly, there is a difficulty in recognizing pathologic versus age-related alter-
ations, such as fibrotic changes, emphysema, airways, and rib cage calcification.
Besides this problem, it is also arduous to discern overlapping pathologies in
patients with multiple comorbidities; for example in the case of heart failure versus
AL GRAWANY
4 C. Bernetti et al.
AL GRAWANY
6 C. Bernetti et al.
However, there are some issues that need to be underlined. US images are
scarcely reproducible, because it is a patient and operator dependent technique,
which relies on the compliance of the patient and the skills of the radiologist.
Moreover, availability of portable equipment can be limited [21].
Elderly patients are characterized by numerous age-related conditions that could
affect the quality, and hence, the interpretation of the images. For instance, their
tissues and vascular vectors are characterized by fibrosis and calcifications, which
determine an increased sonography echogenicity and compound artifacts [10]. This
cohort of patients is also characterized by reduced bowel movements with a conse-
quent increased amount of gas in the lumen of the intestine, which could lead to
difficulty in the evaluation of abdominal organs, such as the pancreas [10].
Doppler analysis represents a useful feature that allows to evaluate the blood
flow. It can be used in case of suspected deep vein thrombosis (DVP) or to evaluate
atherosclerotic plaques of carotid, iliac, femoral, popliteal, and tibial arteries, often
present in older adults, but also to evaluate blood supply of tumors. However,
motion, in particular respiratory movements, often uncontrollable in the elderly,
could alter the visualization of the color maps.
Contrast ultrasound (CEUS) is performed with an intravenous contrast agent
consisting of gaseous microbubbles that remain in the vascular bed and do not reach
the extravascular spaces. Due to the absence of nephrotoxicity and the possibility of
being repeated, CEUS could be proposed as a valid and safe alternative to CT and
MRI with contrast; however, it must be performed by experienced operators in order
to avoid misinterpretations [22, 23].
In addition to the most common studies (abdomen and thyroid gland), ultrasound
can be used for many conditions, for instance musculoskeletal disorders, which are
considered one of the major chronic conditions affecting the senile population.
Vessel evaluation can be performed with color-Doppler, for instance to predict car-
diovascular and neurological events measuring intima-media thickness of the
carotid artery or to follow-up aneurism of the abdominal aorta [23].
This technique, however, uses higher radiation doses than plain radiographs, but
in this age group the long-term risk of cancer from ionizing radiation is of diminish-
ing concern [25].
CT allows to study in-depth findings already detected on radiography or ultra-
sound, but can also be used directly in case of an important clinical suspicion that
requires a more targeted investigation.
Even though some studies can be performed without the use of contrast media, for
instance for small lung nodules, ureteral stones, or brain after trauma, to improve the
diagnostic evaluation of some pathological conditions, the use of iodinated contrast
agents is mandatory. However, in the elderly, intravascular iodine contrast media
should be used with caution and limited to the indications where it is strictly neces-
sary, in order to reduce the risk of contrast medium-induced nephropathy (CIN) to
which they are more exposed [26]. Geriatric patients usually have a background
reduced kidney function due to age-related functional alterations and tend to be
affected by multiple comorbidities, such as heart insufficiency, hypovolemia, diabe-
tes, and hypertension that could contribute to renal impairment. Moreover, poten-
tially nephrotoxic drugs are used in older adults [27].
When administering iodinated contrast media, the value of serum creatinine must
be evaluated and according to age-appropriate guidelines for contrast agents adminis-
tration, the glomerular filtration (eGFR) rate needs to be greater than 30 mg/mL [25].
Some precautions can be put into practice to reduce the risk of CIN, such as
reduction of quantity of contrast media injected, modifications in kV settings, avoid-
ance of nephrotoxic medications, pre- and post-CT hydration, and in some cases a
sessions of dialysis need to be programmed [26].
Moreover, elderly patients usually have poor peripheral venous access due to frag-
ile vessels; hence, often only small cannulas can be positioned, making them more
susceptible to contrast agent extravasation. Establishing secure intravenous access and
performing a saline test injection may reduce the risk of extravasation [25].
One of the main uses of CT in elderly patients is lung CT, which is able to iden-
tify, even with low-dose protocols, the presence of nodules, masses, or pneumonia
[28]. Coronary CT angiography in the senile population is now considered a useful
diagnostic tool for coronary assessment and risk stratification [29]. A full-body CT
with contrast is often performed for tumor staging, because it offers the possibility
to evaluate lymph node stations and the presence of eventual metastases, in order to
adequately plan the therapeutic approach. Finally, non-contrast brain CT is one of
the main tools used in the elderly, in particular in the emergency setting for stroke
or after trauma [30].
MRI is an imaging technique that exploits magnetic fields and radio waves. The
main advantages are the absence of ionizing radiation, the high contrast resolution,
and the direct multiplanar imaging, which make this technique suitable for a wide
range of applications in medical diagnosis [24].
AL GRAWANY
8 C. Bernetti et al.
Due to the long acquisition times, the need to remain still, and the noisy ambient,
MRI is a difficult imaging technique for elderly patients to tolerate. Moreover, in
case of fragile patients that could require monitoring and, in some conditions, life
support equipment, a MRI room, compared to a CT room is a more difficult envi-
ronment to manage [24].
MRI is also burden by numerous contraindications, many of which are present in
elderly patients, such as claustrophobia, cardiac pacemakers or old ferromagnetic
surgical material. Low-field MRI equipment could be used, at the expense of the
quality of the exam, for patients with claustrophobia or mental health problems,
which would require the use of sedation to tolerate the positioning necessary to
perform high-field MRI [2].
In case of the presence of ferromagnetic material, CT or US must be proposed in
lieu of MRI.
The arrangement of the patient on the scanning table must be as comfortable as
possible; in fact this population is more exposed to develop pressure ulcer if posi-
tioned for long on hard surfaces. Hence, when performing MRI in an older popula-
tion, abbreviated, time-efficient MRI protocol that maintains high diagnostic
accuracy could be useful in order to avoid excessive examination times that are
difficult to bare and could lead to motion and breathing artifacts, but also to avoid
the abovementioned complications.
Limited rapid access to MRI is another limitation that must take into consider-
ation when choosing the right examination for elderly patients.
Even though contrast agents used in MRI are less invasive than the ones utilized
for CT, it remains the risk of nephrogenic systemic fibrosis in patients with poor
renal function, such as elderly. However, in some cases it is a necessary tool, for
instance in the evaluation of neoplasms.
Aging is related to neurodegenerative diseases, dementia, and also stroke, which
represent the main cause of disability and the second cause of death, globally, deter-
mining a high financial and social burden on the healthcare system and society [31].
One of the main use of MRI in the elderly is the study of brain ischemic events,
but it is used also to indagate occult fractures and lesions of the spine [31].
In the elderly, surgical treatments are a burden because of higher risk of morbidity
and mortality. Interventional radiology offers a less invasive and safer option for the
diagnostic and therapeutic management of many pathologic conditions in this popu-
lation. Another advantage of IR procedures is that it does not require general anes-
thesia, in fact a local approach with eventual deep sedation is often sufficient [5].
The main complications that might occur include infection and bleeding. The imag-
ing methods most used as a guidance for interventional procedures are fluoroscopy,
ultrasonography, and CT, the choice of which is made according to the anatomical
site and type of procedure.
1 Imaging Techniques in Geriatric Patients 9
References
1. Fawcett R, McCoubrie P. Pitfalls in imaging the frail elderly. Br J Radiol. 2015;88:20140699.
2. Gossner J, Nau R. Geriatric chest imaging: when and how to image the elderly lung, age-
related changes, and common pathologies. Radiol Res Pract. 2013;2013:584793. https://doi.
org/10.1155/2013/584793.
3. Barba BE, Barba JR, Rankin C. Caring for older adults in the radiology department. Are you
prepared? J Radiol Nurs. 2007;26:11–4.
4. Jaul E, Barron J. Age-related diseases and clinical and public health implications for the 85
years old and over population. Front Public Health. 2017;5:335.
5. O’Brien J, Baerlocher MO, Asch M, Myers A. Role of radiology in geriatric care: a primer for
family physicians. Can Fam Physician. 2009;55:32–7.
6. Radiology and primary care in Europe. Insights Imaging. 2010;1:46–52.
7. Chang A, Singh N, Boyd L, Lawson C. Strategies to improve radiographic practices for patients
with Alzheimer’s disease: a systematic review. J Med Imaging Radiat Sci. 2016;47:362–6.
8. Sellier N, Lhoste G, Lemouchi D, Sebbane G, Seror O. [Current problems in geriatrics and
radiology. Teleradiology: organization and evaluation]. J Radiol. 2003;84:1907–12.
9. Faletti C, Borrè A, Tabasso MD. Diagnostic algorithm in the elderly. In: Guglielmi G, Peh
WCG, Guermazi A, editors. Geriatric imaging. Berlin: Springer; 2013. p. 21–51.
10. Chang W-H, Huang C-H, Chien D-K, Huang M-Y, Tsai W, Chang K-S, Tsai C-H. Emergency
sonography for the elderly. Int J Gerontol. 2011;5:1–8.
11. Monfardini L, Vecchi V. Limitations of diagnostic radiology for frail and vulnerable elderly
cancer patients. Aging Health. 2013;9:283–5.
12. Kessler R, Dean AJ. Plain radiography in the elderly. In: Fox JC, editor. Clinical emergency
radiology. 2nd ed. Cambridge: Cambridge University Press; 2017. p. 178–94.
13. DeBlieux P, Mills L. Chest radiograph. In: Fox JC, editor. Clinical emergency radiology. 2nd
ed. Cambridge: Cambridge University Press; 2017. p. 41–54.
14. Vanrusselt J, Vansevenant M, Vanderschueren G, Vanhoenacker F. Postoperative radiograph
of the hip arthroplasty: what the radiologist should know. Insights Imaging. 2015;6:591–600.
15. Krestan C, Hojreh A. Imaging of insufficiency fractures. Eur J Radiol. 2009;71:398–405.
16. Heilmeier U, Youm J, Torabi S, Link TM. Osteoporosis imaging in the geriatric patient. Curr
Radiol Rep. 2016;4:18.
17. Acute abdominal pain in the elderly. Contemp Diagn Radiol. 2017;40:8.
18. Dean AJ, Kessler R. Plain film evaluation of the abdomen. In: Fox JC, editor. Clinical emer-
gency radiology. 2nd ed. Cambridge: Cambridge University Press; 2017. p. 55–78.
19. Melton LJ, Crowson CS, O’Fallon WM. Fracture incidence in Olmsted County, Minnesota:
comparison of urban with rural rates and changes in urban rates over time. Osteoporos Int.
1999;9:29–37.
20. Can B, Kara M, Kara Ö, Ülger Z, Frontera WR, Özçakar L. The value of musculoskeletal
ultrasound in geriatric care and rehabilitation. Int J Rehabil Res. 2017;40:285–96.
21. Ticinesi A, Scarlata S, Nouvenne A, Lauretani F, Incalzi RA, Ungar A, GRETA (Gruppo di
Ricerca sull’Ecografia Toracica nell’Anziano) Group of the Italian Society of Gerontology
and Geriatrics (SIGG). The geriatric patient: the ideal one for chest ultrasonography? A review
from the chest ultrasound in the Elderly Study Group (GRETA) of the Italian Society of
Gerontology and Geriatrics (SIGG). J Am Med Dir Assoc. 2020;21:447–54.e6.
AL GRAWANY
10 C. Bernetti et al.
22. Dietrich CF, Tana C, Caraiani C, Dong Y. Contrast enhanced ultrasound (CEUS) imaging of
solid benign focal liver lesions. Expert Rev Gastroenterol Hepatol. 2018;12:479–89.
23. Recinella G, Marasco G, Tufoni M, Brizi M, Evangelisti E, Maestri L, Fusconi M, Calogero P,
Magalotti D, Zoli M. Clinical role of lung ultrasound for the diagnosis and prognosis of coro-
navirus disease pneumonia in elderly patients: a pivotal study. GER. 2021;67:78–86.
24. Leone A, Balanika A, Cerase A. Infection and miscellaneous. In: Guglielmi G, Peh WCG,
Guermazi A, editors. Geriatric imaging. Berlin: Springer; 2013. p. 141–75.
25. Sadro CT, Sandstrom CK, Verma N, Gunn ML. Geriatric trauma: a radiologist’s guide to imag-
ing trauma patients aged 65 years and older. Radiographics. 2015;35:1263–85.
26. Mehran R, Aymong ED, Nikolsky E, et al. A simple risk score for prediction of contrast-
induced nephropathy after percutaneous coronary intervention: development and initial valida-
tion. J Am Coll Cardiol. 2004;44:1393–9.
27. Wilhelm-Leen E, Montez-Rath ME, Chertow G. Estimating the risk of radiocontrast-associated
nephropathy. J Am Soc Nephrol. 2017;28:653–9.
28. Park JE, Kim Y, Lee SW, Shim SS, Lee JK, Lee JH. The usefulness of low-dose CT scan in
elderly patients with suspected acute lower respiratory infection in the emergency room. Br J
Radiol. 2016;89:20150654.
29. Nijveldt R, Pflederer T, Achenbach S. Coronary CT angiography in the elderly. Neth Heart
J. 2014;22:124–5.
30. Bekelis K, Fisher ES, Labropoulos N, Zhou W, Skinner J. Variations in the intensive use of head
computed tomography for elderly hemorrhagic stroke patients. Radiology. 2015;275:188–95.
31. Cole JH, Franke K. Predicting age using neuroimaging: innovative brain ageing biomarkers.
Trends Neurosci. 2017;40:681–90.
Neurodegenerative Diseases in Geriatric
Patients 2
Camilla Russo, Rossana Senese, and Mario Muto
Abbreviations
C. Russo
Diagnostic and Interventional Neuroradiology, “A. Cardarelli” Hospital, Naples, Italy
Department of Electrical Engineering and Information Technology, Università degli Studi di
Napoli “Federico II”, Naples, Italy
R. Senese
Emicenter European Medical Imaging, Casavatore, Naples, Italy
M. Muto (*)
Diagnostic and Interventional Neuroradiology, “A. Cardarelli” Hospital, Naples, Italy
AL GRAWANY
12 C. Russo et al.
2.1 Introduction
people worldwide, a figure that is expected to double in the next decades due to
population ageing and increasing average life expectancy. Several risk factors have
been called into question for dementia, including sex, ethnicity, educational level,
smoking, drug and alcohol abuse; however, age and genetic factors seem to play a
major role in disease onset and progression [1]. From a clinical perspective, patients
suffering from a neurodegenerative disorder generally realize when their symptoms
began. From here onwards the pattern of cognitive decline is variable, with a period
of relative stability preceding an abrupt and inexorable deterioration. Clinical evolu-
tion speed strictly depends on the putative aetiological mechanism, ranging from
few months in the most aggressive cases to several years in the slowly progressing
diseases [2, 3]. In demented patients, it is supposed to be a certain discrepancy
between actual disease onset and symptoms onset; indeed, due to cellular redun-
dancy in neuronal circuits, symptoms only appear when the number of neurons
spared by neurodegenerative processes is lower than required to maintain a normal
neuronal activity in the affected pathway. Moreover, assuming that the rate of neu-
ronal loss is almost constant during the whole disease course, the observed sudden
clinical deterioration in late-stage dementia is probably due to a collapse in neuron
number beneath a certain threshold [2]. Despite symptom severity, life expectancy
in affected patients is not significantly reduced, unless in case of comorbidities or
direct/indirect involvement of neurological structures responsible for vital functions
(i.e. brainstem centres regulating heart rate, breathing and blood pressure).
The presence of an underlying neurodegenerative process responsible for cogni-
tive decline defines primary dementias. However, severe cognitive impairment is
not only observed in neurodegenerative disorders but also common in vascular,
toxic, paraneoplastic, infectious, metabolic or traumatic insults of the central ner-
vous system (CNS); in these cases, the definition of secondary or potentially revers-
ible dementias may be applied, as some of these conditions may be treatable and
related symptoms partially reversible [4]. Frequently, post-mortem brain examina-
tion represents the only tool for definite differential diagnosis between dementia
types; nevertheless, structural magnetic resonance imaging (MRI) as well as nuclear
medicine (NM) can provide important clues in excluding secondary causes and
hypothesizing the presence of a specific pathogenic mechanism.
This chapter provides a comprehensive overview of the most common neurode-
generative causes of dementia in elderly, classified on the basis of the presumed
underlying pathologic mechanism, focusing on clinical patterns and corresponding
neuroradiological manifestations.
a b
Temporal Lobe
Corteccia Ippocampi
Corteccia frontale Corteccia parietale
Percentile: 0.1 temporale
15.30
Temporal Lobe (% of ICV)
Corteccia frontale
Age (years) 20 25 30 35 40 45 20 25 30 35 40 45
Corteccia temporale Ippocampi
200 99 perc.
14
180 99 perc. 50 perc.
12 10 perc.
160 50 perc. 99 perc.
10 perc. 10 1 perc.
140 1 perc. 50 perc.
10 perc.
8 1 perc.
120
100 Età 6 Età
20 25 30 35 40 45 20 25 30 35 40 45 Corteccia temporale
Iperintensita in FLAIR
Volume 6,25 ml
* I Volumic cerebrali visualizzati sono normalizzati in base alle dimensioni dell testa. II fattore di correzione per questo paziente è 0,71
Fig. 2.2 An example of automated volume reports from two different commercial software, with
reference to age/sex-matched control population and with cortical atrophy patterns: (a) Icobrain
DM report for MRI (icometrix®) and (b) Quantib Neurodegenerative (Quantib® Brain)
16 C. Russo et al.
and neurofibrillary tangles [12, 13]. However, at present NM is only indicated for
those patients with significant diagnostic uncertainty after a comprehensive multi-
domain evaluation, and when a timely diagnosis may affect patient’s manage-
ment [14].
As seen above dementia covers a very wide and varied spectrum of possible disor-
ders, whose early differentiation is increasingly gaining importance especially in
light of new emerging treatments and disease-modifying therapies. In this light,
neuroimaging plays a pivotal role both in terms of research and diagnostic purposes.
Therefore a close communication between referring clinicians and radiologists is
strictly required to ensure the capitalisation of all the information provided by imag-
ing techniques. For such purpose, the structured imaging interpretation then trans-
lated in a shared and standardized language is highly recommended [15]; in
particular the introduction of semi-quantitative visual rating scales (VRS) (which
have largely replaced the use of subjective terms), as well as the development of
several software tools to quantify brain atrophy, is simplifying the communication
among professional involved in the care of demented patients [16]. However, despite
the large use for research purposes, brain atrophy quantification with dedicated
tools is still far from an actual application in daily clinical routine due to its limited
territorial diffusion and its cost, process, and time consumption; conversely, the use
of semi-quantitative VRS is widely adopted both in non-academic and academic
institutes [15, 17]. Several VRS were developed to semi-quantitatively assess the
main imaging features of dementia: brain atrophy, white matter micro-vascular
lesion load and cerebral microbleeds burden. Here we summarize the most relevant
semi-quantitative VRS also suggesting a structured interpretation/reporting scheme
to adopt in daily clinical practice.
AL GRAWANY
2 Neurodegenerative Diseases in Geriatric Patients 17
a b
Fig. 2.3 Scheme of atrophy pattern evaluation by using the global cortical atrophy-frontal (GCA)
score, to be used for each of the individual 13 brain regions assessed separately in each hemi-
sphere; the final score is the sum of all regions. Below, an example of grade 1 (a) and grade 3
(b) atrophy
a b
Fig. 2.4 Scheme of atrophy pattern evaluation by using the medial temporal atrophy (MTA)
score, to assess hippocampal and mesio-temporal atrophy on the coronal plane. Below, an example
of grade 2 (a) and grade 4 (b) atrophy
Structured templates for MCI and dementia assessment are used to provide a check-
list within which all relevant items are included and reported; for the sake of com-
pleteness, other key clinical features should always be included at the beginning of
the structured report to complete the general framework.
2 Neurodegenerative Diseases in Geriatric Patients 19
a b
Fig. 2.5 Scheme of atrophy pattern evaluation by using the Koedam score, to assess parietal lobe
atrophy especially on the sagittal and coronal plane. Below, an example of grade 3 atrophy both on
the sagittal (a) and coronal (b) planes
Suggested Checklist:
• Describe the clinical presentation and state the diagnostic suspicion
• Mention the scan protocol used
• Mention previous imaging examination available for comparison
• Exclude possible mimics (mass lesions, hematomas, and any other surgical or
non-surgical disorder that could explain MCI/dementia)
• Exclude the presence of hydrocephalus (communicating or
non-communicating)
• Describe any diffusion abnormality
• Describe vascular pathology (quantification of WMH and microbleeds burden by
using semi-quantitative scales)
• Describe atrophy pattern (symmetric or asymmetric pattern; supratentorial and/
or infratentorial compartment involvement; quantification by using semi-
quantitative scales)
• Summarize conclusions and final impressions (normal findings according to the
age vs. vascular/neurodegenerative/mixed pathology; pattern consistent with the
clinically suspected dementia disorder; suggest differential diagnosis for
neurodegeneration)
20 C. Russo et al.
a b
Fig. 2.6 Scheme of atrophy pattern evaluation by using the entorhinal cortical atrophy (ERICA)
score, to assess the entorhinal cortex for volume loss. Below, an example of grade 2 atrophy on 3D
T1w (a) and 3D FLAIR (b) images; dotted line indicates the entorhinal cortex, white arrowhead
indicates the enlargement collateral sulcus (with only minimal detachment of the entorhinal cortex
from the cerebellar tentorium, also known as “tentorial cleft sign”)
a b c d
Fig. 2.7 White matter hyperintense (WMH) lesion burden evaluated by using Fazekas’ score on
axial FLAIR MRI. (a) grade 0, (b) grade 1, (c) grade 2, (d) grade 3
2 Neurodegenerative Diseases in Geriatric Patients 21
The overall number of neurodegenerative disorders runs into the hundreds and some
of them overlap clinically or pathologically, making the differential diagnosis quite
challenging. As noted above, two main factors contribute to this phenomenon: on
the one hand a same neurodegenerative disease can affect different areas of the
brain, which leads to a large phenotypical variability specially in early disease
stages; on the other hand some disorders share some common features, thus resem-
bling one another in some clinical manifestations [19, 20]. In terms of classification,
primary neurodegenerative disorders can be classified according to brain abnor-
malities location, clinical manifestations, or pathological mechanism. Following
this last criterion here we give a short overview of typical findings and neuroradio-
logical hallmarks of the most common neurodegenerative disorders in the elderly.
The three most significant groups are represented by:
2.4.1 Tauopathies
thought to be responsible for about 60–80% of all overt dementias, with an age-
specific prevalence that almost doubles every 5 years in subjects aged >65 years.
This exponential increase in AD prevalence in elderly suggests that AD strictly
related to ageing, therefore considered the main risk factor; however, apart from
ageing, other risk factors have also been described (including family history of
dementia, female gender, apolipoprotein E epsilon 4 allele carrier status, smoking
and mutations in the amyloid precursor protein) [22].
As per other forms of dementia, also in AD the neuropathological changes due to
amyloid and tau deposition (leading to the formation of plaques) occur up to
25–30 years before clinical manifestations. From a clinical standpoint we can there-
fore observe a long silent (or pre-clinical) phase, followed by an overt progressive
neurodegenerative disorder (starting when a certain neurotoxicity threshold has
been exceeded). Clinical symptoms include memory loss and MCI in early stages,
then followed by decline in praxis and visuo-spatial abilities, attention deficit and
behavioural changes (with neuropsychiatric symptoms ranging from apathy, depres-
sion and anxiety up to aggressiveness, psychomotor agitation and psychosis). Apart
from AD classical variant initially characterized by anterograde episodic memory
loss, two other clinical (or atypical) variants may be identified: frontal variant AD
(fvAD) and posterior cortical atrophy (PCA). In both cases the same neuropatho-
logical changes of AD can be observed in the brain, however with different spatial
distribution and thus causing different symptoms at onset. In fvAD, the condition
mainly affects frontal lobes, with dominance of behavioural changes, executive dys-
function or a combination of both over a pure memory loss. In PCA early manifesta-
tions are related to visual cortex involvement, with symptoms including difficulty in
recognising faces, measuring distances and perceive the surrounding space; due to
this insidious onset, PCA variant may be difficult to identify and it can take a long
time before considering the proper diagnosis.
Moving to imaging in AD patients, although CT is able to detect atrophy, MRI is
the golden standard to describe the pattern of cortical atrophy and exclude possible
differential diagnoses. Atrophy typically occurs first in hippocampus, perirhinal and
entorhinal cortex, prior to pervasive progression to diffuse cortical atrophy; this
atrophy can be assessed by observing the enlargement of the parahippocampal fis-
sures, or alternatively by using MTA and/or ERICA scores. Medial temporal lobe
atrophy is found both in AD and (to a lesser extent) in other primary neurodegenera-
tive disorders, whereas it is less commonly observed in normal ageing; however,
despite a certain specificity, such volume loss does not appear early in the course of
the disease, thus representing only a late AD manifestation. Posterior cortical atro-
phy or bilateral frontal lobe atrophy are observed in later AD stages coupled to
medial temporal lobe involvement, or in atypical AD variants (PCA and fvAD) as
an early finding coupled to a relative sparing of the medial temporal lobe. In addi-
tion to the above features, WMH due to chronic small vessel disease, cerebral
microbleeds and microinfarcts can also be observed; in these cases, the simultane-
ous vascular and neurodegenerative pathologies double the risk and exacerbate the
symptoms of dementia. Apart from conventional imaging, functional MRI also har-
bour the potential to show a reduced activation in hippocampus and related
2 Neurodegenerative Diseases in Geriatric Patients 23
structures within the medial temporal lobe of AD patients, either during cognitive
paradigms or resting state; similarly, different advanced techniques such as diffu-
sion tensor imaging, quantitative susceptibility mapping and magnetization transfer
have also been used for analysing the damage in the same structures. However at
present these applications have only been exploited for research purposes and used
by limited number of research groups, therefore far from clinical widespread appli-
cations [23].
Structural MRI can be complemented by a variety of NM examinations in case
AD is suspected, both for supporting the diagnosis (even before symptoms onset)
and stratifying the prognosis (in case of overt dementia). SPECT and FDG-PET are
used to detect regional hypoperfusion and hypometabolism, respectively, generally
with a bilateral and symmetric temporo-parietal, precuneus and posterior cingulate
distribution; sensorimotor cortex is generally spared, whereas frontal lobes are usu-
ally involved in later disease stages. Diagnostic specificity can also be increased by
resorting to amyloid-PET and tau-PET; amyloid and tau biding tracers selectively
accumulate in grey matter and medial temporal lobes respectively, with amyloid
deposition occurring before tau deposition. In particular, amyloid binding tracers
are used as a negative predictor of AD, as the absence of captation makes the diag-
nosis unlikely. Conversely tau-binding tracers are not specific for AD, as they are
found positive also in other tauopathies; however, the higher is the accumulation
within hippocampus, entorhinal cortex and temporoparietal cortex, the more severe
is the AD cognitive decline. Therefore we can conclude that, while Aβ plaques
should be considered a disease biomarker, tau-positive inclusions mainly represent
a progression-related biomarker.
AL GRAWANY
2 Neurodegenerative Diseases in Geriatric Patients 25
atrophy. Finally, lvPPA is a disturbance in thinking of the words to use while speak-
ing, narrow attention span, progressive hesitation and difficult comprehension of
complex sentences; on MRI this variant is characterized by a selective left temporo-
parietal atrophy. Metabolic studies with FDG-PET confirmed the presence of hypo-
metabolic areas corresponding to atrophic cortex, with a pattern completely
superposed on the one observed at MRI examination.
peduncle width to superior cerebellar peduncle width ratio; a value of more than 14
is highly suggestive for PSP. A recent version called MRPI2.0 also included ven-
tricle enlargement in the computation, with a final figure obtained by multiplying
the MRPI by the ratio of third ventricular width to frontal horn width; however,
these computations can suffer from inter-rater variability and limited reproducibil-
ity. Therefore easier techniques, such as the above-mentioned midbrain-to-pons
area ratio, should be preferred. The area of the midbrain and pons are calculated on
the midline sagittal plan at the level of ponto-mesencephalic and ponto-medullary
junctions; in PSP patients, this parameter is significantly reduced or even halved,
while in PD MSA and healthy brain is approximately 0.24. An example of MRI
findings in PSP is shown in Fig. 2.8. Functional imaging studies are not entirely
specific for PSP; it has been described that FDG-PET can show hypometabolism in
the frontal lobe and/or midbrain, while I-123 ioflupane SPECT may demonstrate
loss of the normal crescent-shaped tracer uptake in the striatum.
a b c
Fig. 2.8 Most prominent MRI features of PSP: atrophy of the midbrain with the hummingbird
sign on sagittal images due to preserved pontine volume (a, black arrow), associated to cerebellar
atrophy and periacqueductal grey matter hyperintensity (b, black arrowheads); mickey mouse
appearance of the brain stem on the axial plane (c); midbrain-pons ratio <0.12 (d)
2 Neurodegenerative Diseases in Geriatric Patients 27
2.4.2 Synucleinopathies
almost constant finding in both the variants; in more advanced phases sleepiness,
dysphonia, dysphagia, dystonia and excruciating pain can also occur.
MSA can be suspected based on clinical and neuroimaging findings. For MSA-P,
the most reliable sign on conventional MRI is represented by putaminal atrophy,
with relative central hypointensity on T2* images due to iron accumulation and
peripheral hyperintensity on T2w images due to reactive astrogliosis. Conversely, in
MSA-C the most evocative signs are represented by severe isolated atrophy of cer-
ebellum and brainstem (especially olivary nuclei and middle cerebellar peduncle)
coupled to a cruciform signal hyperintensity in the central pons known as hot cross
bun sign (due to the selective degeneration of transverse pontocerebellar fibres and
median pontine raphe nuclei with relatively spared corticospinal tracts) (Fig. 2.9).
Although specific for MSA-C, hot cross bun sign only appears in case of advanced
disease progression. Among NM techniques, I-123 ioflupane SPECT is usually nor-
mal in these patients. In recent times, the identification of new PET radiotracers as
well as the resort to diffusivity analysis coupled to automated volume loss quantifi-
cation on volumetric MRI acquisitions have been proposed as a tool for distinguish
MSA from other synucleinopathies and from tauopathies; however, these studies
are still embryonic and far from clinical widespread application [26].
a b
Fig. 2.9 Most prominent MRI features of MSA-C: severe isolated atrophy of cerebellum
and brainstem on the axial (a) and coronal (b) planes; diffuse ex vacuo dilated cerebellar folia (a,
black arrow), associated with a mild cruciform signal hyperintensity in the central pons (a, white
arrowhead)
2 Neurodegenerative Diseases in Geriatric Patients 29
a b c
Fig. 2.10 Most relevant MRI features of PD: partial loss of the normal swallow tail appearance
on axial SWI at the level of substantia nigra pars compacta (a, white arrow) coupled to mild T2w
signal alteration on the coronal plane (b); no significant atrophy is observed in the supratentorial
compartment (c)
30 C. Russo et al.
symptoms during their lives. This significant overlap between CAA and AD must be
framed in the common presence of amyloid deposits in brain tissue; in CAA such
insoluble oligomers deposition is mainly found in blood vessel primarily at the level
of smooth muscle cells, but also in pericytes and endothelial cells. It was hypothe-
sized that amyloid produced by neurons is drained along the perivascular spaces
within brain parenchyma up to the abluminal portion of the tunica media, the sur-
rounding smooth muscle cells and in the adventitia, where it deposits under specific
conditions thus causing diffuse angiopathy [28].
Generally sporadic and occasionally found as a familiar disorder, CAA is a pos-
sible cause of MCI and dementia; cognitive impairment in this setting can be both
gradual (due to a vascular dementia caused by lobar cerebral microhaemorrhages
and ischemic leukoencephalopathy) or rapidly progressive (in case of recurrent
haemorrhages or inflammatory angiopathy).
At MRI the two primary features of CAA are represented by microbleeds and
WMH (more or less confluent depending on the severity of ischemic leukoencepha-
lopathy and generally sparing subcortical fibres), where the relation between micro-
bleed burden and cognitive impairment severity is known to follow an exponential
trend. Cerebral microbleeds are defined as perivascular deposits of haemosiderin
from millimetric micro-haemorrhages, usually located at grey–white matter junc-
tion or in the cerebellum but sparing basal ganglia and pons; microbleeds can be
distinguished only on T2* sequences as small foci of blooming artefact, with SWI
up to 10 times more sensitive than other T2* images. Other possible findings also
include lobar or cerebellar haemorrhages (that tend to spare the basal ganglia and
pons), convexity subarachnoid haemorrhage and/or superficial siderosis (as a
chronic manifestation of previous convexity subarachnoid haemorrhages, both
symptomatic or asymptomatic), dilated perivascular spaces at the level of centrum
semiovale and corona radiate, and microinfarcts or ischemic lacunae (due to acute,
subacute or chronic ischemic insult).
2.4.4 Others
within brain tissues provides an over increasing substrate for this cascade effect. At
histological examination, the counterparty of these events is the pathological depo-
sition of amyloid plaques within normal brain tissue, coupled to the vacuolization
of neutrophil and normal myelin. Depending on molecular markers observed in
affected patients, sCJD can be further divided into subtypes based on two elements:
the amino acid at codon 129 in the prion protein gene (subtype MM if methionine,
VV if valine, or MV if both) and the size of the protease-resistant core of the abnor-
mal prion protein (subtype 1 if 21 kDa, 2 if 19 kDa or 1 + 2 if both PrPSc types).
sCJD diagnosis relies on the combination of clinical features coupled to the
results in one or more para-clinical tests among electroencephalogram (EEG), cere-
brospinal fluid (CSF) analysis and/or MRI. EEG is generally aspecific, with the
most evocative patterns of alteration only observed in late disease stages. CSF anal-
ysis is probably the most sensitive supportive examination, allowing for the detec-
tion of a specific protein called 14-3-3 (whose expression is correlated to sCJD with
a sensitivity and a specificity of >90%). However MRI abnormalities still represent
the most important hallmark for CJD, with alterations visible even before or in case
of unremarkable findings at EEG and CSF examination [29].
Most common MRI alterations are represented by symmetric or asymmetric, dif-
fuse or focal restriction of water diffusion associated to a less marked increase in
T2-FLAIR signal within cortical and deep grey matter. The aetiology of DWI
abnormalities is still poorly understood, probably related to compartmentalization
within myelin vacuoles or to abnormal deposition of prion protein somehow restrict-
ing free water diffusion. The most typical patterns involve insula, cingulate cortex,
superior frontal gyrus, striatum and thalamus (generally with anterior → posterior
gradient); peri-rolandic cortex, pulvinar and cerebellum are usually (but not invari-
ably) spared. Usually cortical and deep grey matters are both affected, with the
exception of VV1 subtype prominent cortical involvement (almost completely spar-
ing deep grey matter) and of MV2/VV2 exclusive basal ganglia involvement with
very limited/absent cortical abnormalities (Fig. 2.11). Cerebellar atrophy can also
be observed, especially in later CJD stages.
The intensity of the T2-FLAIR equivalent depends on disease severity and dura-
tion; moreover, if NM is performed, the areas of restricted water diffusion and ele-
vated T2-FLAIR signal correspond to PET/CT areas of hypometabolism. MRI
features are therefore crucial in case of sCJD clinical suspicion, supporting the diag-
nosis when typical imaging patterns are observed and possible mimics ruled out,
thus guiding the approach to EEG/CSF interpretation.
a b c
Fig. 2.11 Three possible patterns of signal alteration in sCJD (first row DWI sequences, second
row FLAIR sequences): usual asymmetric basal ganglia involvement with mild DWI restriction of
frontal cortex (a); more unusual bilateral and symmetric basal ganglia involvement (b); rare selec-
tive cortical involvement (c)
instability during cell division caused by the abnormal number of these repeats;
generally the trinucleotide repeats expand until the gene stops functioning.
Huntingtin protein has a prominent role in protein trafficking, vesicle transport,
synaptic (i.e. dopaminergic) signalling and neural cells apoptosis; therefore, its loss
or toxic gain of function is responsible for premature neurodegeneration due to
disruption in many intracellular pathways.
Being HD characterized by an autosomal dominant pattern of inheritance with
complete penetrance, the probability of transmitting the mutated gene is 50% and
all the individuals with the pathologic allele express the clinical phenotype. Although
most serious presentation is generally observed in young adults, HD can also be
diagnosed in elderly in case of short CAG expansion, with the highest recorded
prevalence ranging between 60 and 69 years; in these cases, atypical parkinsonism
as well as cognitive or psychiatric disturbances are the first manifestations, with
chorea and deficit in postural control appearing later on in more advanced stages
(MCI described up to 15 years before motor symptom onset) [30].
Due to the pervasive impact on dopaminergic circuits, HD is mainly character-
ized by the loss of GABAergic neurons within basal ganglia, especially in caudate
2 Neurodegenerative Diseases in Geriatric Patients 33
and putamen nuclei; this neuronal loss is responsible for the most typical HD imag-
ing feature at MRI, characterized by focal atrophy and secondary enlargement of
frontal horns of the lateral ventricles (with increased frontal horns width to intercau-
date distance ratio or intercaudate distance to inner table width ratio). Indeed it has
been demonstrated that striatal atrophy is positively correlated with disease severity
and negatively correlated with the number of CAG triplet repeats. Besides the stria-
tum, extra-striatal atrophy can also be observed in thalamus, hypothalamus, globus
pallidus, limbic system and cerebellum; occasional findings may also include iron
deposition within basal ganglia on SWI and increased T2w signal of the putamen.
In preclinical HD as well as in case of atypical clinical presentation with poor motor
symptoms, FDG-PET showing striatal hypometabolism or DATscan showing
decreased striatal dopamine transporter binding may support MRI findings is guid-
ing the genetic diagnosis.
increasing number of putative genes has been identified, with variable phenotypical
and MRI presentation depending on the specific causative mutation. Generally
affecting children or young adults, NBIA can also be occasionally observed in
elderly population; in these unusual cases, the most common manifestation is repre-
sented by a late-onset atypical parkinsonism. When clinical suspicion is raised,
MRI and DATscan are required to identify typical findings as well as to rule out
PD. The most common NBIA in the elderly is represented by pantothenate kinase-
associated neurodegeneration (PKAN) [33], whose most remarkable MRI feature is
represented by T2w hypointensity within globus pallidi and substantia nigra (cor-
responding on SWI/T2* to susceptibility artefacts due to abundant iron deposition)
with a central hyperintense spot due to myelin vacuolisation (also known as “eye of
the tiger” sign); on spectroscopy, decreased N-acetylaspartate and increased myo-
inositol levels can be observed due to neuronal depletion. Other possible NBIAs,
such as aceruloplasminaemia or neuroferritinopathy, are only exceptionally
observed.
References
1. Chen J, Lin K, Chen Y. Risk factors for dementia. J Formos Med Assoc. 2009;108(10):754–64.
https://doi.org/10.1016/S0929-6646(09)60402-2.
2. Przedborski S, Vila M, Jackson-Lewis V. Neurodegeneration: what is it and where are we? J
Clin Invest. 2003;111(1):3–10.
3. Cloutier S, Chertkow H, Kergoat M, Gauthier S, Belleville S. Patterns of cognitive decline prior
to dementia in persons with mild cognitive impairment. J Alzheimer’s Dis. 2015;47:901–13.
4. Tripathi M, Vibha D. Reversible dementias. Indian J Psychiatry. 2009;51 Suppl 1(Suppl
1):S52–5. https://pubmed.ncbi.nlm.nih.gov/21416018.
5. Martin-macintosh EL, Broski SM, Johnson GB, Hunt CH, Cullen EL, Peller PJ. Multimodality
imaging of neurodegenerative processes: part 1, the basics and common dementias.
AJR. 2016;207:871–82.
6. Martin-macintosh EL, Broski SM, Johnson GB, Hunt CH, Cullen EL, Peller PJ. Multimodality
imaging of neurodegenerative processes: part 2, atypical dementias. AJR. 2016;207:883–95.
7. Custodio N, Montesinos R, Lira D, Herrera-Perez E, Bardales Y, Valeriano-Lorenzo L. Mixed
dementia. A review of the evidence. Dement Neuropsychol. 2017;11(4):364–70.
8. Han R, Liang J, Zhou B. Glucose metabolic dysfunction in neurodegenerative diseases—new
mechanistic insights and the potential of hypoxia as a prospective therapy targeting metabolic
reprogramming. Int J Mol Sci. 2021;22:5887.
9. Bega D, Cindy G, Spies W. Is there a role for DAT-SPECT imaging in a specialty movement
disorders practice? Neurodegener Dis. 2015;15(2):81–6.
10. Morbelli S, Esposito G, Arbizu J, Barthel H, Boellaard R, Bohnen NI. EANM practice guide-
line/SNMMI procedure standard for dopaminergic imaging in Parkinsonian syndromes 1.0.
Eur J Nucl Med Mol Imaging. 2020;47:1885–912.
11. Bega D, Kuo PH, Chalkidou A, Grzeda MT, Macmillan T, Brand C, et al. Clinical utility of
DaTscan in patients with suspected Parkinsonian syndrome: a systematic review and meta-
analysis. Park Dis. 2021;43:1–8.
12. Lemoine L, Gillberg P, Svedberg M, Stepanov V, Jia Z, Huang J, et al. Comparative bind-
ing properties of the tau PET tracers THK5117, THK5351, PBB3, and T807 in postmortem
Alzheimer brains. Alzheimers Res Ther. 2017;9(96):1–13.
13. Leuzy A, Chiotis K, Lemoine L, Gillberg P, Almkvist O, Rodriguez-vieitez E, et al. Tau PET
imaging in neurodegenerative tauopathies—still a challenge. Mol Psychiatry. 2019;24:1112–34.
2 Neurodegenerative Diseases in Geriatric Patients 35
14. Sánchez-juan P, Ghosh PM, Hagen J, Henry M, Grinberg LT, Neil JPO, et al. Practical utility
of amyloid and FDG-PET in an academic dementia center. Neurology. 2014;82(21):230–8.
15. Vernooij M. Outline presentation Imaging in cognitive decline: MRI protocols. Why imaging
in dementia? Conventional MR imaging: main aim. Patterns of atrophy: imaging consider-
ations MTA: perpendicular recons needed; 2019. p. 1–12.
16. Håkansson C, Torisson G, Londos E, Hansson O, Björkman-burtscher IM, Van Westen
D. Reporting frequency of radiology findings increases after introducing visual rating scales in
the primary care diagnostic work up of subjective and mild cognitive impairment. Eur Radiol.
2021;31:666–73.
17. Vernooij MW, Pizzini FB, Schmidt R, Smits M, Yousry TA, Bargallo N, et al. Dementia imag-
ing in clinical practice: a European-wide survey of 193 centres and conclusions by the ESNR
working group. Neuroradiology. 2019;61:633–42.
18. Van Veluw SJ, Lauer A, Charidimou A, Bounemia N, Xiong L, Gregoire B, et al. Evolution of
DWI lesions in cerebral amyloid angiopathy. Evidence for ischemia. Neurology. 2017;89:1–8.
19. Sobański M, Zacharzewska-Gondek A, Waliszewska-Prosól M, Sąsiadek MJ, Zimny A,
Bladowska J. A review of neuroimaging in rare neurodegenerative diseases. Dement Geriatr
Cogn Disord. 2020;49:544–56.
20. Erkkinen MG, Kim M, Geschwind MD. Clinical neurology and epidemiology of the major
neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2018;10(4):a033118.
21. Staffaroni AM, Elahi FM, McDermott D, Marton K, Karageorgiou E, Sacco S, et al.
Neuroimaging in dementia. Semin Neurol. 2018;37(5):510–37.
22. Van Oostveen WM, de Lange ECM. Imaging techniques in Alzheimer’s disease: a review of
applications in early diagnosis and longitudinal monitoring. Int J Mol Sci. 2021;22:2110.
23. Oldan JD, Jewells VL, Pieper B, Wong TZ. Complete evaluation of dementia: PET and MRI
correlation and diagnosis for the neuroradiologist. AJNR. 2021;42(6):998–1007.
24. Albrecht F, Mueller K, Ballarini T, Lampe L, Diehl-schmid J, Fassbender K, et al. Unraveling
corticobasal syndrome and alien limb syndrome with structural brain imaging. Cortex.
2019;117:33–40.
25. Rohrer J. Structural brain imaging in frontotemporal dementia. Biochim Biophys Acta.
2012;3(1822):325–32.
26. Watanabe H, Riku Y, Hara K, Kawabata K, Nakamura T, Ito M, et al. Clinical and imaging
features of multiple system atrophy: challenges for an early and clinically definitive diagnosis.
J Mov Disord. 2018;11(3):107–20.
27. Gayed I, Joseph U, Fanous M, Wan D, Schiess M, Ondo W, et al. The impact of DaTscan in
the diagnosis of Parkinson disease. Clin Nucl Med. 2015;40(5):390–3.
28. Biffi A, Greenberg SM. Cerebral amyloid angiopathy: a systematic review. J Clin Neurol.
2011;7:1–9.
29. Fragoso DC, Lio da Mota Gonçalves Filho A, Torres Pacheco F, Rodi Barros B, Aguiar Littig
I, Hoffmann Nunes R, et al. Imaging of Creutzfeldt-Jakob disease: imaging patterns and their
differential diagnosis. Radiographics. 2017;37(4):234–57.
30. McColgan P, Tabrizi SJ. Huntington’s disease: a clinical review. Eur J Neurol. 2018;25(1):24–34.
31. Cocozza S, Pontillo G, De Michele G, Di Stasi M, Guerriero E, Perillo T, et al. Conventional
MRI findings in hereditary degenerative ataxias: a pictorial review. Neuroradiology.
2021;63:983–99.
32. Lieto M, Roca A, Santorelli FM, Fico T, De Michele G, Bellofatto M, et al. Degenerative and
acquired sporadic adult onset ataxia. Neurol Sci. 2019;40:1335–42.
33. Tofaris GK, Revesz T, Jacques TS, Papacostas S, Chataway J. Adult-onset neurodegeneration
with brain iron accumulation and cortical alpha-synuclein and tau pathology. A distinct clini-
copathological entity. Arch Neurol. 2007;64:280–2.
Neurovascular Emergencies in Geriatric
Patients 3
Giuseppe Maria Di Lella, Luca Ausili Cefaro,
and Cesare Colosimo
3.1 Introduction
growing subset of the population. Effective management of patients who have cere-
brovascular disease depends on accurate diagnosis. The neuroradiological studies
and treatments play a major role in the diagnosis and treatment of patients often
affected also by other age-related symptoms and pathologies (i.e., dementia, diabe-
tes, etc.). The wide diffusion, in the last two decades, of high-quality CT and MRI
systems has allowed better and faster anatomic and functional evaluation, or exclu-
sion, of ischemic and hemorrhagic lesions of arterial or venous origin, especially
with the availability of fast and reliable CT and MR perfusion techniques. The
“need of speed” in these often uncooperating patients is in fact of paramount impor-
tance in order to achieve a proper diagnosis. Neuroradiology plays an established
role also in the therapeutic phase, due to the continuous advance of the materials
and techniques available (i.e., flow diverter stent in the treatment of aneurysms).
Technological advancements have led to advanced interventions such as angio-
plasty, stenting, and carotid endarterectomy for selected stroke patients and
expanded access to acute stroke services through tele stroke programs. In the past
decade, we have also enhanced our understanding of the mechanisms of brain
injury, repair, plasticity, and recovery that hopefully will improve our future post
stroke treatments. In the United States from 1997 to 2007, the stroke death rate fell
44.8%, and the actual number of stroke deaths declined 14.7%, making stroke the
fourth instead of the third leading cause of death.
Stroke is the fifth leading cause of death among elderly persons in the United States
[1]. While stroke can occur at any age, ischemic stroke is predominantly a disease
of the elderly, as age is the most substantial nonmodifiable risk factor. Stroke risk
increases with age, and in 2009, 66% of persons hospitalized for stroke were
65 years or older. The growing longevity of human populations and the associated
multimorbidity explain how the number of accidental strokes is projected to double
between 2010 and 2050, with most strokes occurring in adults over the age of
75 years [2]. Additionally, this age group (>75 years) experiences more hospitaliza-
tion stays and higher mortality post-stroke. Over 80% of strokes result from isch-
emic damage to the brain due to an acute reduction in the blood supply. Around
25–35% of strokes present with large vessel occlusion, and the onset of patients in
this category is often characterized by severe neurological deficits [3, 4].
Stroke is a complex disease that can be caused by multiple potential etiologies;
it is really a heterogeneous disorder with more than 100 pathologies implicated in
the pathogenesis. However the end result is mostly represented by the occlusion of
an artery, which almost immediately leads to hypo perfusion of the tissue segment
supplied by that vessel; the affected parenchyma usually consists of a severely hypo
perfused (cerebral blood flow [CBF] ≤10 mL/100 g/min) central infarct core where
the damage is irreversible. It is bordered by the critically hypo perfused (CBF
10–20 mL/100 g/min) ischemic penumbra (tissue-at-risk), where the injury may be
reversed if timely reperfusion occurs. Collaterals aim at preserving as much
3 Neurovascular Emergencies in Geriatric Patients 39
penumbral tissue as possible. With time (minutes to hours) the infarct core expands
at the expense of the penumbra. This is also helped by the mass effect of the edema-
tous tissue on the neighboring arteries. The penumbra is surrounded by other
involved tissue, which is not at risk of infarction, the so-called benign oligemia
(CBF >20 mL/100 g/min) [5] (Figs. 3.1 and 3.2).
Stroke imaging (CT and/or MR) is crucial in the handling of such patients and
has to be performed in a fast and efficient manner; early diagnosis and assessment
are important in the treatment of acute ischemic stroke (AIS). The main roles of
imaging are: exclude an intracranial hemorrhage, define the ischemic region, distin-
guish between infarct core and penumbra, and finally depict the vessel status. In
particular:
a b c
f
d e
Fig. 3.1 Right occipital ischemic stroke. A 75-year-old woman with visual disturbance (hemi-
anopsia). FLAIR (a), T2 (axial and coronal; b, e), and DWI (d) show the classic appearance and
MR imaging of an acute infarct in the PCA territory, with hyperintensity in the cortex and subcorti-
cal white matter of the right deep occipital lobe. Angio-TOF sequences (c) and T1 C+ MR (f)
confirm the presence of a right P2 segment occlusion
40 G. M. Di Lella et al.
a b c
d e f
Fig. 3.2 Left temporal-parietal ischemic stroke. A 71-year-old man with aphasia and sudden right
hemiparesis; classic appearance of an early subacute cerebral infarct. Axial CT scan (a) in the ER
show a low-density area with loss of SB/SG differentiation and swollen appearance of the involved
temporo-parietal cerebral convolutions; axial MIP view of the CTA (b) shows normal cerebral
intracranial district (“circle of Willis”). MR FLAIR axial and DWI (c, f) and T2 coronal TSE (d)
obtained 48 h after the initial onset of speech difficulties demonstrate an area of hyperintensity in
the same region, associated with hyperintensity in DWI images, due to typical diffusion restriction
in the acute phase (within 7 days). Angio-MRI (TOF 3D, e) confirms the normal parity of arterial
circulation of the base of the skull
loss of the gray–white matter interface, and swelling of the cerebral tissue. The
Alberta Stroke Program Early Computed (ASPECTS) is a CT score system that
can be used to quantify the extent of ischemia: 10 brain regions are assessed
dichotomously for the presence (or not) of early signs of ischemic stroke, result-
ing in a range 0–10, with 1 point subtracted for any evidence of early ischemic
change in each region defined on the CT scan; baseline CT showing a large area
of hypo attenuation is considered as an indicator of poor outcome (ASPECTS <7).
–– Hypo perfusion abnormalities can be accurately measured using perfusion CT
(PCT) and MR perfusion-weighted imaging (PWI); these are able to define the
area of irreversible severe ischemia, with complete loss of oxygen and glucose
supply and resultant depletion of energy stores, cellular necrosis, and cavitation
(“ischemic core”) and also the area surrounding the ischemic core, characterized
3 Neurovascular Emergencies in Geriatric Patients 41
by moderate ischemia and cellular dysfunction but not cell death, which is poten-
tially reversible with prompt reperfusion (“ischemic penumbra”). Perfusion CT
(PCT) is one of the best imaging techniques readily available in an emergency
room to evaluate acute stroke patients for the presence, quantity, and distribution
of the ischemic penumbra, through a dynamic technique involving sequential CT
data acquisition [6] in suspect brain areas during an intravenous bolus injection
of iodinated contrast medium. The most relevant and used parameters are: cere-
bral blood flow (CBF), cerebral blood volume (CBV), average transit time
(MTT), time-to-peak (TTP). In the ischemic penumbra, cerebral perfusion is
impaired, but self-regulation is preserved; vasodilation and collateral recruitment
lead to an increase in CBV. Quantification of ischemic “core” (CBF <30%) and
estimation of “penumbra” or tissue at risk (T-max >6 s) can provide immediate
information for treatment decision-making. Clinical trials have shown that perfu-
sion mismatch ratios of core/penumbra greater than 1.8 may indicate the eligibil-
ity for endovascular treatment (EVT) [7, 8].
Some institutions have MRI available anytime and prefer it over CT, when the
patient’s condition permits, because of the additional information it provides.
Diffusion-weighted magnetic resonance imaging (MRI) is the most useful
method for detecting hyper acute ischemia and the “ischemic core” while PWI,
in selected cases, could be used to evaluate hypo perfusion abnormalities. MRI
can detect abnormal cytotoxic edema (this restricted diffusion areas are seen as
bright on b1000 DWI images and with low signal on the corresponding automati-
cally calculated apparent diffusion coefficient [ADC] maps) in the early stage
and show clear discrimination between ischemic lesions and normal brain tissue.
After the acute phase of decreasing ADC to the lower value, at 1–4 days, the
ADC subsequently rises and “pseudo normalizes” to transient equivalence with
normal brain tissue (although the tissue is infarcted), typically at 1–2 weeks, and
keeps rising until the ADC values would result elevated in the chronic stage [9].
Since the signal intensity on DWI depends on ADC as well as the T2 information
inherent in the b = 0 echo-planar image, net hyper intensity on the DWI images
may be due to low ADC or T2 effects: the “T2 shine-through.” The DWI-FLAIR
mismatch depends on the lack of marked parenchymal hyper intensity on fluid
attenuated inversion recovery (FLAIR) on early MRI study and has been used as
an MRI parameter to widen the treatment window, with intravenous bolus of
r-TPA, in patients with AIS with onset of symptoms beyond 4.5 h and/or in case
of unknown onset time, as in the “wake-up” stroke. Neuroimaging methods, par-
ticularly MRI, based on PWI parameter and DWI lesion volumes, may allow us
to identify the ischemic penumbra and predict brain tissue viability in patients
with AIS, although criteria to define clinically relevant mismatch are not yet
standardized [10] (Fig. 3.3).
–– Obviously the evaluation of intracranial arterial vessels and collateral circula-
tion, made possible through this kind of imaging modalities, is of paramount
importance and is achieved by CT or MR angiography. The study should cover
the entire arterial tree, from the aortic arch to the vertex. MRI provides the advan-
tage of noncontrast imaging of the intracranial arteries using the flow-sensitive
42 G. M. Di Lella et al.
a b c d
e f g h
Fig. 3.3 CT, CTA, CTP, and MRI evaluation in the left MCA territory stroke. Normal appearing
non-contrast CT scan in the ER of a 76-year-old female with sudden onset of aphasia (a). Axial
MIP view of the CTA (b) shows a distal left MCA occlusion, while CT perfusion (c, d) shows an
infarct, with core–penumbra mismatch in the left MCA territory. Non-contrast CT obtained 48 h
after initial onset of speech difficulties (e) shows the classic appearance of an early subacute cere-
bral infarct. Note the wedge-shaped, low-density area involving both the gray and white matter in
the left MCA distribution and in the left thalamus. Follow-up MRI with axial DWI (/ADC) image
(f, g) and FLAIR (h) in the same patient show hyperintensity in the same region within the left
MCA territory. The DWI hyperintensity is due to true diffusion restriction, typical in the acute
phase (within 7 days)
AL GRAWANY
3 Neurovascular Emergencies in Geriatric Patients 43
a b c
d e f
Fig. 3.4 Artery occlusion in a right temporal ischemic stroke. A 71-year-old man with confu-
sional state and sudden onset of left hemiparesis. CT axial documents tenuous right temporo-polar
hypodensity with initial loss of SB/SG differentiation (a). CTA demonstrates the occlusion of the
M1 segment of the right middle cerebral artery (b); note the hyperdensity of the horizontal section
of the rACM on the basal images (a). The CT follow-up after 6 h shows a better defined area of
low-density, with loss of SB/SG differentiation and swollen appearance of the temporo-insulare
cerebral convolutions involved (d). MR axial DWI and T2 coronal TSE confirm the lesion, repre-
sented by an area of T2 hyperintensity in the same region, associated with hyperintensity in DWI
images, due to typical diffusion restriction in the acute phase (within 7 days, c, f). Angio-TOF
sequences document re-habilitation of the right M1 segment of the ACM (e)
recombinant tissue plasminogen activator (r-tPA) remains the standard of care for
patients with moderate to severe neurological deficits who present within 4.5 h from
symptom onset (Acute Ischemic Stroke [Study ECASS-3] and the American Heart
Association/American Stroke Association [AHA/ASA]).
Treatment with intravenous thrombolysis has been tested using imaging bio-
markers to select patients with unknown stroke time onset. These biomarkers com-
prise either penumbral imaging (i.e., perfusion-diffusion MRI or perfusion CT) or
MRI-based tissue-clocking—i.e., the mismatch between a visible ischemic lesion
on diffusion weighted imaging (DWI) and lack of marked parenchymal hyper inten-
sity on fluid attenuated inversion recovery (FLAIR) on MRI (termed DWI-FLAIR
mismatch). The results of recent trials (EXTEND, ECASS4-EXTEND and
EPITHET) have shown, in fact, that patients with ischemic stroke 4,·5–9 h after
stroke onset or with wake-up stroke with evidence of salvageable brain tissue using
CT perfusion or perfusion-diffusion MRI, who were given intravenous Alteplase,
have improved functional outcomes compared with those given placebo. The “ben-
efit to risk” ratio seems to be larger in patients who meet automated perfusion mis-
match criteria [14].
Outcomes for some patients with acute ischemic stroke and moderate to severe
neurological deficits due to proximal artery occlusion are improved with endovas-
cular reperfusion therapy. Efforts to hasten reperfusion therapy, regardless of the
mode, should be undertaken within organized stroke systems of care [15]. It is also
suggested that the EVT should not just be held back based on age, and even patients
over the age of 80 years can benefit from EVT. In conclusion, studies recently dem-
onstrated statistically that endovascular treatment (EVT) combined with intrave-
nous thrombolysis (IVT) has good efficacy and high safety in the treatment of acute
intracranial arterial occlusion, and the combination can significantly improve the
patients’ quality of life, therefore having a high clinical application value [16].
Nontraumatic intracerebral hemorrhage (ICH) results from bleeding into the brain
parenchyma arising from the rupture of an arterial vessel, most often (>80%) a
small arteriole affected by cerebral small vessel diseases (SVD).
Stroke is the second leading cause of death worldwide, and one of the leading
causes of disability. ICH is the second most common subtype of stroke after isch-
emic stroke and accounts for approximately 10–20% of all strokes [17, 18]. A large
meta-analysis recently concluded that most likely the incidence of intracerebral
3 Neurovascular Emergencies in Geriatric Patients 45
hemorrhage has not changed between 1980 and 2006, but consistent across the vari-
ous studies was the finding that the incidence of intracerebral hemorrhage increased
strongly with age, with persons aged 85 years and over having an almost tenfold
increase in yearly risk of intracerebral hemorrhage. Overall case fatality at 1 month
for ICH is reported as 40%. ICH outcomes have been well described in the general
population, but there is a paucity of data regarding complications and outcomes in
the “very elderly” (age >80 years). Available data suggest that patients >85 years
have an almost tenfold increase in yearly risk of ICH compared to patients aged
45–54 years. As the population ages, the incidence of ICH in the very elderly will
likely increase, and the number of Americans aged >65 years is projected to more
than double by 2060 [19, 20].
More than 50% of primary ICH events are directly correlated with hypertension
as a risk factor, whereas ≈30% are known to be associated with cerebral amyloid
angiopathy (CAA). Deep perforator arteriopathy is linked with hypertension
(though not exclusively) and is a frequent cause of nonlobar or deep ICH in the
basal ganglia or brainstem but also contributes to lobar hemorrhage (Fig. 3.5).
CAA is caused by amyloid beta deposition in cortical and leptomeningeal blood
vessels and is a major contributory cause of lobar ICH. Other ICH risk factors are
genetic factors, diabetes, alcohol intake, smoking, oral anticoagulant treatment,
drug abuse, and older age. Intracerebral hemorrhage can occur as a complication of
a preexisting lesion, such as vascular malformation or tumor, which is then referred
to as secondary intracerebral hemorrhage (15–20%) [21–23].
The clinical presentations of ICH and ischemic stroke are similar, typically con-
sisting of abrupt onset of a focal neurologic deficit. Decreased level of conscious-
ness, vomiting, headache, seizures, and very high blood pressure (BP) might suggest
the presence of ICH. However, none of these symptoms/signs is specific enough to
distinguish hemorrhagic from ischemic stroke [24].
Intracerebral hemorrhage is therefore a life-threatening medical emergency that
requires timely diagnosis; brain imaging is essential to reliably distinguish ICH
from ischemic stroke, usually with a rapid noncontrast CT (NCCT) which is highly
sensitive for all forms of acute intracranial hemorrhage, is a fast technique, with
excellent sensitivity, also in noncooperative patients, to identify acute ICH, and
given its wide availability is considered the gold standard for the diagnosis of ICH
in the ER departments. Beyond the diagnosis of ICH, NCCT can provide useful ele-
ments such as ICH location, intraventricular extension, hydrocephalus, presence
and degree of edema, and midline shift or brainstem compression secondary to the
mass effect from the hematoma. Moreover, NCCT allows to quantify the volume of
a hematoma and determine its approximate age, by evaluating the density of the
lesions, measured in Hounsfield units (HU), according to the value of X-ray attenu-
ation corrected for the attenuation coefficient of water; HU for water is equal to 0,
blood is between 30 and 45, gray substance is between 37 and 45, white substance
is between 20 and 30, whereas bone is between 700 and 3000. On CT, an acute ICH
typically presents as a hyper dense mass within the brain parenchyma showing
Hounsfield Units (HU) of 50–70. Within 1–6 weeks, the ICH becomes isodense
(=“subacute” ICH) typically showing a decrease of attenuation of 1.5 HU a day.
46 G. M. Di Lella et al.
a b
c d
Fig. 3.5 “Typical” primary hemorrhage in the right lentiform nucleus. A 72-year-old man with
hypertension and ASA therapy, the axial non-contrast CT study in ER shows the classic appear-
ance of an acute hemorrhage in the R basal ganglia involving the putamen and external capsule
(striatocapsular), with minimal surrounding edema, without significant mass effect (a). CTA, in the
coronal plane, shows mild displacement of the lenticulostriate arteries. There is no spot sign that
would indicate active bleeding. No underlying vascular lesion was found (b). Follow-up MRI with
axial T1 TSE (c) and coronal T2 TSE (d) shows the physiological evolution in the early subacute
phase of the striatocapsular hemorrhage, due to transformation in intracellular methemoglobin
a b c d
e f g h
Fig. 3.6 “Typical” intra-axial hemorrhage in the left thalamus. A 72-year-old woman with sudden
right loss of strength and dysarthria. Axial CT study in ER show a small hyperacute hemorrhage in
the left thalamus with minimal surrounding edema (“typical” intra-axial hemorrhage, a). Axial CT
at 24 h demonstrates huge increase of the blood collection and the mass effect, with extension of
the bleeding in the ventricular system, also in the fourth ventricle (not shown) (b). The subsequent
CT angiography did not show arterial or venous malformations, but demonstrated further enlarge-
ment of the hematoma (d). Follow-up MRI with angio-TOF sequences at 1 month (and CT, c)
evidentiate size reduction of the collection, due to the disappearance of surrounding edema and
physiological evolution of the blood components: hemosiderin in the periphery (black ring) and
mostly deoxyhemoglobin in the core (e–g). No vascular malformation was found on the TOF
angiographic sequence (h)
48 G. M. Di Lella et al.
vital parameters and reduce/eliminate the factors that determine ICH (modifiable
factors), with stabilization of blood pressure, suspension/reduction of anticoagulant
drugs, and possibly administration of prothrombotic drugs. The role of surgical
therapy remains controversial and surgical evacuation of supratentorial hematomas
should be considered only as a lifesaving measure in deteriorating patients. The
only condition in which there is consensus in favor of surgical intervention is in
cerebellar hematomas with clinical or imaging signs of hydrocephalus and/or brain-
stem compression. In these cases, surgical decompression and hematoma evacua-
tion should be performed as soon as possible in the vast majority of cases [32, 33].
a b c d
e f
Fig. 3.7 Ruptured ACA saccular aneurysm in a 74-year-old male. (a) Non-contrast CT scan in ER
at 11 am: widespread, bilateral, and symmetrical subarachnoid hemorrhage secondary to the rup-
ture of an ACA saccular aneurysm. The lesion location was suspected on the non-contrast CT due
to the higher blood density in the lamina terminalis cistern and confirmed with the CT angiography
(b–d). (e, f) DSA, before and after embolization with GDC coils, done on the same day. Note the
complete lesion occlusion and consequent exclusion from the intracranial arterial circulation.
Everything went well, apparently, with progressive albeit partial diappearance of the onset symp-
tomatology, …
scanner, and the skills of the radiologist. If performed within the first 2 days of
SAH, CT scans have 95–100% sensitivity for intracranial hemorrhage. This sensi-
tivity diminishes to 85% within 5 days, 50% after 1 week, 30% after 2 weeks, and
0% after 3 weeks [41–43].
Other than the positive diagnosis of SAH, the initial CT examination can detect
the early complications of hydrocephalus, intraparenchymal hematoma with space
occupying effect and ventricular hemorrhage. Hydrocephalus, which begins with
dilatation of the temporal horns, and a compressive intra-parenchymal hematoma
need to be diagnosed and reported, as they represent life-threatening conditions and
require immediate neurosurgical treatment with the positioning of an external ven-
tricular shunt and/or the evacuation of the hematoma before the treatment of the
cause of the SAH (Fig. 3.9).
Relying on CT of the brain to make a diagnosis of SAH after 1 week is instead
an uncertain and much more challenging affair. Because of the greater availability
and feasibility of CT imaging in patients with suspected subarachnoid hemorrhage,
few studies of MRI in the acute phase after subarachnoid hemorrhage have been
reported. These suggest that in the first few hours and days, MR with proton density
and, nowadays almost exclusively, FLAIR images is as sensitive as CT imaging.
After the initial days, when hyperdensity on CT scans decreases, MR is better for
detecting blood, with fluid attenuation inversion recovery (FLAIR) and T2* images
being the most sensitive techniques [42, 43]. Vascular imaging, usually including
cerebral dye contrast angiography, is needed. CT angiography (CTA) of the circle
50 G. M. Di Lella et al.
a b c
d e f
g h i
Fig. 3.8 (a–i) …but 11 days after the treatment the patient’s history became the “worst case sce-
nario” in a clinical case like this, with the onset of an almost untreatable vasospasm, followed by
multiple ischemic lesions, intraparenchymal bleeding partially along the ventricular catheters, and,
finally, an infectious cerebritis with massive vasogenic edema and ventricular hypertension, that
led this unfortunate patient, a MD Radiologist by the way, to the exitus. This case would be a
reminder that nowadays subarachnoid hemorrhage also represents, also in a “not so old” individ-
ual, a life threatening occurrence, not only at the onset, but also after a routinary successful treat-
ment of the primary lesion
a b
c d
Fig. 3.9 Severe hydrocephalus in ruptured PCoA aneurysm (a, b). Non-contrast CT study in ER
of a 81-year-old woman with severe headache and consciousness alteration shows diffuse sub-
arachnoid hemorrhage (SAH) throughout the basal cisterns and massive intraventricular hemor-
rhage. Note the enlargement of both temporal horns of the lateral ventricles, consistent with early
hydrocephalus. SAH was caused by a ruptured saccular aneurysm located in the PCoA segment of
the right internal carotid artery (not shown). (c, d) Non-contrast CT after the endovascular embo-
lization of the aneurysm and placement of an atrial-ventricular catheter for the treatment of intra-
cranial hypertension
52 G. M. Di Lella et al.
a b c
d e f
Fig. 3.10 Endovascular treatment in an SAH from a saccular L Pcom ruptured aneurysm. A
74-year-old woman with sudden, severe headache. Axial CT study in ER shows extensive sub-
arachnoid hemorrhage (SAH) throughout the basal cisterns and intraventricular hemorrhage
(left>right). Note the enlargement of both temporal horns of the lateral ventricles, consistent with
early hydrocephalus (a, b). Cerebral angiography demonstrates the presence of a saccular Pcom
aneurysm of the left internal carotid artery, which most likely represents the cause of the bleeding
(c). Unsubtracted images during coiling of the aneurysm with balloon assistance for the neck cov-
erage (f). Axial CT after the endovascular embolization of the aneurysm shows an ample parenchy-
mal ischemic area (in the left frontal and parietal regions) due to SAH-related vasospasms, with
mild mass effect and compression of lateral ventricles (d, e)
AL GRAWANY
3 Neurovascular Emergencies in Geriatric Patients 53
therefore developed, which, distorting the local hemodynamic, eliminates the exces-
sive pressure inside the lesion, which suddenly shrinks and could finally disappear.
As a result of these continuous evolution, the percentage of aneurysm malforma-
tions that require a conventional surgical approach has been greatly diminished and
is currently limited to the cases in which the endovascular treatment is not feasible,
due to the patient’s vascular anatomy that could not allow the catheter navigation, or
the complexity of the aneurysm origin, with close relationship with adjacent vessel
that needs to be savaged.
In elderly especially, the endovascular approach has the great advantage of being
much less invasive and therefore more sustainable from often more fragile individu-
als. On the other hand, the vessels are often tortuous, with calcifications and some-
time accompanying stenosis, which in some cases do not allow the catheter to reach
a proper position in order to release the devices (stent and/or coils), required from
the treatment [48–50].
Traumatic brain injury (TBI) is among the leading causes of death and disability
worldwide, with enormous negative social and economic impacts. Traumatic cerebro-
vascular injury (TCVI) is a common pathologic mechanism of traumatic brain injury
with often possible multiple intracranial lesions (Fig. 3.11). Among the elderly, in
recent years, instances of neurotrauma have been increasing [51]. As aged population
grow, so the instances of traumatic brain injury (TBI) in the elderly are increasing. It
has been known that the frequency curve of TBI by age groups has two peaks, in
patients at 15–29 and 65–79 years of age [52]. In recent years, this curve has changed
and currently shows a single peak, only in the elderly age range, resulting from both
decreased frequency in the young and increased frequency in the elderly. In addition,
the peak of frequency in the elderly is continuously shifting toward older ones. Such
changes may be the result of the increased representation of aged individuals in the
population, as well as the reduction in traffic injures, by far more common in the
young people. Age has been proposed as one of the most reliable prognostic factors
following TBI. Both survival and functional outcomes are significantly poorer in the
elderly compared to the younger patients with TBI. It has been also reported that the
duration of hospital stay is significantly longer in the elderly than younger patients
with TBI [53]. The elderly patient frequently presents numerous comorbidities (dia-
betes mellitus, neurodegenerative diseases, arterial hypertension, chronic vascular
encephalopathy) at the time of trauma, which can mask the clinical picture, postpone
treatment, and influence prognosis: as an example, age-related atrophy may provide
space for an intracranial hemorrhage to expand substantially before leading to clini-
cally apparent signs or symptoms that would be detected by the GCS [54].
Morphologically, the distribution of traumatic intracranial lesion varies in the age
groups. Diffuse axonal injury (DAI) is less common in the elderly than the young. In
contrast, focal injury is more common in the elderly. It is well known that the subdu-
ral, contusional, and intracerebral hematomas are more common lesions in the elderly
than the young, although epidural hematomas are less common in the elderly. Those
54 G. M. Di Lella et al.
a b e
c d
Fig. 3.11 SAH, epidural hematoma, and multiple brain contusions in polytrauma. Axial CT scans
in a 69-year-old patient with severe closed head trauma show the classic biconvex configuration of
an acute epidural hematoma in the left parietal region (a) and right frontal and left parietal hemor-
rhagic contusions (the right frontal lesions due to contrecoup mechanism) (b–d) with mild sur-
rounding edema. Traumatic subarachnoid hemorrhage and tiny subdural right hematoma are also
present. Bone CT reconstruction shows a nondisplaced skull fracture of the left parieto-temporal
bone underlying the epidural hematoma (e). Note the subgaleal acute, hyperdense hematoma in the
left temporal area (a)
ADC value on MRI between central and peripheral area. In spite of CBF reduction,
fluid amount is excessive in the area of the cerebral contusion. In addition, hyperemia/
hyperperfusion subsequent to ischemia enhances the increase of post contusional
edema, resulting in delayed deterioration. Delayed traumatic intracerebral hematoma
and expansion of either traumatic acute subdural hematoma or intracerebral hema-
toma may be other important clinical entities for delayed deterioration. A peculiar
entity of delayed deterioration after TBI, the delayed posttraumatic acute subdural
hematoma (DASH), has been reported in elderly patients. DASH has been defined as
an acute subdural hematoma that is not apparent on the initial computed tomography
(CT), and suddenly arise on a follow-up CT after 9–72 h after TBI. Thus, DASH
should be suspected in elderly, anticoagulated, mild TBI patients, including those who
present in the ER with GCS scores of 15 and normal initial CT [55–57]. Current
guidelines recommend “In case of head trauma, the head CT scan is the first line
examination and is recommended for any patient without loss of consciousness or
post-traumatic amnesia, if any of the following is present: neurological deficit, vomit-
ing, severe headache, age over 65 years, suspected skull-base fracture, Glasgow score
<15, coagulopathy, trauma with dangerous mechanism” [58, 59]. A fundamental role
in the approach of the traumatic patient is represented by neuroimaging and in particu-
lar by CT, which through a prompt diagnosis in acute, indicates the management and
helps predict patient outcomes of all ages spectrum. A fundamental role in the
approach to the traumatic patient is represented by neuroimaging and in particular by
CT, which through a timely diagnosis in acute indicates management and helps pre-
dict the outcomes of patients of all age groups. In particular, CT must promptly recog-
nize pathological conditions that require urgent surgery (extensive expansive lesions)
and/or intracranial hypertension. In this context and especially when fractures of the
skull base are present, the integration of a dynamic CT angio study may be useful, for
the evaluation of the main vascular structures in order to exclude arterial dissections,
aneurysms/pseudoaneurysms, and thrombosis of the venous sinuses and of the cere-
bral veins. The unstable clinical conditions of the traumatic patient, the reduced avail-
ability on the territory and the duration of the examination, relegate the MR in urgency
to a secondary role. The damage incurred by TBI can be differentiated into primary
and secondary mechanisms. Post traumatic head lesions included both primary inju-
ries, that are typically defined as the direct mechanical damage caused by trauma
hemorrhagic parenchymal contusions, brain stem injury, traumatic axonal injury
(TAI), parenchymal hematomas, subdural, subarachnoid or extra-dural hematoma,
cranial vault fractures, and secondary injury mechanisms, that are varied and related
to disruption of the blood brain barrier, production of reactive oxygen species and
resultant oxidative stress, metabolic dysfunction, inflammation, and excitotoxicity.
They may become apparent as diffuse cerebral hyperemia, cytotoxic and/or vasogenic
edema, and tissue ischemia [60, 61].
can also be found. t-ESA is more focal and circumscribed than nontraumatic ESA
and is frequently localized in the peri-sylvian regions and in the cerebral sulci adja-
cent to the and/or to epi/sub-dural blood collections [62] (Fig. 3.11).
The clinical presentation appears mostly in the form of headache, classically
defined as maximal at onset and “the worst of life.” The most common cause is
traumatic, also representing the most common form of intracranial hemorrhage in
trauma; approximately 80% of nontraumatic SAH are due to aneurysmal rupture,
with the remainder from idiopathic peri-mesencephalic hemorrhage or other less
common causes.
Noncontrast head CT is the primary means of diagnosis, with the advanced gen-
eration scanners approaching a 100% sensitivity, if completed within 6 h from
symptom onset. The bleeding in the subarachnoid space will result in hyper density
in the first hours on CT-scanner. Within the first 24 h, it is positive in 90/95% of
cases; it should be noted that spontaneous hyper density gradually disappears and
that after a week it is only found in 50% cases. In general, hyper density will depend
on hemoglobin level, amount of blood, and delays between performing the scan and
bleeding [63]. One pitfall might be that the blood and adjacent bone, which both
appear white, can be difficult to distinguish from each other, especially in small
bleeding and in the anemia (sensitivity decreases when the hematocrit is <30%); in
addition, motion artifacts in scans of restless patients can making such scans techni-
cally suboptimal and obscure the diagnosis [64]. In these cases, the Dual Source
scanners may show some edge in the demonstration of such collections.
On MRI the fluid attenuated inversion recovery (FLAIR)/gradient reversal echo
(GRE)/susceptibility weighted imaging (SWI) sequences have a good sensitivity for
the detection of acute SAH in the first 48 h and are complimentary to the CT scans
[65]; however, they are not suitable for a rapid assessment of head injuries. SAH can
be diagnosed by GRE/SWI sequences by its dark signal intensity (“blooming”),
surrounded by the CSF signal intensity. More specifically, the FLAIR sequences,
which are the most sensitive in the first days compared to T1, T2, T2*, show a
hypersignal in the basal cisterns and the sulci of the convexity [64, 66]. The radiolo-
gist evaluation must be careful, because there are other etiologies at the origin of a
hypersignal in FLAIR in the subarachnoid spaces, such as meningitis, hyperoxy-
genation, and metallic artefacts. After a few days from the onset T2* sequences
show hemosiderin deposits, i.e., in the cisterns of the base, or in the cortical furrows.
This modality does not require radiation, though several limitations exist, including
limited availability in the ED, the time required for scanning, the potential for
inducing claustrophobia, and the need for specialist interpretation. MRI/MRA is
optimal for patients who present in a subacute or chronic timeframe [67].
Posttraumatic brain contusions (PTBCs) represent one of the most frequent lesions
in patients with moderate or severe traumatic brain injury (TBI). PTBCs are tradi-
tionally considered primary injuries, but they have an inherent capacity to increase
3 Neurovascular Emergencies in Geriatric Patients 57
in size, generate perilesional edema, and cause mass effect [68]. These lesions are
cortical and due to the impact with the bone surfaces (e.g., the petrous bone, sphe-
noid, cribriform plate, orbit roof) and the dura mater (more resistant and irregular).
There are therefore more common localizations, such as the fronto-basal regions
and the temporal and frontal poles; they typically occur at (“coup”) or in front
(“countercoup”) with respect to the site of the blunt trauma. Bruises frequently are
multifocal and bilateral, usually involving the superficial gray matter that often
bleed, particularly those found in “countercoup” areas. The use in the acute phase is
represented almost exclusively by NCCT; however, it can underestimate the number
and size of blunt foci; there are moreover less common localizations, as is in the
case of Duret hemorrhages, generally associated with other lesions (Fig. 3.12).
Therefore, MRI in a sub-acute/chronic structural phase improves prognostic
modeling after TBI by identifying evidence of neurotrauma that may not be detected
by head CT. They are recognized as hypo dense cortical-subcortical areas and in
some cases a contextual hyper dense component of hemorrhagic significance may
be highlighted.
a b c d
e f g h
Fig. 3.12 Duret brainstem hemorrhage in posttraumatic left subdural hematoma. A 75-year-old
woman with head trauma due to sudden loss of consciousness. A CT study in ER showed an acute
left hemispherical subdural hematoma compressing the hemisphere and lateral ventricle, with
uncal hernia and incarceration of the temporal horn of the lateral ventricle; note left-to-right mid-
line shift (a, b). There is also the evidence of midbrain Duret hemorrhage (usually due to stretch-
ing/tearing of pontine perforators, c). Follow-up non-contrast CT scan after the removal of the
hematoma shows re-expansion of the left ventricular hemisystem (e, f); MRI with FLAIR (d) and
T1 TSE (g) sequences confirm the reduction of the subdural hemorrhage and of the associated
mass effect. CT follow-up after 60 days demonstrates an “ex vacuo” dilation of the horn and trine
of the left lateral ventricle (h)
58 G. M. Di Lella et al.
In patients with severe cerebral contusions, early massive edema occurs within
the period of 24–72 h post-trauma. This type of edema results in progressive eleva-
tion of intracranial pressure (ICP) and clinical deterioration giving rise to a clinical
course termed “talk-and-deteriorate” [69]. DWI measures the freedom of molecular
motion of water in tissue and is useful in identifying pathologic lesions including
foci of axonal injury and infarction. DWI is best used in conjunction with its associ-
ated ADC map, which can distinguish between cytotoxic and vasogenic edema in
the acute and subacute phases and is highly sensitive in the detection of secondary
acute ischemic infarction associated with TBI.
Despite intensive medical therapy, the elevated ICP in patients with early mas-
sive edema is often uncontrollable and fatal.
a b c d
e f g h
Fig. 3.13 Hemorrhagic brain contusions in head trauma. A 73-year-old patient with head trauma
in ASA therapy. Non-contrast CT study in ER demonstrated an ample atypical intra-axial left
frontal hemorrhage with initial surrounding edema and mass effect (a). The next day a CT angio-
graphic exam showed stability of the collection, with no signs of underlying vascular malforma-
tions (b). The subsequent MR study confirmed the diagnosis. T1-w (d) evidentiates the initial
peripheral methemoglobin ring surrounding the oxy-deoxyhemoglobin content of the fresh lesion,
while the SWI (c) sequence shows a little subarachnoid spread and absence of previous hemor-
rhages. After another light head trauma 3 month later, axial CT showed another intra-axial hema-
toma in the right posterior parietal lobe with surrounding edema (e). The subsequent MR study
with TOF sequence confirmed the diagnosis, excluding also in this case signs of vascular malfor-
mations and showing the remnants of the previous frontal hemorrhage (f–h)
compared to CT [74]. The MRI gradient echo sequence (GRE) is able to detect
heme and heme breakdown products, making it a suitable method for discovering
small hemorrhagic lesions. Susceptibility-weighted imaging (SWI) as a variant
sequence of GRE imaging should be considered the “gold standard” for identifying
TAI lesions. It has a higher sensitivity for hemorrhage than GRE, which makes it
more useful for early diagnosis of TAI. Diffusion-weighted imaging (DWI) can
accurately examine nonhemorrhagic lesions. High signal DWI can be used in
patients with early stage TAI. Lesions found represent cellular swelling and cyto-
toxic edema. DWI may aid in predicting clinical outcome after TAI. DWI is more
capable of determining the severity of the injury and estimating the long-term prog-
nosis than MRI techniques [71, 75, 76].
Diffusion tensor imaging (DTI) is an improved form of DWI. It can be used to
evaluate nerve alignment, white matter microstructure, and the morphology around
nerve fibers. Within the first 24 h after trauma, DTI can detect white matter regions
with reduced anisotropy, making it an adequate technique for detecting TAI. CT
scanning is more widely used in the acute phase, due to its much shorter scanning
60 G. M. Di Lella et al.
a b
c d
Fig. 3.14 Bilateral frontal cerebral contusions and TAI of a 65-year-old male (a–d). Non-contrast
CT in ER after close head trauma due to a car accident. The images show bilateral cortical-
subcortical inhomogeneous hypodense parenchymal areas, partially due to vasogenic edema,
directly related with the impact. Also note the multiple small hyperdense foci, more evident in (b),
which represent associated manifestations of traumatic axonal injures (TAI), secondary to the axo-
nal traumatic strain, related to the tissue deformation caused by the differential kinetic energy with
the skull
time. MRI scanning should be performed as soon as the condition of the patient
allows it, so the full extent of trauma can be mapped and white matter volume pro-
spectively followed-up [73, 77].
Acute treatment in the elderly with moderate to severe head injury involves an
acute neurosurgical approach including intracranial pressure monitoring (ICP), cra-
niotomy, and decompression craniotomy; tSAH may impair the absorption of CSF
and may produce hydrocephalus. Posttraumatic vasospasm (PTV) is a significant
3 Neurovascular Emergencies in Geriatric Patients 61
a b c
d e f
Fig. 3.15 Traumatic axonal injury (TAI). Non-contrast CT study in a 67-year-old man involved in
a high-energy head trauma shows tiny hemorrhagic foci in the subcortical white matter (c) and in
the left internal capsule (a). Blood is also present in the right lateral ventricle (b). These micro-
bleeds are typical imaging markers of diffuse axonal injury (DAI). FLAIR images well depict the
edema surrounding the tiny hemorrhagic foci in the left internal capsule (d), while the SWI (e, f)
sequence shows other hemorrhagic foci in the subcortical white matter and in the right lateral
ventricle. Note how the MRI study more accurately demonstrates the presence and the extension
of diffuse axonal damage manifestations
secondary insult to the injured brain. It typically develops between 12 h and 5 days
after the injury and lasts between 12 h and 30 days.
However, preexisting clinical conditions dramatically influence the prognosis in
elderly patients; therefore, invasive surgical treatment in this population remains
controversial.
Epidural hematoma (EDH) represents an extremely rare event in the elderly pop-
ulation: an overwhelming majority of cases arise in fact after a high energy
62 G. M. Di Lella et al.
traumatic brain injury (TBI), with associated skull fracture that involves an
artery, often represented by the middle meningeal artery (MMA). Such events
are typical of the young male population. Other causes, more frequent in the
elderly due to underlying comorbidity, are coagulopathy, secondary effect of
thrombolysis, vascular malformation, neoplasm, epidural anesthesia, or Paget
disease of skull. On the other hand, spontaneous EDHs are rare, and generally
arise from a skull primary or secondary tumor. EDHs, generally unilateral and
supratentorial, derive from an arterial bleeding in 90% of cases and therefore
show a rapid expansion, reaching the maximum size after 36 h. EDHs have the
typical shape of a biconvex lens, often also if of venous origin, are extra-axial
and determine rapid compression and displacement of the underlying brain
parenchyma with huge mass effect: thus a quick diagnosis followed by immedi-
ate surgical approach have paramount importance, in order to avoid a poor out-
come, or the death of the patients due to the brain damage. EDH of suspected
venous origin, with thickness not superior to 1 cm, could be observed with sub-
sequent TC scan in the following 36 h. Although typical in shape, imaging fea-
tures and clinical history, EDHs may pose DD with SDHs, that in some cases
may have biconvex shape: in this case it is useful to remember that EDHs, differ-
ently from SDHs, do not cross the cranial sutures. Other, less frequent DDs are
with extra-axial tumors, like meningiomas or soft component of skull or dural
primary (lymphomas, primary sarcomas) or secondary lesions. Also some infec-
tious/inflammatory event may pose a DD with EDH, epidural empyema from
skull osteomyelitis or granulomatous tubercular osseous localizations. The
NCCT exam is usually diagnostic: a second level study is not necessary and not
recommended, in order to avoid a delay of the surgical treatment, which could in
order cause a significant worsening of the outcome. EDHs appear as a thick
biconvex homogeneously hyper dense collection located under the area of the
skull trauma where, in the case of arterial origin, a fracture responsible for the
torn of an arterial vessel is almost inevitably visible (Fig. 3.16). The density may
also be low or inhomogeneous, due to the contemporary presence of uncoagu-
lated blood: in this case the collection is in the hyperacute phase. The eventual
presence of air (20%) is related to the fracture of a paranasal sinus or mastoid
proximal to the collection. A small, iso or hypodense collection is almost always
of venous origin. MRI is useful in case of nontraumatic collections. In the acute
phase, the content is hypointense in T1 and variable, hypo to hyperintense on
T2wi: in the subacute/chronic phase the T1 signal becomes hyperintense, while
in T2 it appears hypointense in the subacute period and hyperintense in the
chronic phase. The post contrast T1wi may be useful in case of venous collection
in order to demonstrate displacement and patency of dural sinuses.
AL GRAWANY
3 Neurovascular Emergencies in Geriatric Patients 63
a b c
d e f
Fig. 3.16 Epidural hematoma. Non-contrast CT study in a 71-year-old woman with head trauma
shows the classic biconvex appearance of an acute left epidural hematoma, associated with mass
effect, compression of the left hemisphere and lateral ventricle, left-to-right subfalcine herniation
and resulting ample midline shift (a–c). Note the hematoma of adjacent soft tissues (a). Axial CT
study after surgical evacuation shows the disappearance of the epidural blood collection and con-
sequently of the mass effect, with the presence of bilateral frontal pneumocephalus (d). MRI
FLAIR axial image, several days after surgery, showing the presence of tiny bilateral subdural
hematomas and a left subgaleal collection over the craniotomy (e). Bone CT 3D-reconstruction
shows a non-displaced skull fracture of the temporal and parietal bone underlying the hematoma (f)
of SDH [78–80], has increased worldwide along with the augmented percentage of
elderly in the population. Sixty percent of patients are hospitalized due to injuries
sustained in a fall, according to the National Trauma Data Bank in the United States,
55,729 (61%) had sustained injuries from falling [81]. In older patients, reduced
brain parenchyma has been associated with an increased risk of SDH, which may
even occur following minor trauma [82]. SDH usually results from tears in bridging
veins, which cross between the cerebral cortex and the dural sinus [83], or, less
frequently, a rupture of the superior cortical arteries [84]. Blood accumulates in the
space surrounding the brain parenchyma, between the arachnoid mater and the dura
[85]. Increased intracranial pressure caused by a hematoma causes further compres-
sion and damage to the brain tissue (Fig. 3.17). Moreover as the patients grow older,
there is a higher prevalence of comorbidities and increased use of medications,
including anticoagulants and polypharmacy. This can heighten the risk of bleeding
and developing further complications. Fortunately overtime there has been a
decrease in the mortality rate: in the 1990s, the mortality rate for acute SDH was
reported to be as high as 60% [79]. The mortality rate of SDH decreased to a level
of 20% around the year 2000 and has fallen as low as 14% within the last decade
64 G. M. Di Lella et al.
a b
c d
Fig. 3.17 Subdural hematoma. NCCT study in ER of a 81-year-old man with consciousness
reduction and confusion showing right hemispherical subacute subdural hematoma, with signs of
recent rebleeding, that compresses the right hemisphere and lateral ventricle, with right-to-left
subfalcine herniation, resulting in midline shift (a, b). Immediate postsurgical NCCT shows the
thickness reduction of the subdural hematoma (with presence of an air-fluid level) and the associ-
ated reduced compression on the right hemisphere and lateral ventricle (c, d)
[79]. However, acute SDH has been reported to be a poor prognostic factor for those
patients with a traumatic brain injury [86], particularly in elder patients. There was
in fact higher mortality for the elderly following falls compared to young adult
patients, after adjusting for preexisting comorbidities and severity of injury [78].
3 Neurovascular Emergencies in Geriatric Patients 65
Being an almost typical acute neurological event, the patient is generally evaluated
with a noncontrast TC study (NCTC). The recent multislice CT systems generally
available in the radiological departments being nowadays capable to acquire 64 or
more (128, 256, 320 up to 512 sub-mm slices) simultaneously, make generally pos-
sible a good quality study even in an uncooperative patient. An SDH, regardless its
phase, appears as a crescentic extra-axial collection of variable breadth and longitu-
dinal extension along the surface of the affected hemisphere. Less frequently
(15–20%), the SDH can involve both the hemispheres (Fig. 3.18), or be located in
the posterior fossa. In the hyperacute phase (less than 6 h), the subdural collection
appear hypodense or at least heterogeneous, due to the presence of mostly uncoagu-
lated blood. In the acute phase, it appears homogeneously hyperdense in 60% of
cases, while in 40% is mixed hyper-, hypodense with active bleeding (“swirl” sign),
torn arachnoid with CSF accumulation, clot retraction. Rarely the collection is
isodense, due to coagulopathy, anemia (Hgb <8–10 g/dL) ·If no new hemorrhage,
density gradually decreases to become isodense to brain parenchyma in the
a b c
d e f
Fig. 3.18 Bilateral subdural hematomas. NCCT study in ER of a 81-year-old woman with severe
headache, showing bilateral hemispherical subacute subdural hematoma (right>left): in the right,
more sample collection, there are signs of recent rebleeding, represented by the irregular areas of
hyperdensity (a). Note the bilateral compression of the cerebral hemispheres and lateral ventricles,
with right-to-left subfalcine herniation, resulting in midline shift (a–c). Follow-up CT days after
surgery showed the reduction of both the subdural hematomas and of the compression on the
hemispheres and the lateral ventricles. (f) The coronal plane shows the reduction of the right-to-left
subfalcine herniation (d–f)
66 G. M. Di Lella et al.
subacute phase. Post contrast CT is generally unnecessary in this phase, while in the
subacute period it could come in handy, because in certain case a thin SDH could be
indistinguishable from the adjacent brain cortex.
On MRI a SDH generally often follow the fashion of intraparenchymal hemor-
rhage. On TlWI in the hyper acute phase (less than 12 h) appear iso to mildly hyper
intense. In the acute (12 h to 2 days) is mildly hypo intense. On T2WI hyper acute
is mildly hyper intense. In the acute appear hypo intense. On the FLAIR sequence,
it is typically hyper intense to CSF. Signal intensity varies depending on relative Tl
and T2 effects. Acute hematomas can be isointense to CSF due to T2 shortening
effects of intracellular methemoglobin. FLAIR is often the most reliable sequence.
On T2* GRE the collection is hypo intense unless hyper acute. Finally on DWl it
shows heterogeneous signal (nonspecific), but this sequence may be useful in order
to differentiate extra axial empyema (marked central hyper intensity) from hemor-
rhage. Among the differential diagnoses it is possible to consider other subdural
collection, like hygroma, that shows clear CSF without encapsulating membranes,
subdural effusion, made by xanthochromic fluid secondary to extravasation of
plasma from membrane, that appears 1–3 days post-trauma; near CSF density/
intensity, and empyema, that generally has peripheral enhancement and shows
hyper intensity on FLAIR and restricted diffusion on DWl. The epidural hematoma
is a biconvex extra-axial collection, associated with fracture. It may cross dural
attachments, limited by sutures Pachymeningopathies (thickened dura) derives from
chronic meningitis (may be indistinguishable), neurosarcoid, with nodular, “lumpy-
bumpy” appearance, or postsurgical (e.g., shunt), caused by intracranial hypoten-
sion with “slumping” midbrain and tonsillar herniation. Tumors, like meningiomas,
lymphomas, leukemia, metastases are generally dural-based, enhancing masses,
which in some cases may involve the adjacent skull and extracranial soft tissue. In
the peripheral brain infarction, the cortex is involved, not displaced, and is typically
hyper intense on DWl.
Subacute and chronic SDH (sSDH and cSDH) are infrequently the cause of a
neurovascular emergence, due to the subtle onset and slow progression of the
symptoms, also if in some cases is possible to withstand a patient with sudden loss
of consciousness. Nevertheless the sSDH may represent a diagnostic challenge,
especially with a NCCT study; as stated above a thin crescentic collection, with-
out signs of recent rebleeding or older, chronic component, could be difficult to
diagnose from a nonexperienced radiologist. In this case the diagnosis is made
easier by administration of iodinated contrast media, eventually for other mor-
bidities, while a conventional MRI study with a FLAIR sequence never misses the
collection, albeit making sometimes necessary the differential diagnosis (DD)
with other pathologies.
The diagnosis is much more easy in case of chronic SDH, which appears clearly
hypodense on NCCT. In these case, without signs of rebleeding or presence of
membranes inside the collection, it is necessary in some cases to distinguish a cSDH
3 Neurovascular Emergencies in Geriatric Patients 67
from a hygroma: the DD could be made measuring the density of the collection
compared with that of the CSF in the ventricles. The density of a cSDH is gener-
ally higher.
a b c
d e f
Fig. 3.19 Dural sinus thrombosis. A 74-year-old man hospitalized in a COVID+ intensive care
unit for sudden and prolonged loss of consciousness. CT axial and ACT study showed an inhomo-
geneous hemorrhage with initial surrounding vasogenic edema in the left temporal lobe; the angio-
CT demonstrated thrombosis of left transverse and sigmoid sinuses, with involvement of the
homolateral Labbè vein and intracranial tract of the jugular vein (a–c). Follow-up MRI with 3D
TOF angio-sequences confirmed the involvement of the venous structures, with extension to the
extracranial proximal left jugular vein. Also note the enlargement of the vasogenic edema sur-
rounding the hemorrhage (d–f)
References
1. Heron M. Deaths: leading causes for 2017. Natl Vital Stat Rep. 2019;68(6):1–77.
2. Howard G, Goff DC. Population shifts and the future of stroke: forecasts of the future burden
of stroke. Ann N Y Acad Sci. 2012;1268:14–20. https://doi.org/10.1111/j.1749.
3. Saposnik G, Black S. Stroke in the very elderly: hospital care, case fatality and disposition.
Cerebrovasc Dis. 2009;27(6):537–43. https://doi.org/10.1159/000214216.
4. Singer J, Gustafson D, Cummings C, Egelko A, Mlabasati J, Conigliaro A, Levine
SR. Independent ischemic stroke risk factors in older Americans: a systematic review. Aging.
2019;11(10):3392–407. https://doi.org/10.18632/aging.101987.
5. El-Koussy M, Schroth G, Brekenfeld C, Arnold C. Imaging of acute ischemic stroke. Eur
Neurol. 2014;72(5-6):309–16. https://doi.org/10.1159/000362719.
6. Eastwood JD, Lev MH, Provenzale JM. Correlation of early dynamic CT perfusion imag-
ing with whole-brain MR diffusion and perfusion imaging in acute hemispheric stroke.
AJNR. 2003;24:1869–75.
3 Neurovascular Emergencies in Geriatric Patients 69
29. Khosravani H, Mayer SA, Demchuk A, Jahromi BS, Gladstone DJ, Flaherty M, et al.
Emergency noninvasive angiography for acute intracerebral hemorrhage. AJNR Am J
Neuroradiol. 2013;34:1481–7.
30. Wardlaw JM, Smith EE, Biessels GJ, et al. Neuroimaging standards for research into
small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol.
2013;12(8):822–38.
31. Banerjee G, Carare R, Cordonnier C, et al. The increasing impact of cerebral amyloid
angiopathy: essential new insights for clinical practice. J Neurol Neurosurg Psychiatry.
2017;88(11):982–94. Focused review on cerebral amyloid angiopathy.
32. Hilkens NA, van Asch CJJ, Werring DJ, et al. Predicting the presence of macrovascular
causes in non-traumatic intracerebral haemorrhage: the DIAGRAM prediction score. J Neurol
Neurosurg Psychiatry. 2018;89(7):674–9.
33. Hemphill JC, Greenberg SM, Anderson C. Guidelines for the management of spontaneous
intracerebral hemorrhage: a guideline for healthcare professionals from the American Heart
Association/American Stroke Association. Stroke. 2015;46(7):2032–60.
34. Caplan LR, Searls DE, Hon FKS. Cerebrovascular disease. Med Clin North Am.
2009;93(2):353–69. https://doi.org/10.1016/j.mcna.2008.09.004.
35. de Rooij NK, Linn FH, van der Plas JA, Algra A, Rinkel GJ. Incidence of subarachnoid haem-
orrhage: a systematic review with emphasis on region, age, gender and time trends. J Neurol
Neurosurg Psychiatry. 2007;78:1365–72.
36. Fortuny LA, Adams CB, Briggs M. Surgical mortality in an aneurysm population: effects
of age, blood pressure and preoperative neurological state. J Neurol Neurosurg Psychiatry.
1980;43:879–82.
37. Pobereskin LH. Incidence and outcome of subarachnoid haemorrhage: a retrospective popula-
tion based study. J Neurol Neurosurg Psychiatry. 2001;70:340–3.
38. Thom T, Haase N, Rosamond W, et al. Heart disease and stroke statistics—2006 update: a
report from the American Heart Association statistics committee and stroke statistics subcom-
mittee. Circulation. 2006;113:e85–151.
39. Schöller K, Massmann M, Markl G, Kunz M, et al. Aneurysmal subarachnoid hemorrhage in
elderly patients: long-term outcome and prognostic factors in an interdisciplinary treatment
approach. J Neurol. 2013;260(4):1052–60. https://doi.org/10.1007/s00415-012-6758-1.
40. Ostergaard JR. Headache as a warning symptom of impending aneurismal subarachnoid haem-
orrhage. Cephalalgia. 1991;11(1):53–5.
41. van Gijn J, Rinkel GJ. Subarachnoid haemorrhage: diagnosis, causes and management. Brain.
2001;124:249–78.
42. Hostettler IC, Seiffge DJ, Werring DJ. Intracerebral hemorrhage: an update on diagnosis and
treatment. Expert Rev Neurother. 2019;19(7):679–94.
43. Szepesi R, Szell IK, Hortobagyi T, et al. New prognostic score for the prediction of 30-day out-
come in spontaneous supratentorial cerebral haemorrhage. Biomed Res Int. 2015;2015:961085.
44. Fiebach JB, Schellinger PD, Gass A, et al. Stroke magnetic resonance imaging is accurate in
hyperacute intracerebral hemorrhage: a multicenter study on the validity of stroke imaging.
Stroke. 2004;35:502–6.
45. Mitchell P, Wilkinson ID, Hoggard N, et al. Detection of subarachnoid haemorrhage with
magnetic resonance imaging. J Neurol Neurosurg Psychiatry. 2001;70:205–11.
46. Papke K, Kuhl CK, Fruth M, Haupt C, Schlunz-Hendann M, Sauner D, et al. Intracranial aneu-
rysms: role of multidetector CT angiography in diagnosis and endovascular therapy planning.
Radiology. 2007;244(2):532–40.
47. Wintermark M, Uske A, Chalaron M, et al. Multislice computerized tomography angiography
in the evaluation of intracranial aneurysms: a comparison with intraarterial digital subtraction
angiography. J Neurosurg. 2003;98:828–36.
48. Tetri S, Juvela S, Saloheimo P, et al. Hypertension and diabetes as predictors of early death
after spontaneous intracerebral hemorrhage. J Neurosurg. 2009;110(3):411–7.
49. Garbossa D, Panciani PP, Fornaro R, Crobeddu E, Marengo N, Fronda C, Ducati A, Bergui M,
Fontanella M. Subarachnoid hemorrhage in elderly: advantages of the endovascular treatment.
Geriatr Gerontol Int. 2011;12(1):46–9.
3 Neurovascular Emergencies in Geriatric Patients 71
50. Nieuwkamp DJ. Subarachnoid haemorrhage in patients >=75 years: clinical course, treatment
and outcome. J Neurol Neurosurg Psychiatry. 2006;77(8):933–7. https://doi.org/10.1136/
jnnp.2005.0843.
51. Gardner R, Dams-O’Connor K, Morrissey MR, Manley GT. Geriatric traumatic brain
injury: epidemiology, outcomes, knowledge gasp and future directions. J Neurotrauma.
2018;35(7):889–906. https://doi.org/10.1089/neu.2017.5371.
52. Kameyama M, Karibe H, Kawase M, Hayashi T, Hirano T, Tominaga T. Severe head injury
and age in Japan Neurotrauma Data Bank: comparison among project 1998, 2004, and 2009.
Neurotraumatology. 2013;36:10–6.
53. Timler D, Dworzyński MJ, Szarpak Ł, Gaszyńska E, Dudek K, Gałązkowski R. Head
trauma in elderly patients: mechanisms of injuries and CT findings. Adv Clin Exp Med.
2015;24(6):1045–50. https://doi.org/10.17219/acem/27565.
54. Styrke J, Stalnacke BM, Sojka P, Bjornstig U. Traumatic brain injuries in a well-defined popu-
lation: epidemiological aspects and severity. J. Neurotrauma. 2007;24:1425–36.
55. Eisenberg HM, Gary HE Jr, Aldrich EF, Saydjari C, Turner B, Foulkes MA, Jane JA, Marmarou
A, Marshall LF, Young HF. Initial CT findings in 753 patients with severe head injury. A report
from the NIH Traumatic Coma Data Bank. J Neurosurg. 1990 ; 73(5):688-698.
56. Jane JA, Francel PC. Age and outcome of head injury. In: Narayan RK, Wilberger Jr JE,
Povlishock JT, editors. Neurotrauma. New York: McGraw-Hill; 1996. p. 793–804.
57. Tokutomi T, Ogawa T, Ono J, et al. Intracranial diagnosis according to the Traumatic Coma
Data Bank classification of computed tomography imaging in the Japan Neurotrauma Data
Bank. Neurotraumatology. 2005;28:1–5.
58. Smits M, Dippel DWJ, et al. External validation of the Canadian CT head rule
and the New Orleans Criteria for CT scanning in patients with minor head injury.
JAMA. 2005;294(12):1511–8.
59. Guide de bon usage des examens d’imagerie. http://www.sfrnet.org/sfr/professionnels/5-
referentiels-b onnes-p ratiques/guides/guide-b onusage-e xamens-i magerie-m edicale/
index.phtml.
60. Pages P-J, Boncoeur-Martel M-P, Dalmay F, Salle H, Caire F, Mounayer C, Rouchaud
A. Relevance of emergency head CT scan for fall in the elderly person. J Neuroradiol.
2020;47(1):54–8. https://doi.org/10.1016/j.neurad.2019.03.004.
61. Mutch C, Talbott J, Gean A. Imaging evaluation of acute traumatic brain injury. Neurosurg
Clin N Am. 2016;27(4):409–39. https://doi.org/10.1016/j.nec.2016.05.011.
62. Modi NJ, Agrawal M, Sinha VD. Post-traumatic subarachnoid hemorrhage: a review. Neurol
India. 2016;64(Suppl):S8–S13. https://doi.org/10.4103/0028-3886.178030.
63. Gauvrit J-Y, Leclerc X, Ferré J-C, Taschner C-A, Carsin-Nicol B, Auffray-Calvier E, Morandi
X, Carsin M. Imagerie de l’hémorragie sous-arachnoïdienne. J Neuroradiol. 2009;36(2):65–73.
https://doi.org/10.1016/j.neurad.2008.06.005.
64. Edlow JA, Caplan LR. Avoiding pitfalls in the diagnosis of subarachnoid hemorrhage. N Engl
J Med. 2000;342(1):29–36. https://doi.org/10.1056/nejm200001063420106.
65. Agrawal M, Modi N, Sinha VD. Neurological outcome in patients of traumatic subarachnoid
haemorrhage: a study of prognostic factors and role of MRI. IJNT2014;11:10–6.
66. Boesiger BM, Shiber JR. Subarachnoid hemorrhage diagnosis by computed tomography and
lumbar puncture: are fifth generation CT scanners better at identifying subarachnoid hemor-
rhage ? J Emerg Med. 2005;29:23–7.
67. Long B, Koyfman A, Runyon MS. Subarachnoid hemorrhage. Emerg Med Clin North Am.
2017;35(4):803–24. https://doi.org/10.1016/j.emc.2017.07.001.
68. Martinez-valverde T, Vidal-jorge M, Marrtinez-Saez E, et al. Sulfonylurea receptor 1 in
humans with post-traumatic brain contusion. J Neurotrauma. 2015;32(19):1478–87. https://
doi.org/10.1089/neu.2014.3706. Epub 2015 Jun 3.
69. Katayama Y, Tsubokawa T, Miyazaki S, Kawamata T, Yoshino A. Oedema fluid formation
within contused brain tissue as a cause of medically uncontrollable elevation of intracranial
pressure in head trauma patients. Acta Neurochir. 1990;s51:308–10.
70. Gentry LR. Imaging of closed head injury. Radiology. 1994;191(1):1–17.
72 G. M. Di Lella et al.
71. Bruggeman GF, Haitsma IK, Dirven CMF, Volovic V. Traumatic axonal injury (TAI): defini-
tions, pathophysiology and imaging a narrative review. Acta Neurochir. 2021;163:31–44.
72. Gennarelli TA, Thibault LE, Adams JH, Graham DI, Thompson CJ, Marcincin RP. Diffuse
axonal injury and traumatic coma in the primate. Ann Neurol. 1982;12:564–74.
73. Ma J, Zhang K, Wang Z, Chen G. Progress of research on diffuse axonal injury after traumatic
brain injury. Neural Plast. 2016;2016:9746313. https://doi.org/10.1155/2016/9746313.
74. Besenski N. Traumatic injuries: imaging of head injuries. Eur Radiol. 2002;12:1237–52.
75. Takayama H, Kobayashi M, Sugishita M, Mihara B. Diffusion-weighted imaging demon-
strates transient cytotoxic edema involving the corpus callosum in a patient with diffuse brain
injury. Clin Neurol Neurosurg. 2000;102:135–9.
76. Ezaki Y, Tsutsumi K, Morikawa M, Nagata I. Role of diffusion-weighted magnetic resonance
imaging in diffuse axonal injury. Acta Radiol. 2006;47:733–40.
77. Arfanakis K, Haughton VM, Carew JD, Rogers BP, Dempsey RJ, Meyerand ME. Diffusion
tensor MR imaging in diffuse axonal injury. AJNR Am J Neuroradiol. 2002;23:794–802.
78. Rau CS, Lin TS, Wu SC, Yang JC, Hsu SY, Cho TY, Hsieh CH. Geriatric hospitalizations in
fall-related injuries. Scand J Trauma Resusc Emerg Med. 2014;22:63. https://doi.org/10.1186/
s13049-014-0063-1.
79. Ryan CG, Thompson RE, Temkin NR, Crane PK, Ellenbogen RG, Elmore JG. Acute trau-
matic subdural hematoma: current mortality and functional outcomes in adult patients at a
Level I trauma center. J Trauma Acute Care Surg. 2012;73:1348–54. https://doi.org/10.1097/
TA.0b013e31826fcb30.
80. Leitgeb J, Mauritz W, Brazinova A, Janciak I, Majdan M, Wilbacher I, Rusnak M. Outcome
after severe brain trauma due to acute subdural hematoma. J Neurosurg. 2012;117:324–33.
https://doi.org/10.3171/2012.4.JNS111448.
81. Hsieh C-H, Rau C-S, Shao-Chun W, Liu H-T, Huang C-Y, Hsu S-Y, Hsieh H-Y. Risk factors
contributing to higher mortality rates in elderly patients with acute traumatic subdural hema-
toma sustained in a fall: a cross-sectional analysis using registered trauma data. Int J Environ
Res Public Health. 2018;15(11):2426.
82. Ledic D, Girotto D, Pal S, Kolbah B. Risk factors for subdural bleeding in elderly population.
Coll Antropol. 2014;38:1195–8.
83. Shen J, Pan JW, Fan ZX, Zhou YQ, Chen Z, Zhan RY. Surgery for contralateral acute epidural
hematoma following acute subdural hematoma evacuation: five new cases and a short litera-
ture review. Acta Neurochir. 2013;155:335–41. https://doi.org/10.1007/s00701-012-1569-9.
84. Maxeiner H, Wolff M. Pure subdural hematomas: a postmortem analysis of their form and
bleeding points. Neurosurgery. 2002;50:503–8; discussion 508–9.
85. Chisholm KM, Harruff RC. Elderly deaths due to ground-level falls. Am J Forensic Med
Pathol. 2010;31:350–4. https://doi.org/10.1097/PAF.0b013e3181f69c87.
86. Anandasivam NS, Russo GS, Samuel AM, Grant R, Bohl DD, Grauer JN. Injuries associ-
ated with subdural hematoma: a study of the National Trauma Data Bank. Conn Med.
2017;81:215–22.
87. Nasr DM, et al. Mortality in cerebral venous thrombosis: results from national inpatient sam-
ple database. Cerebrovasc Dis. 2013;35(1):40–4.
88. Connor SEJ, Jarosz JMJ. Magnetic resonance imaging of cerebral venous sinus thrombosis.
Clin Radiol. 2002;57(6):449–61.
89. Coutinho JM, Zuurbier SM, Aramideh M, Stam J. The incidence of cerebral venous thrombo-
sis: a cross-sectional study. Stroke. 2012;43(12):3375–7.
AL GRAWANY
Head and Neck in Geriatric Patients
4
T. Popolizio, L. Cassano, A. Pennelli, R. Izzo, G. Fascia,
M. Masciavè, and Giuseppe Guglielmi
Learning Objectives
After reading this chapter, readers should be able to:
4.1 Ear
Chronic suppurative otitis media is a very common disease that should be carefully
treated, as severe complications can develop. Despite the significantly decreased
incidence of chronic suppurative otitis media related complications since the
introduction of antibiotics, this clinical problem has not been eliminated. Chronic
suppurative otitis media remains a serious concern, particularly in developing coun-
tries and socioeconomically poor regions [1–4].
Complications of CSOM can be classed as extracranial (EC) or intracranial (IC).
Extracranial complications include mastoid abscess, petrositis, labyrinthitis, facial
nerve paralysis (FNP), and Bezold’s abscess. Intracranial complications comprise
intracranial abscess (including extradural, epidural, subdural, perisigmoid sinus,
and brain abscesses), lateral sinus thrombophlebitis (LST), meningitis, and otitic
hydrocephalus. The pathophysiology of complications of CSOM remains some-
what of a mystery. The pathways of EC and IC complications include thrombophle-
bitis of the venules of the adjoining cranial bones, bone erosion by pressure or
enzymatic actions, preformed pathways, and hematogenous spread [5–8].
Coalescent Mastoiditis
The treatment for acute mastoiditis can fail and sometimes there is an enzymatic
destruction of the mastoid septa and the development of an intramastoid empyema.
TC can visualize these erosive changes very early [9, 10].
Subperiosteal Abscess
It typically occurs via direct extension of the inflammatory debris through a defect
in the external context of the mastoid sinus. This passage can occur in any direction:
post-auricular, common as the bone is particularly thin (Macewan’s triangle) or
infero-medial, medial to the attachment of sternocleidomastoid (resulting in a
Bezold’s abscess) [11].
Bezold’s abscess is comparable to the subperiosteal abscess but takes place
through a bony defect at the mastoid tip, medial to the insertion of the posterior
belly of digastric.
CT can demonstrate coalescent mastoiditis with erosion of the medial mastoid
tip [12].
This defect could let infection expand into the neck.
CT also shows thickening of the omolateral sternocleidomastoid muscles with
surrounding inflammatory changes, heterogeneous post contrast enhancement with
rim enhancing cystic lesion [13, 14].
Petrositis
It is an infection with involvement of bone at petrous apex of the temporal bone.
CT is the modality of choice to evaluate bony changes in the temporal bone,
revealing debris within the petrous apex, air cells, and erosive lysis of bony septa
[15–18].
Disruption of the anterior or posterior bony cortex may occur resulting in fulmi-
nant intracranial complication (meningitis, empyema, dural sinus thrombosis, cra-
nial neuropathy).
4 Head and Neck in Geriatric Patients 75
At the onset diagnosis of petrus, apex inflammatory disease is best made with
high-resolution CT. Later MR imaging becomes important to assess intracranial
complications [19, 20].
Labyrinthitis
The acute stage results when bacteria and other pathogenic noxae filled the perilym-
phatic spaces, inducing acute inflammatory response.
The CT is normal at this stage, and the endolymphatic space is spared.
The majority of patients will not have enhancement or any imaging findings.
Sometimes there is enhancement of the normally not enhanced fluid-filled spaces of
the labyrinth on T1-weighted images [21, 22].
The enhancement may persist long after symptoms, there is an accumulation of
gadolinium within inflamed labyrinthine membrane because of the breakdown of
the labyrinthine vasculature [10].
The combination of pathological labyrinthine enhancement and thickening/
enhancement of the seventh nerve should induce suspicion of EAC [23, 24].
If acute labyrinthitis does not resolve, the progression to chronic disease results
first in fibrous change and then in ossification.
The fibrous stage is characterized by fibroblastic proliferation within the peri-
lymphatic spaces (begins after 2 weeks after the onset of infection). CT is normal
while T2-weighed MR images show replacement of the normal high signal of the
fluid-filled spaces of the labyrinth, detected at the cochlear apex [25, 26].
The ossify stage consists of pathological ossification of the membranous laby-
rinth [27, 28].
The CT appearance is characterized by high-density bone deposition within the
membranous labyrinth, from hazy increase in density within fluid spaces of the
membranous labyrinth (mild disease) or focal areas of bony invasion on fluid spaces
(moderate disease) or with total obliteration by bony replacing fluid spaces (severe
disease) [29, 30].
MRI will show loss of normal signal fluid in membranous labyrinth in
T2-weighted images as hypointense foci in labyrinth.
of the mastoid cells and reducing the vascular perfusion of the mucosa and decreases
the tissue penetration of antibiotics [32]. The inflammatory process on the facial
nerve canal, through canal dehiscence or invasion of infectious microorganisms,
results in inflammation and edema of the nerve inside its canal. The venous return
decreases and the pressure increases on the nerve, which leads to nerve dysfunction.
As the limits of the facial nerve canal are narrow, the accumulation of purulent
secretion inside the canal leads to mechanical compression and ischemic neuritis.
Persistent inflammation of the middle ear can also cause dehiscence of the facial
nerve canal, which leads to consequent facial paralysis [33].
Gradenigo’s Syndrome
It consists of the triad: suppurative otitis media, abducent nerve palsy, retroorbital
pain due to the extension of inflammation into Meckel cave. In patients with sup-
purative otis media infection may spread to the petrous apex of the temporal bone
and may be via pneumatized air cell tracts, through vascular channels or as a result
of direct extension through fascial planes. Abducent nerve is close to the petrous
apex separated from it only by dura mater, lies medial and adjacent to the trigeminal
ganglion, passing through Dorello canal. Extradural inflammation secondary to api-
cal petrositis may affect the above structures and generate the symptoms of
Gradenigo’s syndrome [34–40].
Sinus Thrombophlebitis
Sinus thrombophlebitis is caused by spread of infection to the inner wall of the
venous sinus, leading to the formation of a thrombus through the action of fibrin and
platelets, a process called endophlebitis [46].
4 Head and Neck in Geriatric Patients 77
a b
Fig. 4.1 Necrotizing external otitis. Meningo-encephalitis complication. Post-contrast T1-w cor-
onal (a) and axial (b): high contrast enhancement of right middle ear and mastoid with focal
thickening of the meningeal sheets in the middle crania fossa close to the petrous bone. In the late
sequences (b) a small area of vivid contrast enhancement with blurred borders can be appreciated
in the adjoining brain parenchyma. Findings are suggestive for meningo-encephalitis. NCCT coro-
nal MPR reconstruction (c): the right residual middle ear is filled with homogeneously hypodense
tissue. No erosions of the epitympanic recess are evident. The stapes shows some bone changes.
The tegmen tympani is eroded. The superior semicircular canal is not well depicted. On the left, a
small amount of homogeneous hypodense tissue is attached to the cochlear promontory
78 T. Popolizio et al.
Alterations in sensory functions, vision, balance, and hearing are some of the most
common disturbances seen in the aging population and lead to dramatic social and
4 Head and Neck in Geriatric Patients 79
a b
Fig. 4.2 Necrotizing external otitis. Carotid stenosis complication. Axial (a) CT and coronal (b)
CT images show on the right side erosion of the glenoid joint, the jaw condyle, the apex of the
petrous part of the temporal bone, the carotid canal, and clivus. Hypodense tissue fills the middle
ear and mastoid cavities. There are no signs of bone or ossicular chain erosion. Post-contrast T1-w
MRI (c and d) shows avid enhancement of temporo-mandibular joint that extends to the petrous
part of the temporal bone, clivus pterygoid space, skull base, and cavernous sinus. Encasement of
the intrapetrous segments of the internal carotid artery can also be appreciated, which appear ste-
notic as depicted in the 3D MRI TOF (e). Also, the lesion expands through the masticatory space,
engaging the lateral pterygoid muscles, and the prevertebral space involving the posterior wall of
rhinopharynx, which is swelled. It focally overpasses the midline affecting the left lateral ptery-
goid muscle. Autologous 99mTc-HMPAO-labeled leukocyte scintigraphy (f) shows accumulation
in the right mastoid bone, skull base, rhinopharynx, and clivus
80 T. Popolizio et al.
functional disability. Among the senses affected by increasing age, hearing loss is
the most common. Presbycusis, or age-related hearing loss (ARHL), is a term that
refers to hearing loss as a result of physiologic and pathologic changes associated
with increasing age. As the aging population continues to grow, greater focus is
placed on understanding and attempting to reverse this sensory loss for the benefit
of geriatric patients. Today, there is an established although still evolving concept of
the workings of the outer ear, middle ear, and inner ear.
Presbycusis may present insidiously and be confounded by various medical, psy-
chological, and pharmacologic factors. Only after thorough history, examination,
and audiological testing can a diagnosis of presbycusis be made after excluding
concurrent medical and pharmacologic effects. In general, the first signs of ARHL
can be seen in late middle age with high-frequency hearing losses in the realm of
conversation frequencies, ultimately progressing subtly to lower frequency tones.
The range of human auditory frequencies spans 20–20,000 Hz, with speech fre-
quencies ranging from 400 to 5000 Hz, with the greatest loss in hearing seen in
frequencies greater than or equal to 2000 Hz. The loss of this linguistic information
results in many of the complaints in presbycusis. The loss of meaning is seen in
deterioration of speech intelligibility, the loss of clear separation between words
results in speech sounding mumbled, and the loss of syllables causes difficulty dis-
cerning similarly sounding words. Patients with presbycusis rely on conversational,
emotional, and postural context clues to compensate for their hearing impairment,
requiring a greater amount of higher order cognitive functioning to understand daily
conversations.
a c
Fig. 4.3 External auditory canal exostoses—hearing loss. NCCT Ax (a) and Cor (b): broad-based
and focal circumferential bony overgrowth of the osseous external auditory canal causing stenosis
of the left auditory canal. Stenosis external acoustic. X-ray (c) of implant shows how well it is the
curve of the skull
MRI can visualize the fluid content of the membranous labyrinth. Visualization
of the vestibulocochlear nerve in the fluid-filled internal auditory canal and cerebel-
lopontine angle is only possible by the MRI [66, 67].
Postoperative imaging is required when a malfunction of the device is suspected
[63]. However, the authors perform—and recommend to do so—a postoperative
examination in every patient to confirm the correct position of the implant electrode.
Information regarding basal electrode location helps improving programming accu-
racy, associated frequency allocation, and audibility with appropriate deactivation
of extracochlear electrodes [68, 69].
Postoperative CT has turned out to be useful in visualizing position of the elec-
trode array by using HRCT or cone-beam computed tomography (CBCT).
Successful cochlear implantation requires that the electrode be confined to the scala
tympani. In general, CBCT is associated with lower dose and less metal artifacts
when compared to HRCT (Fig. 4.3) [70].
4.2 Nose
4.2.1 Rhinosinusitis
Based on the Medical Expenditure Panel Survey for 2007, which concerned
225.1 million Americans, Bhattacharyya estimated the prevalence of chronic rhino-
sinusitis (CRS) (with nasal polyps or without nasal polyps) at 0.2% or 490/10,000
AL GRAWANY
4 Head and Neck in Geriatric Patients 83
• An increase in the volume and a decrease in the elasticity of the nasal mucosa
[75, 76]
• A reduced or absent nasal cycle, partly due to a declining ciliary efficacy
• Atrophy of the supporting fibro fatty tissues of the nose, with a potential loss of
support for the nasal structures (narrowing of the nasal valve), which gives rise
to more nasal obstruction
• A higher incidence of rhinorrhea with more mucus due to increased glandular
activity and more viscous secretions and excess mucus crusting [77]
a b
c d
Fig. 4.4 Acute rhinosinusitis. NCCT bone algorithm, axial (a) and coronal MPR (b). The maxil-
lary sinus is filled with hypodense tissue that also involves the upper and middle ethmoid cells and
the sphenoid sinus. Focal interruptions of lamina papyracea, cribriform plate, orbital floor, and the
maxillary bone are evident. Reticulation of the medial intraorbitary fat tissue is present suggesting
involvement. On post-contrast T1-w MRI, axial (c) and coronal (d) images, the pathologic tissue
has low signal on T1-w and intermediate on T2-w (not shown), fuzzy margins and determines
structural bone changes and focal interruptions. Internal fluid components are present, which show
proton diffusion restriction on DWI (not shown), compatible with abscess. Also, there is involve-
ment of pterygopalatine fossa, infratemporal fossa, lateral pterygoid muscle, the choana, and the
let nasal cavities
Sinonasal inverted papilloma (IP) is one of the most common benign epithelial
tumors of the nose and paranasal sinuses. It accounts for 0.5–4% of all nasal tumors,
with a male/female ratio of 2–4:1.1 It originates from the Schneiderian membrane
that lines both nasal and paranasal areas. The invagination of such epithelial
4 Head and Neck in Geriatric Patients 85
membrane within the submucosal stroma is the typical histological aspect of this
tumor. The age at onset varies, but it is mostly encountered between the fifth and
sixth decades of life. Although the precise etiology is not clear, several external fac-
tors and a relationship with some subtypes of the human papilloma virus are reported
in almost 40% of patients. It has been suggested that sinonasal IP can progress to
squamous cell carcinoma; some recent articles stated that alteration of cell cycle–
related proteins may contribute to the malignant transformation from IP to squa-
mous cell carcinoma. As initially reported, the origins of tumor were observed in the
lateral nasal wall (82%), maxillary sinus (53.9%), ethmoid sinus (31.6%), frontal
sinus (6.5%), and sphenoid sinus (3.9%) [83–85].
The proposed treatment for IP has always been a radical surgical removal based
on the recurrence rates and the possibility of malignant transformation/association
with malignant lesions. As stated in the literature, we confirmed that the tumor pre-
sented a pedicle and a single site of attachment. The research of the pedicle’s attach-
ment is facilitated by radiological examination because, according to several recent
articles, the site of tumor attachment can be frequently predicted by both computed
tomography (CT) and magnetic resonance imaging (MRI) scans. Moreover, in most
cases, the tumor’s pedicle and the site of attachment can be accurately unveiled dur-
ing surgery, and the tumor’s extension can be precisely studied [86, 87].
Even though combined CT and MRI are useful for preoperative assessment of
sinonasal IP, differentiation of IP from other malignant sinonasal tumors is often
difficult because the overlap of imaging features. CT demonstrates soft tissue den-
sity mass with enhancement. The location of the mass leads toward the correct diag-
nosis. As the mass enlarges, it results in bony remodeling and resorption [85, 88, 89].
In 40% of cases, intralesional calcifications can be observed, representing resid-
ual bone fragments. The presence of focal hyperostosis has been correlated to the
point of origin of the lesions [90].
MRI often demonstrates a distinctive gross mucosal morphology of IP, called
convoluted cerebriform pattern (CCP), a “striated” imaging, seen on both T2 and
contrast-enhanced T1-weighted images, with characteristic alternating hypointense
and hyperintense bands.
In T1-weighted images, it appears isointense to muscle, in T2 generally hyperin-
tense to muscle with alternating lines. In roughly 50% of cases IP enhance, the
lesions are heterogeneous with alternating hypointense and hyperintense bands
[91, 92].
The presence of central necrosis requires consideration of an associated malig-
nancy [93–96].
IP can show an aggressive pattern of bone destruction because it may cross the
cribriform plate into the cranial anterior fossa.
They can erode the skull base comparable to aggressive cancer and because of
this signal intensity characteristics place on those of malignancies [97, 98].
Recurrences may be distinguished from postoperative thickening by dynamic
enhanced MRI because they have earlier and greater enhancement than granulation
tissue (Fig. 4.5) [99].
86 T. Popolizio et al.
4.2.3 Dacryocystitis
a b
c d
Fig. 4.5 Inverted papilloma. NCCT axial (a) and coronal MPR reconstruction (b): expansive
dense mass in the left maxillary sinus erodes the lamina papyracea and invades the nasal fossa.
There are signs of bone remodeling (thinning and bowing) and resorption. The cribriform plate is
intact. On MRI, the lesion has low signal on T1-w (c), axial (d), and coronal (e) T2-w images
showing a “striated pattern” sign. On post-contrast T1-w MRI image (f), the lesion shows periph-
eral enhancement. DWI coronal image (g) shows restricted diffusion
4 Head and Neck in Geriatric Patients 87
e f
highlights the role of the interprofessional team in the care of patients with this
condition [100, 101].
The etiology of acquired dacryostenosis is multifactorial and is not fully under-
stood. Some cases may be related to trauma, neoplasm, systemic disease, radio-
therapy, or chemotherapy. However, in most cases, the cause is “involutional” and
classified as “idiopathic.” Some authors have reported that the cause is secondary to
anatomic changes in the diameter of the bony lacrimal canal, which occurs with
aging. Women, in particular, have a smaller diameter of the lacrimal duct that tends
to narrow with time. A congenital narrowness within the lacrimal drainage system
is generally regarded as a disposition for lacrimal stenosis [102].
88 T. Popolizio et al.
Some authors suggest that the cause may be from ascending inflammation from
the region of the nose and sinus cavities. A descending infection from the conjunc-
tiva has also been suggested as a cause of acquired dacryostenosis. Clinical studies
indicate that nasal disease is sporadic in patients undergoing DCR [103].
Familial predisposition and osteoporotic changes have also been suggested as
being predisposing factors.
The obstruction is more frequently situated at the level of the nasolacrimal duct
or puncta and less frequently at the level of the canaliculi. The incidence is higher
among older people and in women [104].
Diagnosis is usually made clinically; however, imaging may help to exclude
complications. It is important distinguishing between acute and chronic dacryocys-
titis [100].
Acute dacryocystitis is commonly associated with preseptal cellulitis.
Complications include orbital cellulitis (limited to preseptal tissues), corneal
involvement, lacrimal sac mucocele and, rarely orbital abscess. The most common
organisms implicated are Staphylococcus aureus in acquired cases and S. pneumo-
nia in congenital cases although cultures and smears expressed punctual secretions
as desirable [105].
Chronic dacryocystitis is a result of chronic obstruction due to systemic disease,
repeated infection, dacryoliths, and chronic inflammatory debris of the nasolacrimal
system. Some common systemic diseases include Wegener’s granulomatosis, sar-
coidosis, and systemic lupus erythematosus.
Acquired states are typically due to repeated trauma, surgeries, medications, and
neoplasms. Among traumatic causes of nasolacrimal obstruction, nasoethmoid frac-
tures seem to be most common.
Imaging features pointing to acute dacryocystitis include thick rim enhancement
and extensive adjacent soft tissue preseptal cellulitis.
MRI is the imaging modality of choice in the evaluation of orbital cellulitis
because of its superior soft tissue and contrast resolution. It is essential to evaluate
the extent of the orbital infection, underlying paranasal sinus involvement, as well
as detect complications of orbital cellulitis, especially intracranial spread. Orbital
cellulitis causes diffuse, edematous infiltration of the orbital connective tissue that
is best demonstrated by the high signal intensity in T2-weighted fat-saturated
sequences. Other findings are swelling and ill-defined margins of the extraocular
muscles and exophthalmos. Orbital cellulitis may be complicated by an abscess,
which may form in the extraconal or intraconal orbit separate from the bone.
Periorbital cellulitis is a preseptal process, which is limited to the soft tissues
anterior to the orbital septum. It usually occurs due to the contiguous spread of
infection from adjacent structures such as the teeth and face. Computed tomography
and MRI demonstrate diffuse soft tissue thickening anterior to the orbital septum.
Infection in orbit, whether as a result of periorbital cellulitis extending across the
orbital septum or due to sinusitis, constitutes an emergency.
MRI is better than CT, in fact it provides excellent contrast resolution in the orbit
with the demonstration of pathologies in the intraconal and extraconal compart-
ments. The ability to depict cross-sectional anatomy and pathology with better tis-
sue characterization and even without administering intravenous gadolinium-based
4 Head and Neck in Geriatric Patients 89
a b
Fig. 4.6 Dacryocystitis. NCCT Ax (a), e Cor MPR reconstruction (b). Well-circumscribed round
lesions with fluid core around the inner canthus, with adjacent soft tissue thickening and fat strand-
ing, scalp melanoma is noticed
Cutaneous squamous cell carcinoma (cSCC) accounts for approximately 20% of all
non-melanoma skin cancers, which is the most common malignancy worldwide.
Although less than 5% of head and neck cSCC (HNcSCC) metastasize, lymph
node metastases in the parotid and/or neck are potentially lethal and require morbid
multimodal regional therapy with surgery and adjuvant radiotherapy (RT). Although
the fundamental treatment approach has remained largely unchanged, there have
been several advances that may impact survival.
There have been very few studies examining trends in survival of HNcSCC, par-
ticularly metastatic HNcSCC. Gnanasekaran et al. recently examined the trends in
prognosis of patients with metastatic HNcSCC over the last 30 years within a single
Australian institution. The authors reported improved cancer-specific survival over
90 T. Popolizio et al.
time despite treating increasing numbers of elderly patients and more aggressive
cancers.
The radiologist’s roles include evaluating the full local extent of the primary,
detecting perineural tumor, and assessing regional nodal and distant spread of disease.
CT scanning or MRI can be helpful in defining the extent of disease. CT scan-
ning is useful for determining the presence of bone or soft tissue invasion and for
evaluating cervical lymph nodes at risk for metastasis.
cSCCs predominantly exhibited a flattened configuration, superficial ulcer for-
mation, protrusion into the subcutaneous tissue, ill-demarcated deep tumor margin,
and peritumoral fat stranding.
Conventional MRI sequences are also superior to CT for a variety of findings that
influence the therapeutic choice such as laryngeal cartilage invasion, invasion of the
skull base, perineural spread, detection of retropharyngeal lymph nodes in nasopha-
ryngeal carcinoma, extranodal spread in metastatic neck nodes and vascular and
lymphatic invasion.
On T2-weighted MRI the show ill-demarcated, flattened, cutaneous lesion, with
superficial ulcer formation and protrusion and infiltration into subcutaneous fat tis-
sue. Fat-suppressed T2-weighted image is useful for the evaluation of peritumoral
fat stranding. MRI criteria based on the analysis of signal intensity and enhance-
ment patterns after injection of gadolinium have had a major impact on the assess-
ment of deep tumor spread. In most HNSCCs, the actual invasion of bony and
cartilaginous structures is often preceded by tumor-induced inflammation. In laryn-
geal and hypopharyngeal HNSCCs, careful analysis of signal intensities on T1 and
T2 sequences has improved differentiation between tumor and inflammation: mod-
erate enhancement on T1 and moderately high signal on T2 indicate tumor involve-
ment, whereas high signal on T2 and vivid enhancement correspond histologically
to peritumoral inflammation (Fig. 4.7).
The most prevalent primary tumor sites in the head and neck region in
elderly patients seem to be—depending on the series—the oral cavity or the
larynx, each comprising up to one half of all primaries, with the tendency to
overcome their incidence among younger-aged patients. A trend of fewer hypo-
pharyngeal cancer cases in the elderly patient group was also observed.
Considering the tumor stage at presentation, it appears that the occurrence of
an advanced disease (T3, T4) at the primary site is comparable to or even
reduced when matched with that observed in younger age groups, but the
regional lymphatics are primarily less frequently infiltrated by cancer cells in
older patients. Apparently, an increase in the disease severity that would be
expected from the usual delay in diagnosis in older people, probably reflecting
age-related inequalities in access to health care due to a variety of social and
behavioral factors is successfully compensated by a less aggressive biology of
the disease in the elderly [107–109].
Incidence of benign and malignant salivary gland tumors in major portion of the
world ranges from 1 to 2 cases per 100,000 people per year There is no specific
predilection of occurrence of these tumors in any particular gender, although
Warthin’s tumor is more common in males and acinic cell tumor in females. Site-
wise incidence varies for both benign and malignant tumors. Seventy-five to eighty
percent of benign tumors occur in the parotid glands, 5–10% in submandibular
glands, and only 1–2% in sublingual glands. Malignant tumors are more common in
sublingual glands (80%) and least in parotid glands (17–20%). Benign tumors affect
a mean age group of 40 years, and malignant tumors affect an age group of 55 years
[110–114].
Most frequently found in the superficial lobe of the parotid gland, it presents as a
firm, slow-growing asymptomatic mass which is smooth, rounded, lobular, and
mobile with a rubbery consistency causing ear lobule to be raised [115, 116].
On light microscopy, morphologically complex and diverse cellular elements are
seen. Both epithelial and myoepithelial elements are present myxoid to extreme cel-
lular [117].
Surgical excision is the treatment of choice. Historically, enucleation was prac-
ticed which resulted in inadequate surgery and recurrences. Superficial parotidec-
tomy is the most widely accepted technique in the treatment of pleomorphic
adenomas in the superficial lobe of the parotid gland, and total gland excision with
facial nerve preservation is carried out [118, 119].
AL GRAWANY
4 Head and Neck in Geriatric Patients 93
Complete excision like all other tumors is the treatment of choice. Elective
regional lymph node dissection is not indicated, because distant metastasis is more
common than cervical (regional) node involvement [127–131].
The diagnosis of primary squamous cell carcinoma is limited to the major glands.
It occurs between 7 and 95 years of age, the mean age being 60.5 years with a
male predilection of 2:1. Parotid gland is the most commonly involved followed by
submandibular and sublingual glands [132].
Surgical management is the mainstay of treatment. Parotidectomy with or with-
out facial nerve preservation depending on the case is needed for parotid tumors.
Submandibular sialoadenectomy is needed for submandibular gland tumors. A neck
dissection is done in clinically positive necks at the slightest suspicion [116].
Pleomorphic adenoma is the most common salivary gland tumor and is character-
ized by cytomorphological and architectural diversity. On CT and MR images, PAs
are shown as well-circumscribed rounded masses, most commonly located within
the parotid gland, sometimes joined by characteristic lobulated contour enhance-
ment. On T2-weighted images, typical PAs show marked hyperintensity, which
reflects the abundant myxochondroid stroma, with a hypointense rim indicating the
fibrous capsule. The intensity signal within the tumors varies due to the cellular
density, proportion of epithelial and stromal components, and type of stromal com-
ponents. In addition, a variety of secondary histological changes, including fibrosis,
lipometaplasia, ossification, cystic degeneration, and infarction, occur rarely in PAs.
T1-weighted images after contrast administration usually demonstrate homoge-
neous enhancement [133, 134].
It is the second most common benign tumor arising in the parotid gland after benign
mixed tumor. On CT, Warthin’s tumors usually appear as small (2–4 cm, rare
>10 cm), ovoid, smoothly marginated masses. They are homogenous soft tissue
density lesions without calcifications. Cyst formation with homogenous material is
common (30%). The cyst wall is usually thin and fairly smooth. The presence of a
mural nodule helps to distinguish Warthin’s tumors with large cystic components,
septa or multiple adjacent cystic lesions from first branchial cleft cysts or lympho-
epithelial cysts [135–137].
4 Head and Neck in Geriatric Patients 95
On MRI solid and cystic components show low T1-weighted signal, but
cystic areas may show high signal secondary to proteinaceous debris and/or
hemorrhage. In T1-weighted images after contrast administration solid compo-
nents show minimal contrast enhancement. In T2-weighted images, solid com-
ponents appear intermediate to high signal, with high signal in cystic foci,
intermediate signal in Proton Density-weighted images, while in STIR images
the lesions become more conspicuous, especially the cystic components.
Warthin’s tumors show significant restriction of diffusion. The differential
diagnosis of Warthin’s tumor includes benign mixed tumor, benign adenopathy,
lymphoma, benign lymphoepithelial lesions—HIV, adenoid cystic or mucoepi-
dermoid carcinoma, as well as squamous cell carcinoma and melanoma nodal
metastasis [138–140].
MEC has been classified histologically as low, intermediate, or high grade accord-
ing to intracystic components, mitotic figures, neural invasion, necrosis, and cellular
anaplasia.
MRI findings are variable reflecting their histological nature, which seems to
have certain tendencies depending on the tumor grade.
Tumors show inhomogeneous low to intermediate signal intensity on
T2-weighted images, reflecting high cellularity, with an ill-defined margin, reflect-
ing peritumoral inflammatory changes rather than invasive tumor growth. In the
intermediate-grade MECs, tumors showed intermediate signal intensity on
T2-weighted images. Among the low-grade MECs, most tumors had a hyperin-
tense area on T2-weighted images because of the existence of abundant mucin-
secreting cells. High-grade tumors, on the other hand, have lower signal on T2 and
poorly defined margins and infrequent cystic areas. On T1 images following
administration of contrast, there is heterogeneous enhancement of solid compo-
nents [141–144].
Lymph node metastasis was seen often in high-grade MECs.
On CT images, low-grade tumors appear as well-circumscribed lesions, usually
with cystic components. The solid components enhance and calcification is some-
times seen. They have appearances similar to benign mixed tumors. High-grade
tumors have poorly defined margins, infiltrate locally, and appear solid.
Adenoid cystic carcinoma has a propensity for perineural spread. A high-grade vari-
ant is evidenced by a copious of pleomorphic cells, loss of the classic biphasic
epithelial-myoepithelial growth pattern, and comedonecrosis. CT and MRI are
96 T. Popolizio et al.
considered reliable and convenient methods for diagnostic and prognostic predic-
tion, they can both be helpful for demonstrating the extent of invasion in oral cavity-
associated adenoid cystic carcinoma, which can reach the inferior alveolar nerve for
perineural spread by direct invasion through the mandible. It is usually best depicted
on MRI. Low-grade tumors tend to be well-defined, in contradistinction to high-
grade tumors, which appear infiltrative. However, on T1-weighted images both the
subtypes are usually hypo to isointense, on T2 slightly hyperintense, with higher
grades being markedly hypointense and homogeneously enhancing after contrast
administration. In particular, involvement of cranial nerves and tumoral infiltration
around the nerves and osseous structures is optimally assessed via non-contrast
T1-weighted and contrast-enhanced, fat-suppressed T1-weighted MR sequences.
Perineural spread typically appears as enlargement and abnormal enhancement of
the affected nerve and widening or obliteration of the nerve canal. Considering ade-
noid cystic carcinoma of the oral cavity can attain, overrun, and infiltrate the inferior
alveolar nerve by first eroding through the mandibular cortex, and infiltrating
through the bone marrow, CT can be complementary to MRI.
MRI features showed large tumor size, irregular shape, ill-defined margin, extrapa-
rotid infiltration, low-intermediate signal intensity in the solid portions on
T2-weighted images and the presence of central necrosis. On contrast-enhanced
T1-weighted image with the fat-suppression technique, the mass has a central unen-
hanced area and can infiltrate the subcutaneous fat, mandibular ramus, and parapha-
ryngeal space.
The ill-defined margins and extraparotid infiltration, which reflect the invasive
growth of the tumor cells. The appearances of SCC originating in the parotid gland
on MRI can be similar to other more common parotid malignancies (e.g., adenoid
cystic carcinoma and muco-epidermoid carcinoma) (Figs. 4.8 and 4.9) [125, 145].
Fig. 4.8 Parotid gland adenocarcinoma. CECT (a) shows an expansive lesion with avid enhance-
ment located in the superficial lobe and in the deep portion of the gland. Peripheral stranding and
central fluid areas are evident. MRI confirms the presence of an infiltrating lesion with fuzzy bor-
ders that shows isointense signal on T1 (b) and low signal on T2-w (c) and high contrast enhance-
ment on post-contrast T1-w (d). Irregular stranding can be appreciated in the adipose tissue around
and posteriorly to the deep portion on the gland (sagittal post-contrast T1-w (e and f)). Bulky and
coalescent nodal metastases are also present
4 Head and Neck in Geriatric Patients 97
a b
c d
e f
98 T. Popolizio et al.
a b
c d
Fig. 4.9 Parotid gland cystadenoma. The MRI shows expansive lesion located in the deep part of
the parotid gland that deforms its contour and comes close to the external carotid and the internal
jugular vein, without imaging findings of infiltration. The lesion has high signal intensity on T2-w
(a) with an isointense small central component with corresponding high signal intensity on fat
saturated T1-w without contrast (b), suggesting proteinaceous components. On post-contrast T1-w
(c), contrast enhancement is avid in the deeper part of the lesion, while it is more nuanced and
inhomogeneous in the superficial portions. A fluid component with peripheral enhancement is also
noticed. On DWI (d) the lesion shows restriction in the diffusion of proton density. There are also
two lymph nodes in the superficial layers of the gland
4.6 Osteonecrosis
The bisphosphonates play a major role in the treatment and prevention of these
skeletal related events, together with radiation therapy, surgery, analgesics, and
standard anticancer therapy. The primary goal of these therapeutic strategies is to
4 Head and Neck in Geriatric Patients 99
improve the quality of life, as the disease is usually incurable at this stage. The
occurrence of osteonecrosis of the jaw (ONJ) associated with the use of bisphospho-
nates is a potentially new side effect that can have a severe impact on the daily
functioning of the affected individuals, causing great concern among patients, den-
tists, and the medical community [146–148].
Osteonecrosis of the jaw has historically been linked with exposure to white
phosphorous (“phossy jaw”) and in more recent times with radiotherapy and
chemotherapy.
Changes in the socioeconomic fabric of society, resulting in safer working envi-
ronments and the banning of white phosphorus, have caused phossy jaw to be noth-
ing more than a historical curiosity. Osteoradionecrosis, on the other hand, is a
well-defined entity that can be adequately managed with combined hyperbaric oxy-
gen therapy and surgery, although it can develop many years after initial treatment
[6]. Chemotherapy as a cause of ONJ has been infrequently reported in the literature
and is poorly understood, but the presence of infection and dentures seems to be
important in the development of this disorder.
Diagnostic criteria put forward by an expert panel have been published and con-
firm ONJ as a clinical diagnosis. The disorder is defined as the persistence of
exposed bone in the oral cavity after adequate treatment for 6 weeks, in the absence
of local metastatic disease and without previous radiation therapy to the affected
area. This definition is, however, deceptively simple, as the differentiation between
osteonecrosis complicated by infection and osteomyelitis with secondary osteone-
crosis can be difficult, if not impossible. Although these criteria will help uniform
reporting, there is some ambiguity regarding the role of other diagnostic proce-
dures, such as pathology, imaging, and microbiology. Moreover, it is unclear what
an “adequate treatment” should entail [119, 149].
Clinical examination reveals an exposed alveolar ridge with sequestra of necrotic
bone, often with a foul smelling discharge. The surrounding gingival and mucosal
tissues are usually inflamed and painful to touch. The lesions can become multiple,
as one study identified 2.3 areas of ONJ per patient. The mandible is affected in the
majority of cases (60–80%), with the lingual posterior area being particularly sus-
ceptible, which may relate to the thinness of the mucosa in this region and can be
easily traumatized during normal mastication. Fistulization to the maxillary sinus
and the skin can occur and pathological fractures of the mandible have also been
reported [150].
The aspect of ONJ at radiography, CT, and MR imaging is variable and is non-
specific at both anatomic imaging and functional imaging. Imaging takes part in
determining the extent of the disease, diagnosing early stages of osteonecrosis,
identifying a potential association between metastasis to the jaw and ONJ lesions,
excluding other diseases of the jaws, diagnosing complications such as fractures,
and evaluating the jaw before performance of orofacial procedures [151].
The appearance of ONJ at radiography and CT is variable and includes ill-
defined areas of lucency or low attenuation, permeative appearance, cortical destruc-
tion, bony sequestrum, periosteal reaction, or sclerotic changes [152].
100 T. Popolizio et al.
b c
Fig. 4.10 Osteonecrosis. Patient with history of retromolar trigone carcinoma that underwent
surgery and radiation therapy. Orthopantomography (a) documents fixation device in the left man-
dibular bone and the presence of osteolysis discontinuity of bone fragments. Tumefaction of
adjoining small parts is also evident. NCCT with bone algorithm (b) better demonstrates bone
rarefaction and microfractures. On PET 18F-FDG (c) there is high metabolic activity (SUV max
7.09) suggesting inflammation
4 Head and Neck in Geriatric Patients 101
References
1. Lin Y-H, Lin M-Y. Bezold abscess. Ear Nose Throat J. 2015;94(6):E45–6.
2. Abada RL, Mansouri I, Maamri M, Kadiri F. Complications of chronic otitis media. Ann
Otolaryngol Chir Cervicofac. 2009;126(1):1–5. https://doi.org/10.1016/j.aorl.2008.10.006.
3. Pont E, Mazón M. Indications and radiological findings of acute otitis media and its compli-
cations. Acta Otorrinolaringol Esp. 2017;68(1):29–37.
4. Govea-Camacho LH, Pérez-Ramírez R, Cornejo-Suárez A, Fierro-Rizo R, Jiménez-Sala CJ,
Rosales-Orozco CS. Diagnosis and treatment of the complications of otitis media in adults.
Case series and literature review. Cir Cir. 2016;84(5):398–404.
5. Chengazi HV, Desai A, Bhatt AA. Emergency radiologic approach to mastoid air cell fluid.
Emerg Radiol. 2021;28(3):633–40.
6. Kovoor JM, Kademian J, Moritani T, Neal MH, Birkeland AC, Spector ME. Head and neck.
p. 715–75.
7. Mansour S, Magnan J, Nicolas K, Haidar H. Complications of chronic otitis media.
p. 383–414.
8. Nelson D, Jeanmonod R. Bezold abscess: a rare complication of mastoiditis. Am J Emerg
Med. 2013;31(11):1626.e3–4.
9. Bhutta MF, Monono ME, Johnson WD. Management of infective complications of oti-
tis media in resource-constrained settings. Curr Opin Otolaryngol Head Neck Surg.
2020;28(3):174–81.
10. Lemmerling M. Acute otomastoiditis and its complications. p. 53–9.
11. Lyoubi H, Berrada O, Lekhbal A, Abada RA, Mahtar M. Bezold’s abscess: an extremely rare
complication of suppurative mastoiditis: case report and literature review. Int J Surg Case
Rep. 2020;77:534–7.
12. Minks DP, Porte M, Jenkins N. Acute mastoiditis—the role of radiology. Clin Radiol.
2013;68(4):397–405.
13. Lesser FD, Derbyshire SG, Lewis-Jones H. Can computed tomography and magnetic reso-
nance imaging differentiate between malignant pathology and osteomyelitis in the central skull
base? J Laryngol Otol. 2015;129(9):852–9. https://doi.org/10.1017/S0022215115001991.
14. Meltzer PEKG. Pyocutaneous osteomyelitis of the temporal bone, mandible, and zygoma.
Laryngoscope. 1959;169:1300–16.
15. McKellop JA, Bou-Assaly W, Mukherji SK. Emergency head & neck imaging: infections and
inflammatory processes. Neuroimaging Clin N Am. 2010;20(4):651–61.
16. El-Kashlan HK, Harker LA, Shelton C, Aygun N, Niparko JK. Complications of temporal
bone infections. In: Flint PW, Haughey BH, Lund VJ, Niparko JK, Richardson MA, Robbins
KT, editors. Cummings otolaryngology head and neck surgery. 5th ed. Philadelphia, PA:
Mosby; 2010. p. 1979–98.
17. Chole RA, Donald PJ. Petrous apicitis: clinical considerations. Ann Otol Rhinol Laryngol.
1983;92(6 Pt 1):544–51.
18. Finkelstein Y, Marcus N, Mosseri R, Bar-Sever Z, Garty BZ. Streptococcus acidomini-
mus infection in a child causing Gradenigo syndrome. Int J Pediatr Otorhinolaryngol.
2003;67(7):815–7.
19. Chole RA, Sudhoff HH. Chronic otitis media, mastoiditis, and petrositis. In: Flint PW,
Haughey BH, Lund VJ, Niparko JK, Richardson MA, Robbins KT, editors. Cummings oto-
laryngology head and neck surgery. 5th ed. Philadelphia, PA: Mosby; 2010. p. 1963–78.
20. Watkyn-Thomas FW. The treatment of petrositis: (section of otology). Proc R Soc Med.
1936;29(3):267–74.
21. Wanna GB, Dharamsi LM, Moss JR, Bennett ML, Thompson RC, Haynes DS. Contemporary
management of intracranial complications of otitis media. Otol Neurotol. 2010;31(1):111–7.
22. Penido Nde O, Borin A, Iha LC, Suguri VM, Onishi E, Fukuda Y, Cruz OL. Intracranial
complications of otitis media: 15 years of experience in 33 patients. Otolaryngol Head Neck
Surg. 2005;132(1):37–42.
102 T. Popolizio et al.
23. Booth TN, Roland P, Kutz JW, Lee K, Isaacson B. High-resolution 3-D T2-weighted imaging
in the diagnosis of labyrinthitis ossificans: emphasis on subtle cochlear involvement. Pediatr
Radiol. 2013;43(12):1584–90.
24. Mattiola LR, Makowiecky M, Guimarães de Salles CE, Cardoso MP, Cahali S. Labyrinthitis
ossificans. Report of one case and literature review. Int Arch Otorhinolaryngol. 2008.
Accessed 20 May 2019.
25. Paparella MM, Sugiura S. The pathology of suppurative labyrinthitis. Ann Otol Rhinol
Laryngol. 1967;76:554–86.
26. Torok N. Tympanogenic labyrinthitis. Otolaryngol Clin N Am. 1972;5:45–57.
27. Cody Larson MD, Scott A, Jorgensen MD, Alexander J, Towbin MD, Richard Towbin
MD. Labyrinthine ossificans. Appl Radiol. Accessed 30 May 2019.
28. Page N, Kearns D. Radiology quiz case 2. Labyrinthitis ossificans secondary to suppurative
labyrinthitis. Arch Otolaryngol Head Neck Surg. 2011;137:303–5.
29. Swartz JD, Mandell DM, Faerber EN, Popky GL, Ardito JM, Steinberg SB, et al. Labyrinthine
ossification: etiologies and CT findings. Radiology. 1985;157(2):395–8.
30. deSouza C, Paparella MM, Schachern P, Yoon TH. Pathology of labyrinthine ossification. J
Laryngol Otol. 1991;105:621–4.
31. Laurens MB, Becker RM, Johnson JK, Wolf JS, Kotloff KL. MRSA with progression from
otitis media and sphenoid sinusitis to clival osteomyelitis, pachymeningitis and abducens
nerve palsy in an immunocompetent 10-year-old patient. Int J Pediatr Otorhinolaryngol.
2008;72(7):945–51.
32. Sternberg I, Ronen S, Arnon N. Recurrent, isolated, post-febrile abducens nerve palsy. J
Pediatr Ophthalmol Strabismus. 1980;17(5):323–4.
33. Lee NH, Ban JH, Park CY, Kim CC. A case of petrositis with abducens palsy. Korean J
Otolaryngol-Head Neck Surg. 2006;49(8):869–72.
34. Kong SK, Lee IW, Goh EK, Park SE. Acute otitis media induced petrous apicitis presenting
as Gradenigo syndrome: successfully treated by ventilation tube insertion. Am J Otolaryngol.
2011;32(5):445–7.
35. Lutter SA, Kerschner JE, Chusid MJ. Gradenigo syndrome: a rare but serious complication
of otitis media. Pediatr Emerg Care. 2005;21(6):384–6; Marianowski R, Rocton S, Ait-Amer
JL, Morisseau-Durand MP, Manach Y. Conservative management of Gradenigo syndrome in
a child. Int J Pediatr Otorhinolaryngol. 2001;57(1):79–83.
36. Burston BJ, Pretorius PM, Ramsden JD. Gradenigo’s syndrome: successful conservative
treatment in adult and paediatric patients. J Laryngol Otol. 2005;119(4):325–9; Shin HS,
Yoo HK, Suh YR. A case of unusual petrositis. Korean J Otolaryngol-Head Neck Surg.
1964;7(2):59–62.
37. Gillanders DA. Gradenigo’s syndrome revisited. J Otolaryngol. 1983;12(3):169–74.
38. Peters GB III, Bakri SJ, Krohel GB. Cause and prognosis of nontraumatic sixth nerve palsies
in young adults. Ophthalmology. 2002;109(10):1925–8.
39. Lee KC, Chio C, Park KY, Park MS. A case of Gradenigo’s syndrome combining epidural
abscess and Bezold’s abscess. Korean J Otolaryngol-Head Neck Surg. 1989;32(3):567–71.
40. Minotti AM, Kountakis SE. Management of abducens palsy in patients with petrositis. Ann
Otol Rhinol Laryngol. 1999;108(9):897–902.
41. Colpaert C, Van Rompaey V, Vanderveken O, Venstermans C, Boudewyns A, Menovsky T, de
Veuster I, Van de Heyning P, Hamans E. Intracranial complications of acute otitis media and
Gradenigo’s syndrome. B-ENT. 2013;9(2):151–6. PMID: 23909122.
42. Derić D, Arsović N, Dordević V. [Pathogenesis and methods of treatment of otogenic brain
abscess]. Med Pregl. 1998;51(1–2):51–5.
43. Mattos JL, Colman KL, Casselbrant ML, Chi DH. Intratemporal and intracranial com-
plications of acute otitis media in a pediatric population. Int J Pediatr Otorhinolaryngol.
2014;78(12):2161–4.
44. Swartz JD, Hagiwara M. Inflammatory diseases of the temporal bone. p. 1183–229.
45. Goldstein NA, Casselbrant ML, Bluestone CD, Kurs-Lasky M. Intratemporal complications of
acute otitis media in infants and children. Otolaryngol Head Neck Surg. 1998;119(5):444–54.
46. Graß SK, Welkoborsky HJ, Bersch C. Lateral sinus thrombosis—a rare complication of an
acute mastoiditis or infected cholesteatoma. Laryngorhinootologie. 2016;95(1):37–42.
AL GRAWANY
4 Head and Neck in Geriatric Patients 103
47. Park SN, Yeo SW, Rhyoo JY, Lee HY. A case of cavernous sinus thrombophlebitis and
abducence nerve palsy secondary to petrositis. Korean J Otolaryngol-Head Neck Surg.
2002;45(1):82–5.
48. Hendershot EL, Wood JW, Bennhoff D. The middle cranial fossa approach to the petrous
apex. Laryngoscope. 1976;86(5):658–63.
49. Chandler JR. Malignant external otitis. Laryngoscope. 1968;1968(78):1257–94.
50. Mahdyoun P, Pulcini C, Gahide I, Raffaelli C, Savoldelli C, Castillo L, et al. Necrotizing
otitis externa: a systematic review. Otol Neurotol. 2013;34(4):620–9.
51. Sylvester MJ, Sanghvi S, Patel VM, Eloy JA, Ying YM. Malignant otitis externa hospitaliza-
tions: analysis of patient characteristics. Laryngoscope. 2016;127(10):2328–36.
52. Kwon BJ, Han MH, Oh SH, Song JJ, Chang KH. MRI findings and spreading patterns of
necrotizing external otitis: is a poor outcome predictable? Clin Radiol. 2006;61(6):495–504.
Spreading patterns with MRI imagings with clear examples of intracerebral complications.
53. Glikson E, Sagiv D, Wolf M, Shapira Y. Necrotizing otitis externa: diagnosis, treatment, and
outcome in a case series. Diagn Microbiol Infect Dis. 2017;87(1):74–8.
54. Stevens SM, Lambert PR, Baker AB, Meyer TA. Malignant otitis externa: a novel stratifica-
tion protocol for predicting treatment outcomes. Otol Neurotol. 2015;36(9):1492–8.
55. Soudry E, Hamzany Y, Preis M, Joshua B, Hadar T, Nageris BI. Malignant external otitis:
analysis of severe cases. Otolaryngol Head Neck Surg. 2011;144(5):758–62.
56. Chawdhary G, Pankhania M, Douglas S, Bottrill I. Current management of necrotis-
ing otitis externa in the UK: survey of 221 UK otolaryngologists. Acta Otolaryngol.
2017;137(8):818–22.
57. Marsot-Dupuch K, Meyer B. Cochlear implant assessment: imaging issues. Eur J Radiol.
2001;40:119–32.
58. Berrettini S, Baggiani A, Bruschini L, et al. Systematic review of the literature on the clinical
effectiveness of the cochlear implant procedure in adult patients. Acta Otorhinolaryngol Ital.
2011;31:299–310.
59. Carlson ML, Sladen DP, Gurgel RK, Tombers NM, Lohse CM, Driscoll CL. Survey of the
American Neurotology Society on cochlear implantation: part 1, candidacy assessment and
expanding indications. Otol Neurotol. 2018;39:e12–9.
60. Vesseur A, Free R, Snels C, et al. Hearing restoration in cochlear nerve deficiency: the choice
between cochlear implant or auditory brainstem implant, a meta-analysis. Otol Neurotol.
2018;39:428–37.
61. Peng KA, Kuan EC, Hagan S, Wilkinson EP, Miller ME. Cochlear nerve aplasia and hypopla-
sia: predictors of cochlear implant success. Otolaryngol Head Neck Surg. 2017;157:392–400.
62. Birman CS, Brew JA, Gibson WPR, Elliott EJ. CHARGE syndrome and cochlear implanta-
tion: difficulties and outcomes in the paediatric population. Int J Pediatr Otorhinolaryngol.
2015;79:487–92.
63. Frau GN, Luxford WM, Lo WW, Berliner KI, Telischi FF. High-resolution computed tomog-
raphy in evaluation of cochlear patency in implant candidates: a comparison with surgical
findings. J Laryngol Otol. 1994;108:743–8.
64. Aldhafeeri AM, Alsanosi AA. Management of surgical difficulties during cochlear implant
with inner ear anomalies. Int J Pediatr Otorhinolaryngol. 2017;92:45–9.
65. Deep N, Dowling E, Jethanamest D, Carlson M. Cochlear implantation: an overview. J
Neurol Surg Part B Skull Base. 2018;80:169–77.
66. Sennaroglu L. Cochlear implantation in inner ear malformations—a review article. Cochlear
Implants Int. 2010;11:4–41.
67. Sennaroğlu L, Bajin MD. Classification and current management of inner ear malformations.
Balkan Med J. 2017;34:397–411.
68. Birman CS, Elliott EJ, Gibson WPR. Pediatric cochlear implants: additional disabilities
prevalence, risk factors, and effect on language outcomes. Otol Neurotol. 2012;33:1347–52.
69. Hellingman CA, Dunnebier EA. Cochlear implantation in patients with acute or chronic
middle ear infectious disease: a review of the literature. Eur Arch Otorhinolaryngol.
2009;266:171–6.
70. Amin N, Sethukumar P, Pai I, Rajput K, Nash R. Systematic review of cochlear implantation
in CHARGE syndrome. Cochlear Implants Int. 2019;20:266–80.
104 T. Popolizio et al.
71. Fasunla AJ, Nwaorgu OGB. Adult chronic rhinosinusitis: spectrum of clinical features in a
tertiary health institution and literature review. East Cent Afr J Surg. 2011;16(1):12–8.
72. Lanza DC, Kennedy DW. Adult rhinosinusitis defined. Otolaryngol Head Neck Surg.
1997;117(3 pt 2):S1–7.
73. Benninger MS, Ferguson BJ, Hadley JA. Adult chronic rhinosinusitis: definitions, diagnosis,
epidemiology, and pathophysiology. Otolaryngol Head Neck Surg. 2003;129(3 Suppl):S1–32.
74. Bhattacharyya N. A comparison of symptom scores and radiographic staging systems in
chronic rhinosinusitis. Am J Rhinol. 2005;19(2):175–9.
75. Fokkens W, Lund V, Bachert C, Clement P, Hellings P, Holmstrom M, Jones N, Kalogjera
L, Kennedy D, Kowalski M, Malmberg H, Mullol J, Passali D, Stammberger H, Stierna
P. EAACI position paper on rhinosinusitis and nasal polyps executive Summary. Allergy.
2005;60(5):583–601.
76. Lund VJ, Kennedy DW. Staging for rhinosinusitis. Otolaryngol Head Neck Surg.
1997;117(suppl):S35–40.
77. Hopkins C, Browne JP, Slack R, Lund V, Brown P. The Lund-Mackay grading system for
chronic rhinosinusitis: how is it used and what does it predict. Otolaryngol Head Neck Surg.
2007;137(4):555–61.
78. Bhattacharyya N. The role of CT and MRI in the diagnosis of chronic rhinosinusitis. Curr
Allergy Asthma Rep. 2010;10(3):171–4.
79. Arango P, Kountakis SE. Significance of computed tomography pathology in chronic rhino-
sinusitis. Laryngoscope. 2001;111(10):1779–82.
80. Kenny TJ, Duncavage J, Bracikowski J, Yildirim A, Murray JJ, Tanner SB. Prospective
analysis of sinus symptoms and correlation with paranasal computed tomography scan.
Otolaryngol Head Neck Surg. 2001;125(1):40–3.
81. Bhattacharyya N, Fried MP. The accuracy of computed tomography in the diagnosis of
chronic rhinosinusitis. Laryngoscope. 2003;113(1):125–9.
82. Karkos PD, Khoo LC, Leong SC, Lewis-Jones H, Swift AC. Computed tomography and/or
magnetic resonance imaging for pre-operative planning for inverted nasal papilloma: review
of evidence. J Laryngol Otol. 2009;123(7):705–9.
83. Loevner L, Sonners A. Imaging of neoplasms of the paranasal sinuses. Neuroimaging Clin
N Am. 2004;4:625–46.
84. Wassef SH, Batra PS, Barnett S. Review article: Skull base inverted papilloma: a comprehen-
sive review. ISRN. 2012:1–34.
85. Jeon TY, Kim HJ, Chung SK, Dhong HJ, Kim HY, Yim HJ, Kim ST, Jeon P. Sinonasal
inverted papilloma: value of convoluted cerebriform pattern on MR imaging. AJ Neuroradiol.
2007;29:1556–60.
86. Wright EJ, Chernichenko N, Ocal E, Moliterno J, Bulsara KR, Judson BL. Benign inverted
papilloma with intracranial extension: prognostic factors and outcomes. Skull Base Rep.
2011;1:145–50.
87. Chawla A, Shenoy J, Chokkappan K, Chung R. Imaging features of sinonasal inverted papil-
loma: a pictorial review. Curr Probl Diagn Radiol. 2015;10:1–7.
88. Maroldi R, Farina D, Palvarini L. Magnetic resonance imaging findings of inverted papil-
loma: differential diagnosis with malignant sinonasal tumors. Am J Rhinol. 2004;18:305–10.
89. Anari S, Carrie S. Sinonasal inverted papilloma: narrative review. J Laryngol Otol.
2010;124:705–15.
90. Head CH, Sercaz JA, Luu Q, Collis J, Blackwell K. Radiographic assessment of inverted
papilloma. Acta Otolaryngol. 2007;127:515–20.
91. Alba JR, Armengot M, Díaz A, Pérez A, Rausell N, Basterra J. Inverted papilloma of the
sphenoid sinus. Acta Otorhinolaryngol Belg. 2002;56(4):399–4.
92. Lisan Q, Laccourreye O, Bonfils P. Sinonasal inverted papilloma: from diagnosis to treat-
ment. Eur Ann Otorhinolaryngol Head Neck Dis. 2016;133(5):337–41.
93. Ozturk K, Gawande R, Gencturk M, Boegel K, Caicedo-Granados E, Cayci Z. Imaging fea-
tures of sinonasal tumors on positron emission tomography and magnetic resonance imaging
including diffusion weighted imaging: a pictorial review. Clin Imaging. 2018;51:217–28.
4 Head and Neck in Geriatric Patients 105
94. Salomone R, Matsuyama C, Filho G, De Alvarenga ML, Neto EM, Chaves AG. Bilateral
inverted papilloma. Case report and review of literature. Rev Bras Otorrinolaringol.
2008;74(2):293–6.
95. Strek P, Zagolski O, Skladzien J, Oles K, Konior M, Hydzik-Sobocinska K. Endoscopic
surgical treatment of patients with isolated sphenoid sinus disease. Otolaryngol Pol.
2007;61(3):254–9.
96. Lee JT, Bhuta S, Lufkin R, Castro DJ. Isolated inverting papilloma of the sphenoid sinus.
Laryngoscope. 2003;113(1):41–4.
97. Lee DK, Chung SK, Dhong H-J, Kim HY, Kim H-J, Bok KH. Focal hyperostosis on CT
of sinonasal inverted papilloma as a predictor of tumor origin. AJNR Am J Neuroradiol.
2007;28:618–21.
98. Bhalla RK, Wright ED. Predicting the site of attachment of sinonasal inverted papilloma.
Rhinology. 2009;47:345–8.
99. Liang N, Huang Z, Liu H, Xian J, Huang Q, Zhou B. Bone involvement: histopathologi-
cal evidence for endoscopic management of sinonasal inverted papilloma. Laryngoscope.
2017;127:2703–8.
100. Wilkins RB, Pressly JP. Diagnosis and incidence of lacrimal calculi. Ophthalmic Surg.
1980;11:787–9.
101. Weber AL, Rodriguez-DeVelasquez A, Lucarelli MJ, Cheng HM. Normal anatomy and
lesions of the lacrimal sac and duct: evaluated by dacryocystography, computed tomography,
and MR Imaging. Neuroimaging Clin N Am. 1996;6:199–216.
102. Kassel EE, Schatz CJ. Anatomy, imaging, and pathology of the lacrimal apparatus. In: Som
PM, Curtin HD, editors. Head and neck imaging. 5th ed. St. Louis, MO: Elsevier-Mosby;
2011. p. 757–853.
103. Hurwitz JJ, Edward Kassel EE, Jaffer N. Computed tomography and combined
CT-dacryocystography (CT-DCG). In: Hurwitz JJ, editor. The lacrimal system. New York:
Lippincott-Raven; 1996. p. 83–5.
104. Choi SC, Lee S, Choi HS, Jang JW, Kim SJ, Lee JH. Preoperative computed tomography
findings for patients with nasolacrimal duct obstruction or stenosis. Korean J Ophthalmol.
2016;30:243–50.
105. Tawfik KO, Ishman SL, Altaye M, Meinzen-Derr J, Choo DI. Pediatric acute otitis media in
the era of pneumococcal vaccination. Otolaryngol Head Neck Surg. 2017;156(5):938–45.
106. Taxy JB. Squamous carcinoma in a major salivary gland: a review of the diagnostic consider-
ations. Arch Pathol Lab Med. 2001;125:740–5.
107. Christe A, Waldherr C, Hallett R, et al. MR imaging of parotid tumors: typical lesion char-
acteristics in MR imaging improve discrimination between benign and malignant disease.
AJNR Am J Neuroradiol. 2011;32:1202–7.
108. Batsakis JG, McClatchey KD, Johns M, et al. Primary squamous cell carcinoma of the parotid
gland. Arch Otolaryngol. 1976;102:355–7.
109. Ying YL, Johnson JT, Myers EN. Squamous cell carcinoma of the parotid gland. Head Neck.
2006;28:626–32.
110. Heaton CM, Chazen JL, van Zante A, Glastonbury CM, Kezirian EJ, Eisele DW. Pleomorphic
adenoma of the major salivary glands: diagnostic utility of FNAB and MRI. Laryngoscope.
2013;123(12):3056–60.
111. Yarington CT Jr, Yonkers AJ, Carter WS. Salivary gland tumors. Nebr State Med
J. 1971;56(4):146–51.
112. Spiro RH. Salivary neoplasms: overview of a 35-year experience with 2,807 patients. Head
Neck Surg. 1986;8:177–84.
113. Abdel Razek AAK, Mukherji SK. State-of-the-art imaging of salivary gland tumors.
Neuroimaging Clin N Am. 2018;28:303–17.
114. Thoeny HC. Imaging of salivary gland tumors. Cancer Imaging. 2007;7:52–62.
115. Phillips PP, Olsen KD. Recurrent pleomorphic adenoma of the parotid gland: report of 126
cases and a review of the literature. Ann Otol Rhinol Laryngol. 1995;104:100–4.
106 T. Popolizio et al.
116. Tsushima Y, Matsumoto M, Endo K, et al. Characteristic bright signal of parotid pleomor-
phic adenomas on T2-weighted MR images with pathological correlation. Clin Radiol.
1994;49:485–89; Ikeda K, Katoh T, Ha-Kawa SK, et al. The usefulness of MR in establishing
the diagnosis of parotid pleomorphic adenoma. AJNR Am J Neuroradiol. 1996;17:555–59.
117. Motoori K, Yamamoto S, Ueda T, et al. Inter- and intratumoral variability in magnetic reso-
nance imaging of pleomorphic adenoma: an attempt to interpret the variable magnetic reso-
nance findings. J Comput Assist Tomogr. 2004;28:233–46.
118. Mandelblatt SM, Braun IF, Davis PC, et al. Parotid masses: MR imaging. Radiology.
1987;163:411–4.
119. Epstein JB, Wong FL, Stevenson-Moore P. Osteoradionecrosis: clinical experience and a pro-
posal for classification. J Oral Maxillofac Surg. 1987;45(2):104–10.
120. Minami M, Tanioa H, Oyama K, et al. Warthin’s tumor of the parotid gland: MR-pathologic
correlation. AJNR Am J Neuroradiol. 1993;14:209–14.
121. Liu XW, Xie CM, Li H, Zhang R, Geng ZJ, Mo YX, et al. Nasopharyngeal adenoid cystic
carcinoma: magnetic resonance imaging features in ten cases. Chin J Cancer. 2012;31:19–28.
122. Shimamoto H, Chindasombatjaroen J, Kakimoto N, Kishino M, Murakami S, Furukawa
S. Perineural spread of adenoid cystic carcinoma in the oral and maxillofacial regions: evalu-
ation with contrast-enhanced CT and MRI. Dentomaxillofac Radiol. 2012;41:143–51.
123. Joe VQ, Westesson PL. Tumors of the parotid gland: MR imaging characteristics of various
histologic types. AJR Am J Roentgenol. 1994;163:433–8.
124. Tian Z, Li L, Wang L, Hu Y, Li J. Salivary gland neoplasms in oral and maxillofacial regions:
a 23-year retrospective study of 6982 cases in an eastern Chinese population. Int J Oral
Maxillofac Surg. 2010;39:235–42.
125. Goode RK, Auclair PL, Ellis GL. Mucoepidermoid carcinoma of the major salivary glands:
clinical and histopathologic analysis of 234 cases with evaluation of grading criteria. Cancer.
1998;82:1217–24.
126. Jansisyanont P, Blanchaert RH Jr, Ord RA. Intraoral minor salivary gland neoplasm: a single
institution experience of 80 cases. Int J Oral Maxillofac Surg. 2002;31:257–61.
127. Khalek Abdel Razek AA. Diffusion-weighted magnetic resonance imaging of head and neck.
J Comput Assist Tomogr. 2010;34:808–15.
128. Meyers M, Granger B, Herman P, Janot F, Garrel R, Fakhry N, et al. Head and neck ade-
noid cystic carcinoma: a prospective multicenter REFCOR study of 95 cases. Eur Ann
Otorhinolaryngol Head Neck Dis. 2016;133:13–7.
129. Coca-Pelaz A, Rodrigo JP, Bradley PJ, Vander Poorten V, Triantafyllou A, Hunt JL, et al.
Adenoid cystic carcinoma of the head and neck—an update. Oral Oncol. 2015;51:652–61.
130. van Weert S, Lissenberg-Witte BI, Bloemena E, Leemans CR. Mucoepidermoid carcinoma
of the head and neck: CRTC1/3 MAML 2 translocation and its prognosticators. Eur Arch
Otorhinolaryngol. 2021;279(5):2573–81. https://doi.org/10.1007/s00405-021-07039-2.
131. Takashima S, Wang J, Takayama F, et al. Parotid masses: prediction of malignancy using
magnetization transfer and MR imaging findings. AJR Am J Roentgenol. 2001;176:1577–84.
132. Casselman JW, Mancuso AA. Major salivary gland masses: comparison of MR imaging and
CT. Radiology. 1987;165:183–9.
133. Celebi I, Mahmutoglu AS, Ucgul A, Ulusay SM, Basak T, Basak M. Quantitative diffusion-
weighted magnetic resonance imaging in the evaluation of parotid gland masses: a study with
histopathological correlation. Clin Imaging. 2013;37:232–8.
134. Yoshino N, Yamada I, Ohbayashi N, et al. Salivary glands and lesions: evaluation of appar-
ent diffusion coefficients with split-echo diffusion-weighted MR imaging–initial results.
Radiology. 2001;221:837–42.
135. Thompson L. World Health Organization classification of tumours: pathology and genetics of
head and neck tumours. Ear Nose Throat J. 2006;85:74.
136. Habermann CR, Gossrau P, Graessner J, et al. Diffusion-weighted echo-planar MRI: a valu-
able tool for differentiating primary parotid gland tumors? Rofo. 2005;177:940–5.
4 Head and Neck in Geriatric Patients 107
5.1 Introduction
In the United States (US), adults aged ≥75 years represent 6% of the entire popula-
tion and the number of US citizens older than 80 years is expected to rise approxi-
mately by 25 million in the next 30 years.
Cardiovascular disease is the most frequent single cause of death in persons over
65 years of age, and more than 60% of myocardial infarctions occur in patients over
75 years of age [1]. The increase in life expectancy will likely cause an increase in
myocardial infarction cases [2]. Cardiovascular diseases such as coronary artery
disease, arrhythmias, heart failure and valve disease increase in incidence with
increasing age [3]. Age itself affects cardiac physiology and remodelling, making
challenge to distinguishing age-related changes to pathology. Main cardiovascular-
related changes included increased diffuse fibrosis causing cardiovascular stiffness
with reduced vascular compliance, ventricular diastolic and systolic dysfunction,
and alteration in conduction system; increased lipid endothelial deposition with pro-
gressive atherosclerosis and fibrocalcific valve degeneration.
Cardiac Computed Tomography (CCT) and Magnetic Resonance Imaging (MRI)
are able to provide a deep characterization of myocardial anatomy, function and
remodelling, crucial for screening, diagnosis, risk stratification and therapy guid-
ance. Furthermore, imaging has a pivotal role in planning of interventional proce-
dures, rapidly developed in the last years aimed to safely treat more fragile patients
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 109
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_5
110 A. Palmisano et al.
[4]. The understanding of cardiac disease typically occurring in elderly and the
potential value of each cardiac imaging modality may help to decide on the most
suitable imaging modality and its timing [5]. This chapter provides an overview of
cardiac imaging modalities for the most frequent cardiovascular diseases in the
elderly population.
5.2.1 Introduction
Coronary artery disease (CAD) constitutes one of the main causes of mortality in
the Western world. Many important studies such as the NHANES (National Health
and Nutrition Examination Survey), the FHS (Framingham Heart Study), MESA
(Multi-Ethnic Study of Atherosclerosis) and the CHS (Cardiovascular Health
Study) stated that the prevalence of CAD drastically increases in the elderly popu-
lation with men affected more than women [6–8]. The development of an athero-
sclerotic plaque in the intima of the coronary arteries is the primary cause of the
disease. The majority of coronary atherosclerotic plaques will not determine any
symptoms; however, some can clinically manifest in various forms that are gener-
ally classified as chronic coronary syndrome (CCS) and acute coronary syn-
drome (ACS).
CCS include the so-called Stable Angina in which thoracic pain is exacerbated
by the presence of an atherosclerotic plaque in the intima layer of the coronary
arteries which induces significant stenosis of the coronary lumen and reduction of
blood flow supply. This results in faster exhaustion of the coronary flow reserve and
the subsequent insufficiency of oxygen supply to the myocardium, which typically
occurs after physical exercise or in any situation with increased oxygen demand.
ACS is commonly triggered by the rupture or the erosion of the fibrous cap of an
atherosclerotic plaque, which leads to rapid platelet aggregation and thrombus for-
mation. This can determine a sudden variable degree of obstruction to the coronary
blood flow with downstream myocardial damage, resulting in three different clini-
cal manifestations: unstable angina, non-ST-elevation myocardial infarction
(NSTEMI), or ST-elevation myocardial infarction (STEMI). In some cases, sudden
arrhythmic death may be the first clinical manifestation of CAD.
lumen. Often, these plaques are not associated with relevant luminal stenosis and
exhibit a thin, fibrous cap with a large necrotic core. Other typical features of vul-
nerability are plaque vascularization, high plaque volume, matrix metalloproteinase
expression and collagenase activity, and macrophage infiltration of the fibrous
cap [11].
Complete vessel occlusion can result in myocardial necrosis and subsequent
STEMI or NSTEMI, whereas partial vessel occlusion can lead to ischemia without
myocardial necrosis that clinically manifest as unstable angina [12].
However, the majority of atherosclerotic plaques remain clinically silent. These
slow-growing plaques induce progressive vessel stenosis with flow-limiting condi-
tion, whose symptoms eliciting stable coronary artery disease became manifest dur-
ing physical exercise and all condition requiring an increased oxygen demand for a
mismatch between the myocardial oxygen demand and myocardial oxygen con-
sumption. The reduction of coronary flow reserve is proportional to the degree of
luminal narrowing; however, it is further worsened by the endothelial dysfunction,
which typically occur in atherosclerosis causing impaired vasodilation indepen-
dently by the degree of stenosis.
Cardiac imaging can be used to unravel the coronary artery disease by either trig-
gering the ischemic process or via the direct visualization of coronary artery steno-
ses [13].
The majority of coronary stenosis do not cause ischemia and consequently do not
require revascularization. Ischemia was found in approximately 50% of patients
with obstructive CAD (stenosis ≥ 50%); therefore, the functional significance of
stenosis should be considered uncertain, and an imaging-based stress examination
is frequently required to identify myocardial ischemia in the setting of CCS [14].
Ischemia can be triggered by physical exercise or pharmacologic stress [15]. Single-
photon emission computed tomography (SPECT), positron emission tomography
(PET) myocardial perfusion, stress echocardiography, stress computed tomography
and stress magnetic resonance (MR) imaging are the most commonly used tests
[16]. Recent studies stated that the Stress MRI and PET have similar accuracy and
are suitable to rule out hemodynamically significant coronary artery diseases in a
wide range of pre-test probability, while stress SPECT and echocardiography are
less reliable [17]. Stress MRI has also the advantage, over PET, of combining a
multiparametric characterization of the myocardium.
The direct visualization of coronary anatomy can be obtained invasively with
coronary angiography (ICA) or non-invasively with coronary computed tomog-
raphy angiography (CCTA). ICA offers the best spatial resolution and can be
combined with the measurement of the fractional flow reserve (FFR) to quantify
the stenosis significance and guide revascularization if its value is lower than
0.8 [16].
112 A. Palmisano et al.
In the recent years, CCTA emerged has a valid alternative to ICA in patients with
low-intermediate likelihood for CAD thanks to its negative predictive value close to
100% [13]. Despite CCTA is a pure anatomic method, the continuous technical and
methodological development opened the possibility of a CT-based assessment of func-
tional significance of stenosis using CT stress perfusion or non-invasive virtual FFR.
Each of the outlined strategies presents certain limitations. Ischemia testing has
limited sensitivity and specificity, cannot identify where coronary plaques are
located, and cannot perform stenosis grading and plaque characterization. Invasive
coronary angiography is associated with potential complications for its invasiveness
and high radiation exposure, as well as higher costs compared to non-invasive tech-
niques. CCTA can suffer from poor image quality in patients with arrhythmias or
tachycardia or can be misinterpreted determining false-positive results in case of a
very high calcium burden [13].
For these reasons, choosing the best imaging strategy must take into account
patient characteristics, pre-test probability, medical expertise and available tech-
nologies, preferring a non-invasive anatomical imaging strategy (CCTA) in case of
no previous history of CAD, low to intermediate clinical likelihood and when infor-
mation about the presence and the burden of coronary atherosclerosis is desired to
tailor the patient management.
While the use of imaging is well established for diagnosing CAD in symptomatic
patients, its role in primary prevention is still unclear. In asymptomatic individuals,
traditional risk factors such as hypertension, diabetes mellitus, sex and family his-
tory are used to estimate the risk of future major cardiac events and the need for
risk-lowering treatments. Non-enhanced cardiac CT has a crucial role in risk strati-
fication via the evaluation of coronary calcium score and will be discussed in the
following sections.
Non-invasive cardiac imaging has limited role in the setting of acute coronary
obstruction [18, 19], but has gained a growing role in patients with low-risk acute
chest pain or with a clinical-angiographic diagnosis of Myocardial Infarction with
Non-Obstructed Coronary Arteries (MINOCA).
CCTA has rapidly gained a central role to rule out CAD in patients with acute chest
pain and a relatively low pretest probability of ACS. However, it cannot exclude other
important causes of acute chest pain with unobstructed coronaries, such as acute myo-
carditis, myocardial infarction with normal coronary arteries, and cardiomyopathies.
A few recent studies showed the possibility to obtain information about myocardial
scar and extracellular volume also in CT; however, these techniques still need experi-
enced readers [20–22] and require large studies to define its generalizability.
Cardiac MRI is widely adopted in patients with non-obstructed coronary arteries
at ICA, being able to provide differential diagnosis of ACS impacting on patients’
treatment and management. Moreover, it plays a role in patients risk stratification
and in the diagnosis of complications [18].
5.2.3.1 CCTA
Minimum technological requirement and patients’ preparation are crucial to obtain
a diagnostic image quality. 64-detector row CT is considered the minimum standard
for CCTA.
5 Heart Diseases in Geriatric Patients 113
Image quality improves substantially when the heart rate is regular and lower
than 65 beats/min. In the absence of contraindication, the administration of beta-
blockers and nitrates to lower the heart rhythm and to vasodilate coronary arteries is
recommended to improve image quality and coronary evaluation. Different acquisi-
tion strategies are available to obtain the best image quality based on patient’s heart
rate, BMI and ability to breath-hold. In general, retrospective ECG-gated helical
acquisition enables the reconstruction of image data sets at arbitrary time points
during the cardiac cycle. This technique provides excellent flexibility for identify-
ing the optimal cardiac phase without motion artefacts at the cost of higher radiation
exposure. Prospective ECG-triggered acquisition substantially reduces radiation
exposure but requires a lower and regular heart rate.
In the elderly population, considering the lower impact of radiation exposure,
retrospective ECG-gated helical acquisition often represents a good choice, but the
application of the tube-current modulation during the cardiac cycle to the retrospec-
tive ECG-gated helical acquisition is anyway recommended, in order to maintain
radiation exposure at reasonable level.
Chronic kidney disease can represent an important limitation to the exam in this
category of patients. However, dual energy acquisition offers the possibility to per-
form CCTA with very low dose of contrast agent.
Calcium Scoring
Calcium hydroxyapatite deposition in coronary arteries always occurs in the intima
associated with coronary atherosclerotic plaque formation; here, inflammation trig-
gers processes similar to osteogenesis. The majority of elderly patients presents
with coronary calcium without any symptoms. Non-contrast cardiac CT is used to
establish the coronary artery calcium score (CACS), also known as the Agatston
score, considering each calcified plaque using dedicated software. Finally, scores
for all detected lesions are summarized to obtain the total score. Non-contrast CT
should be ECG-gated and acquired at 120 kVp; however, recent studies documented
strong correlation with CACS measured from non ECG-gated non contrast CT scan
and also from ECG-gated low dose (80 kVp) scan [23].
In asymptomatic individuals the presence and the extent of coronary calcium
correlate to total atherosclerotic plaque burden and to the risk of future cardiovascu-
lar events [24]. Any coronary calcium, including a single focus of calcium, indicates
increased atherosclerotic cardiovascular disease risk over the next 10–15 years,
while its absence indicates less than 1% risk for atherosclerotic cardiovascular
events in the next 10 years [25, 26].
The use of Coronary Artery Disease Reporting and Data System (CAD-RADS)
[31] to report the results of a CCTA examination should be promoted to improve
standardization and communications among physicians.
a b c
Fig. 5.1 (a–d) CCTA of a 77-year-old man with previous PCI on left anterior descending artery
(a), circumflex (b) and right coronary artery (c). CCTA showed good patency of the coronary
artery stent on left anterior descending artery (a) and circumflex artery (b), without signs of neo-
intimal hyperplasia, differently from stents on the right coronary artery with evidence of intrastent
stenosis >50% in the proximal and medium stent and total occlusion of the distal stent
5 Heart Diseases in Geriatric Patients 115
a b c d
Fig. 5.2 (a–d) CCTA of a 78-year-old man with previous CABG: left internal mammary artery on
left anterior descending artery (arrow in a), and a venous graft for the posterior descending artery
(arrow in c). CCTA documented good patency of both grafts as showed in 3D volume rendering (a
and c) and multiplanar reconstruction (b and d)
contrast media, with good accuracy compared to MRI [20–22]. Different scanning
protocol were tested based on low kV single energy scan or dual energy acquisition
[20–22].
a b
c d
Fig. 5.3 (a–d) CMR images of a 45-year-old man with STEMI. CMR images were performed
6 days after STEMI due to culprit lesion on left circumflex artery, promptly treated with PCI. CMR
showed oedema on the lateral mid-basal wall (a and c) involving 34% of myocardial mass associ-
ated with transmural post-ischemic LGE (b and d) involving 23% of myocardial mass. Endocardial
hypointensity was recognizable in the endocardium of the injured myocardium both on STIR
images (asterisks in a and c) and LGE images (asterisks in b and d) referrable to myocardial haem-
orrhage and microvascular obstruction respectively
[43, 56]. In this setting, imaging is indicated for the assessment of the LV ejection
fraction before hospital discharge [47, 56] especially when echocardiography is
suboptimal or inconclusive [47, 57].
Moreover, CMR represents a game-changer in all the cases of cTnT elevation of
unknown origin, outlining the diagnosis of all the conditions that can mimic AMI,
such as Takotsubo cardiomyopathy, myocarditis, and MINOCA. CMR is an impor-
tant management tool in this setting since it has a crucial role in guiding therapy
[42, 58].
In patients with chornic coronary syndrome with intermediate stenosis [59, 60],
stress cardiac MRI is an effective imaging to identify myocardial ischemia and via-
bility, resulting fundamental for guiding patients’ revascularization. Inducible isch-
emia is detected as an area of myocardial hypointensity in a coronary territory,
extending from the endocardium to the epicardium, recognizable during the first
pass perfusion at the peak of myocardial enhancement, not visible at rest perfusion
and in the absence of scar at LGE.
118 A. Palmisano et al.
5.3.1 Introduction
Heart failure represents the latest stage of every cardiovascular disease. It is defined
as ischemic heart failure if it is a consequence of CAD or as non-ischemic heart
failure if it is due to other diseases (e.g., hypertension, idiopathic cardiomyopathies,
valvular diseases, inflammation, auto-immune diseases, nutritional deficiency,
infections, and drugs). These conditions can trigger two primary pathophysiological
alterations: volume (i.e., valvular regurgitation) or pressure overload (i.e., hyperten-
sion, aortic stenosis) and systolic dysfunction (i.e., CAD, idiopathic cardiomyopa-
thies) [62].
Clinically, it can manifest as left HF, whose symptoms are mainly due to low
cardiac output with fatigue and syncope or pulmonary congestion with dyspnoea
and pulmonary oedema. Instead, jugular vein engorgement, hepatic congestion, and
peripheric oedema are the typical signs of right HF [63].
EF is an essential parameter in patients affected by HF, and European guidelines
use EF to classify HF in three different categories: HF with preserved ejection frac-
tion (HFpEF, >50%), HF with mid-range reduction of EF (HFmrEF, 40–49%), and
HF with reduced ejection fraction (HFrEF, <40%). This classification is of para-
mount importance because it reflects aetiologies and comorbidities of HF, with an
important impact on prognosis and therapy [61].
This classification also reflects the pathophysiological mechanism of HF, which
can be distinguished in systolic and diastolic HF. In systolic dysfunction, there is a
contractile insufficiency of the left ventricle that loses its capability to guarantee an
expected cardiac output, leading to HFrEF (i.e., CAD, DCM) [61]. Dilated
Cardiomyopathy (DCM) is a form of HFrEF and is defined as a left ventricular dila-
tion and systolic dysfunction in the absence of coronary artery disease or abnormal
loading conditions proportionate to the degree of LV impairment. It can be due to
many causes, such as alcohol consumption, genetic aetiology (e.g., lamin A/C muta-
tion or myotonic dystrophy), anthracycline therapy history, HIV infection,
5 Heart Diseases in Geriatric Patients 119
scanner in the recent years, CCTA can accurately evaluate cardiac structure and
function. Moreover, using Late Contrast Enhancement (LCE) scan, CT could also
identify non-ischemic cardiac disease underlying HF and may provide a quantifica-
tion of myocardial stiffness through the measurement of ECV with results compa-
rable to CMR [73, 74].
1. The extent of the scar: if it is <15%, LGE can predict a good response to
CRT [85].
2. The location of the myocardial scar: if it involves the postero-lateral wall, the
myocardial scar is predictive of a lower response.
3. The presence of fibrosis in the site of LV lead placement, which may reduce the
effectiveness of CRT [86].
Furthermore, some authors discovered that pacing in a site with <50% scar trans-
murality was associated with response to CRT [87].
In conclusion, CMR can provide a complete cardiac assessment for CRT, since it
is the gold standard to evaluate the size and the function of the chambers, determine
prognosis, and provide invaluable information about the probability of response to
CRT via LGE.
5.3.4 Amyloidosis
The high accessibility and the ability to describe both cardiac structure and func-
tion make echocardiography the first-line tool in CA assessment. The typical echo-
cardiographic findings in CA comprise a small left ventricle with concentric
hypertrophy, a sparkling myocardial appearance, a biatrial enlargement with an
increased atrial septum thickness, and a restrictive physiology [90].
Recent ultrasound techniques such as speckle tracking echocardiography were
more sensitive than traditional echocardiography in detecting global and regional
cardiac function changes. Several groups have shown the good sensitivity and speci-
ficity of basal to apical longitudinal strain ratio for differentiating CA from other
cardiac pathologies [91, 92].
CMR is the imaging tool of choice to diagnose CA because of its tissue charac-
terization capabilities [93]. LGE is historically the cornerstone of CMR to diag-
nose CA in patients whose kidney function allows contrast medium administration.
The typical LGE pattern in CA is represented by diffuse and inhomogeneous myo-
cardial hyperenhancement; sometimes, subendocardial circumferential hyperen-
hancement can be present in some patients while others can also show transmural
hyperenhancement [94]. Some researchers suppose that these different LGE pat-
terns are representative of different phases due to continuous amyloid deposition,
with progression from no LGE to transmural LGE [95]. Another typical sign of
amyloid deposition is the abnormal myocardial and blood-pool kinetics demon-
strated in the inversion time scout sequence [96]. Native myocardial T1 mapping
enables CA diagnosis without the need to administer gadolinium since very high
T1 values are a typical feature of CA [97]. The integration of native T1 with post-
contrast T1 values allows a non-invasive quantification of ECV [97]. High value of
T1 mapping and ECV represent one of the earliest imaging signs for cardiac amy-
loid deposition (Fig. 5.4); they can be altered even when LGE is absent [98].
Instead, T2 mapping can show oedema in both types of amyloidosis, but this is
generally larger in light chain CA [93]. While the integrated use of clinical find-
ings, electrocardiography, echocardiography, and CMR makes the diagnosis of
cardiac amyloidosis possible, these methodologies cannot accurately contribute to
the characterization of the type of the underlying amyloid deposition. Myocardial
scintigraphy with bone avid tracers has high sensitivity and specificity to diagnose
ATTR CA; the ease of access, imaging simplicity, low cost and high specificity for
ATTR CA are some of the advantages of this diagnostic tool [99–101]. In conclu-
sion, cardiac imaging represents a critical tool for the diagnosis of CA. The step-
wise use of various imaging modalities in conjunction with the clinical and
laboratory data diagnose this pathology in most patients; therefore, cardiac biopsy,
the gold standard diagnostic test to confirm and provide typization of amyloidosis,
is no longer routinely performed in clinical practice due to its invasive nature and
limited availability.
5 Heart Diseases in Geriatric Patients 123
a b c
d e f
Fig. 5.4 CMR images of a 73-years-old man with cardiac amyloid. Cine SSFP image (a) shows a
concentric myocardial hypertrophy (end-diastolic wall thickness of mid-basal septum: 19 mm;
end-diastolic mass: 147 g) with slight diffuse edema (global T2 mapping: 58 ms; normal value <50
ms) and marked increase of native T1 mapping (b–c) with global T1: 1220 ms (normal value<
1045 ms) and of ECV values (e, f) with global ECV of 42% (normal value <27%) suggestive of a
huge expansion of the extracellular space. LGE showed endocardial hyperitensity with a gradient
from the bases to the apex (d). CMR findings were suggestive for cardiac amyloid
5.4.1.2 Pathophysiology
A normal aortic valve has an opening area between 3 and 4 cm2; a progressive
reduction of the aortic valve area leads to a significant obstruction to the transvalvu-
lar flow and results in a transvalvular gradient. These haemodynamic changes cause
an increase in pressure afterload and ventricular wall stress that stimulates hypertro-
phy of the left ventricular myocardium [105, 106]. In the early stages of the disease,
left ventricular hypertrophy (LVH) reduces parietal stress and preserves the systolic
function of the left ventricle; over time, LVH can be maladaptive with literature
evidence that the hypertrophic myocardium represents an adverse prognostic marker
in various clinical conditions [107–109]. LVH in patients with aortic valve stenosis
is highly heterogeneous and is only weakly correlated with the extent of valve
obstruction; it is more closely associated with age, male sex and obesity [110–115].
The heterogeneity of the myocardial response in terms of LVH has critical prognos-
tic implications; it has been shown that with the same valvular obstruction, the find-
ing of an inappropriate left ventricular mass significantly increases the mortality of
patients with AS aortic [116]. The negative prognostic impact of an inappropriate
left ventricular mass can be related to an increase in myocyte apoptosis and intersti-
tial myocardial fibrosis deposition [117–119].
CT Imaging
CT with its high spatial resolution images of the aortic annuls and root represents a
central modality for planning of transcatheter procedure of aortic valve replacement
(TAVR/TAVI). In TAVI candidates, CT imaging provides a comprehensive assess-
ment, including the analysis of the aortic annulus, the characterization of the valve
anatomy (bicuspid or tricuspid), the burden of the aortic valve and aortic root calci-
fication, and the evaluation of the peripheral vascular accesses [121, 122] (Fig. 5.5).
The evaluation of aortic valve calcifications by CT has been proposed to estimate
the severity of AS when the echocardiographic parameters are discordant: the cut-
off value of AV calcifications associated with severe AS is ≥1274 AU in women and
≥2065 AU in men [123]. The evaluation of the aortic annulus anatomy is of para-
mount importance for a successful TAVI outcome. The annulus diameter dictates
the aortic prosthetic size, making this measurement a critical point for procedural
success; any error at this level could result in serious complications [124–126]. The
aortic annulus has an oval shape with long and short diameters; CT is exceptionally
accurate to obtain these measurements, as well as the perimeter and the area of the
aortic annulus [127]. Another critical parameter determining the success of TAVI is
5 Heart Diseases in Geriatric Patients 125
a b e
Fig. 5.5 CT angiography for planning of transcatheter aortic valve replacement in a 70-years-old
woman. CT images shows a fibrocalcific degeneration of the aortic valve (a), with tricuspid mor-
phology and moderate calcifications, characterized by severe stenosis with aortic valve planimetric
area equal to 0.7 cm2 (yellow area in b). CT images was used to characterize landing zone (multi-
planar and 3D reconstruction of the aortic root in c and d respectively) and pheriperal accesses (e)
the height of the coronary ostia from the annulus plane, since the prosthetic valve or
the native valve cusps displaced by the TAVI procedure, may cause coronary ostium
obstruction and may impede the coronary ostial flow [128]. The recommended
annulus-ostia length is >10–11 mm for the Edwards-Sapien valve, while there is no
recommendation for Core Valve [128].
CMR Imaging
Cardiac magnetic resonance (CMR) is not routinely used in the diagnostic work-
flow of AS; the main reason for this is that AS traditionally has been considered as
a disease of the valve while the myocardium has been largely ignored. On the
contrary, recent studies documented a progressive myocardial remodelling in AS
126 A. Palmisano et al.
5.4.2.1 Introduction
Mitral regurgitation is the second most frequent valvular heart disease in western
world [103]. Surgery is the first-choice in symptomatic patients with severe MR
[120]; however, surgery is often denied to elderly patients because of their comor-
bidities [134]. The denial of surgical treatment to elderly patients with significant
MR contributed to the tumultuous spread of percutaneous techniques in treating this
pathology [135] with imaging playing a pivotal role in procedural planning and
guiding.
CT Imaging
Transcatheter mitral valve repair should be performed when the heart team evalu-
ates a high surgical risk for the patient. In functional MR, TMVR is indicated in
patients with severe MR where medical therapy and cardiac resynchronization ther-
apy (CRT) are not effective [140]. In primary MR, TMVR is indicated in the pres-
ence of comorbidities such as CKD, COPB, advance age, and severe impairment of
left ventricular function.
Two main transcatheter mitral valve repair strategies for severe MR include
edge-to-edge repair (MitraClip) and annuloplasty rings (Cardioband) [141–143].
MitraClip is the selected treatment of moderate to severe degenerative and func-
tional MR, while the Cardioband system consists of a percutaneous annuloplasty
that can be used in settings of functional MR resulting from systolic dysfunction or
annular dilatation. As mentioned above, echocardiography is an initial tool to assess
mitral valve structure and disease; however, this can be challenging in patients with
limited acoustic windows also considering the complexity of the mitral valve appa-
ratus. In these cases, CCTA allows a full cardiac cycle acquisition, outlining similar
morphologic findings to echocardiography and MRI such as mitral annulus geom-
etry, dimensions and calcifications, morphology and mobility of valve and sub-
valve apparatus, with an improved spatial resolution. It can also be used to assess
the anatomy of coronary arteries and of the aortic valve and interatrial septum, but
it cannot assess the flow that can only be analysed via echocardiography or MRI
[144, 145]. Furthermore, CTCA can easily exclude the presence of an atrial throm-
bus which would represent a contraindication to percutaneous mitral valve repair.
Critical anatomic selection criteria for MitraClip device in functional MR are
Coaptation length (≥2 mm), and depth (<11 mm). In contrast, Flail gap (<10 mm)
and width (<15 mm) are the ones fundamental for MitraClip in primary MR
128 A. Palmisano et al.
MR Imaging
Even if echocardiography represents the main tool for diagnosis and follow-up of
MR, MRI remains the gold standard for comprehensive assessment of the pathology
and has shown higher accuracy than echocardiography to evaluate MR severity and
guiding surgery. Phase contrast sequences can directly or indirectly quantify MR
severity [147]. Tissue characterization with LGE and mapping may also help in the
identification of the aetiology of secondary MRI and may provide information about
myocardial remodelling.
5.5 Conclusion
References
1. Mehta N, Chokshi NP, Kirkpatrick JN. Cardiac imaging in the geriatric population: what do
we think we know, and what do we need to learn? Prog Cardiovasc Dis. 2014;57:204–14.
2. Pecer G. US bureau of the census: projections of the population of the United States, by age,
sex, and race: 1988 to 2080. Current Population Reports, Series P-25, No. 1018. Washington,
DC: US Government Printing Office; 1989.
3. Alexander KP, et al. Acute coronary care in the elderly, part I. Circulation. 2007;115:2549–69.
4. Zamorano JL, et al. EAE/ASE recommendations for the use of echocardiography in new trans-
catheter interventions for valvular heart disease. J Am Soc Echocardiogr. 2011;24:937–65.
5. Wong CY, Green P, Williams M. Decision-making in transcatheter aortic valve replace-
ment: the impact of frailty in older adults with aortic stenosis. Expert Rev Cardiovasc Ther.
2013;11:761–72.
6. Kuller LH, et al. 10-Year follow-up of subclinical cardiovascular disease and risk of coronary
heart disease in the cardiovascular health study. Arch Intern Med. 2006;166:71–8.
7. McClelland RL, Chung H, Detrano R, Post W, Kronmal RA. Distribution of coronary artery
calcium by race, gender, and age: Results from the Multi-Ethnic Study of Atherosclerosis
(MESA). Circulation. 2006;113:30–7.
8. Benjamin EJ, et al. Heart disease and stroke statistics’ 2017 update: a report from the
American Heart Association. Circulation. 2017;135(10):e146–603.
5 Heart Diseases in Geriatric Patients 129
29. Tanaka A, et al. Non-invasive assessment of plaque rupture by 64-slice multidetector com-
puted tomography—comparison with intravascular ultrasound. Circ J. 2008;72:1276–81.
30. Motoyama S, et al. Multislice computed tomographic characteristics of coronary lesions in
acute coronary syndromes. J Am Coll Cardiol. 2007;50:319–26.
31. Leipsic J, et al. SCCT guidelines for the interpretation and reporting of coronary CT angi-
ography: a report of the Society of Cardiovascular Computed Tomography Guidelines
Committee. J Cardiovasc Comput Tomogr. 2014;8:342–58.
32. Andreini D, et al. Coronary in-stent restenosis: assessment with CT coronary angiography.
Radiology. 2012;265:410–7.
33. Dai T, Wang JR, Hu PF. Diagnostic performance of computed tomography angiography in
the detection of coronary artery in-stent restenosis: evidence from an updated meta-analysis.
Eur Radiol. 2018;28:1373–82.
34. Chiurlia E, Menozzi M, Ratti C, Romagnoli R, Modena MG. Follow-up of coronary artery
bypass graft patency by multislice computed tomography. Am J Cardiol. 2005;95:1094–7.
35. Chan M, et al. A systematic review and meta-analysis of multidetector computed tomography
in the assessment of coronary artery bypass grafts. Int J Cardiol. 2016;221:898–905.
36. Frazier AA, et al. Coronary artery bypass grafts: assessment with multidetector CT in the
early and late postoperative settings. Radiographics. 2005;25:881–96.
37. Celeng C, et al. Anatomical and functional computed tomography for diagnosing hemody-
namically significant coronary artery disease: a meta-analysis. JACC Cardiovasc Imaging.
2019;12:1316–25.
38. Zhao R, Shan Y, Zou L, Zhao H, Zheng S. Solitary fibrous tumor of the seminal vesicle.
Medicine (Baltimore). 2019;9:10–3.
39. Branch KR, et al. Myocardial computed tomography perfusion. Cardiovasc Diagn Ther.
2017;7:452–62.
40. Danad I, et al. Diagnostic performance of cardiac imaging methods to diagnose ischaemia-
causing coronary artery disease when directly compared with fractional flow reserve as a
reference standard: a meta-analysis. Eur Heart J. 2017;38:991–8.
41. Ko BS, et al. Computed tomography stress myocardial perfusion imaging in patients
considered for revascularization: a comparison with fractional flow reserve. Eur Heart
J. 2012;33:67–77.
42. Broncano J, et al. Cardiac MRI in patients with acute chest pain. Radiographics. 2021;41:8–31.
43. Bodi V, et al. Prognostic value of a comprehensive cardiac magnetic resonance assessment
soon after a first ST-segment elevation myocardial infarction. JACC Cardiovasc Imaging.
2009;2:835–42.
44. Eitel I, et al. Comprehensive prognosis assessment by CMR imaging after ST-segment eleva-
tion myocardial infarction. J Am Coll Cardiol. 2014;64:1217–26.
45. Karamitsos TD, et al. Contained left ventricular rupture after acute myocardial infarction
revealed by cardiovascular magnetic resonance imaging. Circulation. 2012;125:2278–80.
46. Sharma A, Kumar S. Overview of left ventricular outpouchings on cardiac magnetic reso-
nance imaging. Cardiovasc Diagn Ther. 2015;5:464–70.
47. Ibanez B, et al. 2017 ESC guidelines for the management of acute myocardial infarction in
patients presenting with ST-segment elevation. Eur Heart J. 2018;39:119–77.
48. McAlindon EJ, et al. Measurement of myocardium at risk with cardiovascular MR: compari-
son of techniques for edema imaging. Radiology. 2015;275:61–70.
49. Mahrholdt H, Wagner A, Judd RM, Sechtem U. Assessment of myocardial viability by car-
diovascular magnetic resonance imaging. Eur Heart J. 2002;23:602–19.
50. McCrohon JA, et al. Differentiation of heart failure related to dilated cardiomyopathy and
coronary artery disease using gadolinium-enhanced cardiovascular magnetic resonance.
Circulation. 2003;108:54–9.
51. Rajiah P, Desai MY, Kwon D, Flamm SD. MR imaging of myocardial infarction.
Radiographics. 2013;33:1383–412.
52. de Waha S, et al. Prognosis after ST-elevation myocardial infarction: a study on cardiac mag-
netic resonance imaging versus clinical routine. Trials. 2014;15:1–9.
5 Heart Diseases in Geriatric Patients 131
98. Lin L, et al. The prognostic value of T1 mapping and late gadolinium enhancement cardio-
vascular magnetic resonance imaging in patients with light chain amyloidosis. J Cardiovasc
Magn Reson. 2018;20:2.
99. Rapezzi C, et al. Role of (99m)Tc-DPD scintigraphy in diagnosis and prognosis of hereditary
transthyretin-related cardiac amyloidosis. JACC Cardiovasc Imaging. 2011;4:659–70.
100. Falk RH, Quarta CC, Dorbala S. How to image cardiac amyloidosis. Circ Cardiovasc
Imaging. 2014;7:552–62.
101. Perugini E, et al. Noninvasive etiologic diagnosis of cardiac amyloidosis using
99mTc-3,3- diphosphono-1,2-propanodicarboxylic acid scintigraphy. J Am Coll Cardiol.
2005;46:1076–84.
102. Nkomo VT, et al. Burden of valvular heart diseases: a population-based study. Lancet.
2006;368:1005–11.
103. Iung B, et al. A prospective survey of patients with valvular heart disease in Europe: the Euro
Heart Survey on valvular heart disease. Eur Heart J. 2003;24:1231–43.
104. Otto CM. Calcific aortic stenosis—time to look more closely at the valve. N Engl J Med.
2008;359:1395–8.
105. Grossman W, Jones D, McLaurin LP. Wall stress and patterns of hypertrophy in the human
left ventricle. J Clin Invest. 1975;56:56–64.
106. Carabello BA. The relationship of left ventricular geometry and hypertrophy to left ventricu-
lar function in valvular heart disease. J Heart Valve Dis. 1995;4 Suppl 2:S132–8.; discus-
sion S138–9.
107. Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. Prognostic implications of echo-
cardiographically determined left ventricular mass in the Framingham Heart Study. N Engl
J Med. 1990;322:1561–6.
108. Spirito P, et al. Magnitude of left ventricular hypertrophy and risk of sudden death in hyper-
trophic cardiomyopathy. N Engl J Med. 2000;342:1778–85.
109. Schillaci G, et al. Continuous relation between left ventricular mass and cardiovascular risk
in essential hypertension. Hypertension. 2000;35:580–6.
110. Salcedo EE, et al. Determinants of left ventricular hypertrophy in patients with aortic steno-
sis. Cleve Clin J Med. 1989;56:590–6.
111. Kupari M, Turto H, Lommi J. Left ventricular hypertrophy in aortic valve stenosis: preventive
or promotive of systolic dysfunction and heart failure? Eur Heart J. 2005;26:1790–6.
112. Lavie CJ, Milani RV, Patel D, Artham SM, Ventura HO. Disparate effects of obesity and left
ventricular geometry on mortality in 8088 elderly patients with preserved systolic function.
Postgrad Med. 2009;121:119–25.
113. Dweck MR, et al. Left ventricular remodeling and hypertrophy in patients with aortic steno-
sis: insights from cardiovascular magnetic resonance. J Cardiovasc Magn Reson Off J Soc
Cardiovasc Magn Reson. 2012;14:50.
114. Gunther S, Grossman W. Determinants of ventricular function in pressure-overload hypertro-
phy in man. Circulation. 1979;59:679–88.
115. Orlowska-Baranowska E, et al. Influence of ACE I/D genotypes on left ventricular hypertro-
phy in aortic stenosis: gender-related differences. J Heart Valve Dis. 2004;13:574–81.
116. Cioffi G, et al. Prognostic effect of inappropriately high left ventricular mass in asymptomatic
severe aortic stenosis. Heart. 2011;97:301–7.
117. Hein S, et al. Progression from compensated hypertrophy to failure in the pressure-
overloaded human heart: structural deterioration and compensatory mechanisms. Circulation.
2003;107:984–91.
118. Bishopric NH, Andreka P, Slepak T, Webster KA. Molecular mechanisms of apoptosis in the
cardiac myocyte. Curr Opin Pharmacol. 2001;1:141–50.
119. Cheng W, et al. Stretch-induced programmed myocyte cell death. J Clin Invest.
1995;96:2247–59.
120. Baumgartner H, et al. 2017 ESC/EACTS guidelines for the management of valvular heart
disease. Eur Heart J. 2017;38:2739–91.
134 A. Palmisano et al.
121. Salgado RA, et al. Preprocedural CT evaluation of transcatheter aortic valve replacement:
what the radiologist needs to know. Radiographics. 2014;34:1491–514.
122. Tops LF, et al. Noninvasive evaluation of the aortic root with multislice computed tomog-
raphy implications for transcatheter aortic valve replacement. JACC Cardiovasc Imaging.
2008;1:321–30.
123. Clavel M-A, et al. The complex nature of discordant severe calcified aortic valve disease
grading: new insights from combined Doppler echocardiographic and computed tomographic
study. J Am Coll Cardiol. 2013;62:2329–38.
124. Willson AB, et al. 3-dimensional aortic annular assessment by multidetector computed
tomography predicts moderate or severe paravalvular regurgitation after transcatheter aortic
valve replacement: a multicenter retrospective analysis. J Am Coll Cardiol. 2012;59:1287–94.
125. Barbanti M, et al. Anatomical and procedural features associated with aortic root rupture dur-
ing balloon-expandable transcatheter aortic valve replacement. Circulation. 2013;128:244–53.
126. Blanke P, et al. Prosthesis oversizing in balloon-expandable transcatheter aortic valve
implantation is associated with contained rupture of the aortic root. Circ Cardiovasc Interv.
2012;5:540–8.
127. Delgado V, et al. Automated assessment of the aortic root dimensions with multidetector row
computed tomography. Ann Thorac Surg. 2011;91:716–23.
128. Ribeiro HB, et al. Coronary obstruction following transcatheter aortic valve implantation: a
systematic review. JACC Cardiovasc Interv. 2013;6:452–61.
129. Dweck MR, et al. Midwall fibrosis is an independent predictor of mortality in patients with
aortic stenosis. J Am Coll Cardiol. 2011;58:1271–9.
130. Krayenbuehl HP, et al. Left ventricular myocardial structure in aortic valve disease before,
intermediate, and late after aortic valve replacement. Circulation. 1989;79:744–55.
131. Taylor AJ, Salerno M, Dharmakumar R, Jerosch-Herold M. T1 mapping basic techniques and
clinical applications. JACC Cardiovasc Imaging. 2016;9:67–81.
132. Flett AS, et al. Diffuse myocardial fibrosis in severe aortic stenosis: an equilibrium con-
trast cardiovascular magnetic resonance study. Eur Hear journal Cardiovasc Imaging.
2012;13:819–26.
133. Flett AS, et al. Equilibrium contrast cardiovascular magnetic resonance for the mea-
surement of diffuse myocardial fibrosis: preliminary validation in humans. Circulation.
2010;122:138–44.
134. Mirabel M, et al. What are the characteristics of patients with severe, symptomatic, mitral
regurgitation who are denied surgery? Eur Heart J. 2007;28:1358–65.
135. Sala A, Alfieri O. Percutaneous treatment of mitral valve regurgitation: where do we stand?
Int J Cardiol. 2019;288:137–9.
136. Levine RA, Schwammenthal E. Ischemic mitral regurgitation on the threshold of a solution:
from paradoxes to unifying concepts. Circulation. 2005;112:745–58.
137. Enriquez-Sarano M, et al. Functional anatomy of mitral regurgitation: accuracy and outcome
implications of transesophageal echocardiography. J Am Coll Cardiol. 1999;34:1129–36.
138. Dulgheru R, Bruls S, Lancellotti P. How I look at the regurgitant mitral valve—a stepwise
echocardiographic assessment. Eur Hear journal Cardiovasc Imaging. 2021;22:491–3.
139. Lancellotti P, et al. Recommendations for the echocardiographic assessment of native valvu-
lar regurgitation: an executive summary from the European Association of Cardiovascular
Imaging. Eur Heart J Cardiovasc Imaging. 2013;14:611–44.
140. Stone GW, et al. Transcatheter mitral-valve repair in patients with heart failure. N Engl J
Med. 2018;379:2307–18.
141. Feldman T, Young A. Percutaneous approaches to valve repair for mitral regurgitation. J Am
Coll Cardiol. 2014;63:2057–68.
142. Feldman T, et al. Percutaneous mitral repair with the MitraClip system: safety and midterm
durability in the initial EVEREST (Endovascular Valve Edge-to-Edge REpair Study) cohort.
J Am Coll Cardiol. 2009;54:686–94.
5 Heart Diseases in Geriatric Patients 135
143. Maisano F, et al. Cardioband, a transcatheter surgical-like direct mitral valve annuloplasty
system: early results of the feasibility trial. Eur Heart J. 2016;37:817–25.
144. Delgado V, et al. Assessment of mitral valve anatomy and geometry with multislice computed
tomography. JACC Cardiovasc Imaging. 2009;2:556–65.
145. Alkadhi H, et al. Mitral annular shape, size, and motion in normals and in patients with
cardiomyopathy: evaluation with computed tomography. Investig Radiol. 2009;44:218–25.
146. Feuchtner GM, et al. Cardiac CT angiography for the diagnosis of mitral valve prolapse:
comparison with echocardiography1. Radiology. 2010;254:374–83.
147. Uretsky S, Argulian E, Narula J, Wolff SD. Use of cardiac magnetic resonance imaging in
assessing mitral regurgitation: current evidence. J Am Coll Cardiol. 2018;71:547–63.
Vascular Diseases in Geriatric Patients
6
Gloria Caredda, Giuseppe Guglielmi, and Luca Saba
6.1 Introduction
Over time, the mean age of the population has progressively increased, with a large
number of geriatric subjects. It is predicted that in 2030 they will represent the 19%
of the general community in the USA, with about 19 millions of people over
85 years of age, with an increase of 22% of the geriatric population in 2050 and of
32% in 2100.
In this scenario, there has been a parallel increase in the number of the age-
related diseases, thus it is possible to consider the presence of a concrete “demo-
graphic transition.” In particular, the geriatric syndromes play a fundamental role,
and they are generally related to a vascular chronic disease. More in depth, about
40 millions of people over 65 years of age are affected by a cardiovascular disease,
which, together with other cerebrovascular conditions, represents the main cause of
death in this age range, constituting an important healthcare, economic, and
social issue.
Such a relative matter leads to the necessity of developing new prevention and
treatment strategies, considering the vascular diseases at the base of a syndromic
process, which might require a different patient management with respect to the
single disease affecting a healthy subject. This is important, in particular, for the
female patients, because, with aging and the hormone physiological changes, these
diseases often occur with an insidious onset.
Generally, the aging process of the cardiovascular system is characterized by a
stiffening of the vascular walls and the occurrence of atherosclerosis, affecting both
the large vessels and the microvasculature, leading to organic systemic alterations.
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 137
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_6
138 G. Caredda et al.
The main risk factors related to vessel aging, which lead to vasculature system
diseases, are represented by patient aging, smoking, other conditions such as diabe-
tes mellitus, hypertension, hyperlipidemia, and other factors (some of them non-
modifiable) like race, ethnicity (with an increased risk in African American and
Hispanic subjects), chronic kidney disease, metabolic syndrome, high levels of
C-reactive protein, β2-microglobulin, cystatin C, lipoprotein, and homocysteine.
Among these factors, the main modifiable one is smoking, which increases the risk
of peripheric vascular disease by about four times, with an onset that precedes by
10 years than that in non-smokers. Moreover, smoking patients usually have a worse
prognosis.
Aside from these conditions, in geriatric patients, vascular trauma plays a key
role. Their incidence is lower if compared to their young counterpart, but, at the
same time, they are more severe. Indeed, in this age range, the mortality for vascular
trauma (40% for falls) is higher than that of non-geriatric adults and, with respect to
young people, death is more frequent in the emergency departments. The main
cause is represented by car accidents, followed by falls (with an incidence of 32%
in subjects over 75 years of age), with a predominance of abdominal and upper
limbs harms, more frequent than in non-geriatric adults.
This is related to the physiological processes of this age range, with alterations
affecting the nervous and the muscle-skeletal systems, which involve balance, mak-
ing these patients more susceptible to falls. Furthermore, if multiple diseases are
present, trauma management is more challenging, thus determining high morbidity
and mortality [1–3].
In the geriatric population, some aging-related changes show an impact in the car-
diovascular system, both as microscopic and as macroscopic alterations, leading to
several organic diseases. In particular, the oxidative stress, caused by high levels of
reactive oxygen species (ROS), has a crucial role in vasculature aging, reducing the
production of the endothelium-derived nitric oxide (NO), thus leading to vascular
stiffening, low parietal elasticity, and reduced tissue perfusion. Even aged dysfunc-
tional mitochondria contribute to the production of ROS. Moreover, while in young
subjects the result of ROS activity induces the activation of an antioxidant pathway,
characterized by the production of Nrf2, the aging-related changes inactivate this
process, making vasculature more sensible to ROS activity and more prone to
inflammation, with an increase of endothelial apoptosis. This is also due to proin-
flammatory changes in gene expression in the endothelial and smooth muscle cells,
which contribute to induce several conditions, from atherogenesis to microvascula-
ture diseases and aneurysm formation, by promoting the production of proinflam-
matory cytokines, determining the “senescence-associated secretory phenotype.”
Furthermore, senescent endothelial cells show a reduced angiogenetic and repara-
tive ability.
6 Vascular Diseases in Geriatric Patients 139
The expression “peripheral artery disease” (PAD) includes the conditions involving
the arteries aside from the cranial and the cardiac territories. About 200 million
people in the world are affected by PAD, with a similar prevalence between men and
postmenopausal women. More frequently it is caused by atherosclerosis, which is
considered the most important determinant, but it can be due to diabetes mellitus,
smoking [6], vasculitis and other noninflammatory arteriopathies as well. This con-
dition is usually combined with coronary artery disease (CAD) and cerebrovascular
disease (CVD). An association between the lower extremity, the carotid, and the
renal arterial territory disease is frequent as well [7].
Two forms of PAD can be distinguished, the proximal one, which affects the
aortoiliac and the femoropopliteal tracts, and the distal subtype, involving the ter-
ritories distal to the popliteal arteries. In some patients, the latter type is character-
ized by a calcification of the middle layer of the wall, leading to a poorly compressible
vessel and a high mortality (Practice 2016).
PAD can be assessed by estimating the segmental blood pressure in different levels
of the limb and evaluating the ankle-brachial index, the ratio between the systolic
blood pressure measured in the ankle and the systolic blood pressure assessed in the
arm, measured after 5–10 min of rest in a supine position [8]. The normal and patho-
logic values are reported in Table 6.1. In subjects with non-compressible arteries,
instead, the toe-brachial index might be assessed. Even exercise testing may be
valuable, using a treadmill or measuring the maximum walking times without
symptoms. Finally, in cases of critical leg ischemia, the evaluation of transcutane-
ous oximetry provides the clinician with important information (Practice 2016).
140 G. Caredda et al.
6.5 Imaging
The first-line technique for PAD evaluation is ultrasonography (US) with Doppler
imaging. It is a noninvasive and highly available tool, able to assess the atheroscle-
rotic plaque and monitor the patency of the revascularized vessels (Practice 2016).
The typical flow pattern of arterial vessels is triphasic, due to the high resistance of
muscle territory irroration, but it is lost in case of exercise or ischemia [9]. The
severity of the disease varies according to the grade of stenosis and to the value of
peak systolic velocity (PSV) and velocity ratio (VR), which are summarized in
Table 6.2. In particular, a stenosis of 50–99% is hemodynamically significant when
the PSV at the level of the lesion is double than that measured in a proximal arterial
segment (>200 cm/s with turbulence) [9] (Трансплантати 2017).
In particular, Doppler US (DUS) is the first-line imaging technique for the
assessment of the carotid territories, being able to distinguish patients with a high
risk for ipsilateral ischemic stroke and identify those subjects who are candidate for
endarterectomy or, on the other hand, for the only best medical treatment [8]
(Fig. 6.1).
a b
Fig. 6.1 (a–c) A 75-year-old male patient with left stroke. The MRI TOF shows the stenosis of
70% NASCET at the origin of the ICA (white open arrow). The FAT SAT MPRAGE shows the
presence of hyperintese signal in the plaque due to the presence of IPH
a b
Fig. 6.2 (a, b) A 78-year-old male patient with right stroke. The CTA shows in the right ICA the
presence of plaque in the ICA characterized by hypodense components and positive rim sign
(white arrows)
142 G. Caredda et al.
a b
Fig. 6.3 (a, b) A 84-year-old female patient. The CTA shows bilateral calcified plaque
CT angiography is also the first-line method for the evaluation of patients with
acute mesenteric ischemia and renal artery disease, providing a map of the arterial
vessels and demonstrating the presence of vascular thrombi [8].
The atherosclerotic plaque may show some features that might increase the risk for
stroke, which can be detected through diagnostic imaging evaluation. The presence
of intraplaque hemorrhage, a lipid-rich necrotic core (LRNC) and the assessment of
the fibrous cap is well evaluated with MRI. In particular, CT is able to detect the
lipidic plaque components as well, but MRI better discriminates between LRNC
and intraplaque hemorrhage. In addition, MRI also provides information about a
thin or damaged fibrous cap.
Moreover, a plaque is considered active when intraplaque inflammation and neo-
vascularization are detected, better estimated with a CT scan.
Other information about plaque vulnerability are obtained with the evaluation of
plaque thickness and surface morphology, both well analyzed through DUS, CT,
and MRI. In particular, among the three possible types of plaque surface (smooth,
irregular, or ulcerated), the ulcerated form has the major risk for stroke.
Finally, it is useful to assess the volume of the plaque, with MRI providing better
information about soft tissue contrast and CT allowing a better spatial resolu-
tion [12].
6.7 Treatment
The main therapeutic strategy is to remove the cardiovascular risk factors, through
healthy life choices and medications. In addition, regular walking exercise is able to
increase the development of the collateral circulation, improving the claudication
pain. Finally, the last therapeutic approach is revascularization, performed in
patients with critical ischemia (Practice 2016).
6.7.1 Aneurysms
Another condition, typical for old adults, is the development of aneurysms, found in
the 3–4% of subjects older than 65 years of age, more frequently in the infrare-
nal aorta.
Aortic aneurysms are defined as a weakness of the aortic wall with the presence
of an aortic diameter >30 mm and an increase of more than 50% with respect to the
normal vessel, determining a dilatation with a saccular or fusiform morphology. The
aneurysms with a diameter >55 mm in men and >50 mm in women are at risk of
rupture.
144 G. Caredda et al.
6.8 Diagnosis
In most cases, aneurysms are an incidental finding, casually noticed in other exami-
nations or identified thanks to their typical calcifications pattern. In other subjects,
they are clinically detected because of the presence of an abdominal palpable mass.
However, more frequently aneurysms are recognized through diagnostic imaging.
US, CT, and MR imaging (MRI) are the methods usually performed in order to
obtain aneurysm’s information such as its diameters [13–16]. The important mea-
surements that should be evaluated when assessing an aortic aneurysm include the
aneurysmal sac, the arterial portions proximal and distal to the dilatation, and the
features of the vascular access that would be used in case of stent-graft positioning,
in particular the ilio-femoral tract, including calcifications and thrombi [16].
6.8.1 Ultrasound
In patients with a non-ruptured aneurysm, US and DUS are the main techniques for
disease screening and monitoring. Indeed, they are noninvasive methods which
allow arterial diameter measurement with high reproducibility.
6 Vascular Diseases in Geriatric Patients 145
With 2D imaging, MIP and MPR images, CT angiography is able to assess the
aneurysm’s diameters, providing important information about the necessity for
operative treatment.
Before contrast administration, a first scan is useful for identifying parietal cal-
cifications in the aorto-iliac tract. An important pitfall that should be considered is
represented by vascular tortuosity, which might lead to obtain an erroneous value of
the vessel diameters, as well as their measurement in an axial plain.
In addition, CT has a pivotal role in examining the vascular anatomy for a proper
procedural planning, identifying the correct device suitable for the single patient, by
assessing the aorto-iliac length and the aneurysm’s diameters.
Finally, CT has a higher sensitivity than conventional angiography in identifying
the possibility of procedural and post-procedural complications, especially after
EVAR, more frequently represented by endoleaks [14, 16].
The last studies analyze the disease from a wider point of view, considering not only
the vascular diameters but also the properties of the arterial wall. Indeed, a different
risk of progression and rupture has been observed in aneurysms with similar
146 G. Caredda et al.
diameters. The use of particular tracers (i.e., radio-labeled blood cells) allows the
localization of inflammatory and metabolic changes in the vascular walls. The main
technique used for functional imaging is single-photon emission computed tomog-
raphy (SPECT) with the employment of radioisotopes such as 99mTc, 111In, 123I, and
131
I. With this technique, the activity of the parietal thrombi can be evaluated, by
detecting the focal activity of platelets and polymorphonucleates [14].
Several molecular probes are also used with techniques such as MRI, magnetic
resonance spectroscopy (MRS), PET, optical bioluminescence, optical fluores-
cence, and targeted US. In the different stages of the disease, alterations in particu-
lar markers are detectable, providing important information that go beyond the
measurement of the vascular diameter by itself. Indeed, an increment of the parietal
activity is considered expression of wall inflammation, correlated with a major risk
of rupture. Even modifications in extracellular components such as elastin and col-
lagen are indicative for parietal instability, and thus increased risk of rupture [13,
14]. For instance, MRI images can be acquired using a particular probe, which
consist in the ultrasmall superparamagnetic particles of iron oxide (USPIO), which
marks areas of inflammation by detecting the activity of the macrophages. All of
these information provided can be used as a guide for appropriate pre-procedural
decision-making [15].
6.9 Treatment
a b
Fig. 6.4 (a, b) A 81-year-old male patient with AAA. The CTA shows the presence of infrarenal
aneurysm with endoluminal moderate eccentric thrombus (white arrows)
a b
Fig. 6.5 (a, b) A 75-year-old male patient with AAA. The CTA shows the presence of infrarenal
aneurysm with endoluminal severe eccentric thrombus (white arrows)
148 G. Caredda et al.
References
1. Olin JW, Sealove BA. Peripheral artery disease: current insight into the disease and its
diagnosis and management. Mayo Clin Proc. 2010;85(7):678–92. https://doi.org/10.4065/
mcp.2010.0133.
2. Strandberg TE, Pitkälä KH, Tilvis RS, O’Neill D, Erkinjuntti TJ. Geriatric syndromes—vascu-
lar disorders? Ann Med. 2013;45(3):265–73. https://doi.org/10.3109/07853890.2012.727022.
3. Ungvari Z, Tarantini S, Donato AJ, Galvan V, Csiszar A. Mechanisms of vascular aging. Circ
Res. 2018;123(7):849–67. https://doi.org/10.1161/CIRCRESAHA.118.311378.
4. Alvis BD, Hughes CG. Physiology considerations in geriatric patients. Anesthesiol Clin.
2015;33(3):447–56. https://doi.org/10.1016/j.anclin.2015.05.003.
5. Kovacic JC, Moreno P, Nabel EG, Hachinski V, Fuster V. Cellular senescence, vascular dis-
ease, and aging: part 2 of a 2-part review: clinical vascular disease in the elderly. Circulation.
2011;123(17):1900–10. https://doi.org/10.1161/CIRCULATIONAHA.110.009118.
6. Thiruvoipati T, Kielhorn CE, Armstrong EJ. Peripheral artery disease in patients with diabetes:
epidemiology, mechanisms, and outcomes. World J Diabetes. 2015;6(7):961–9. https://doi.
org/10.4239/wjd.v6.i7.961.
7. Imori Y, Akasaka T, Ochiai T, Oyama K, Tobita K, Shishido K, Nomura Y, Yamanaka F,
Sugitatsu K, Okamura N, Mizuno S, Arima K, Suenaga H, Murakami M, Tanaka Y, Matsumi
J, Takahashi S, Tanaka S, Takeshita S, Saito S. Co-existence of carotid artery disease, renal
artery stenosis, and lower extremity peripheral arterial disease in patients with coronary artery
disease. Am J Cardiol. 2014;113(1):30–5. https://doi.org/10.1016/j.amjcard.2013.09.015.
8. Vlachopoulos C, Georgakopoulos C, Tousoulis D. Diagnostic modalities in peripheral artery
disease. Curr Opin Pharmacol. 2018;39:68–76.
9. Verim S, Taşçı I. Doppler ultrasonography in lower extremity peripheral arterial disease. Turk
Kardiyol Dern Ars. 2013;41(3):248–55. https://doi.org/10.5543/tkda.2013.76429.
10. Lau JF, Weinberg MD, Olin JW. Peripheral artery disease. Part 1: clinical evaluation
and noninvasive diagnosis. Nat Rev Cardiol. 2011;8(7):405–18. https://doi.org/10.1038/
nrcardio.2011.66.
11. Mathew RC, Kramer CM. Recent advances in magnetic resonance imaging for peripheral
artery disease. Vasc Med. 2018;23(2):143–52. https://doi.org/10.1177/1358863X18754694.
12. Saba L, Saam T, Jäger HR, Yuan C, Hatsukami TS, Saloner D, Wasserman BA, Bonati LH,
Wintermark M. Imaging biomarkers of vulnerable carotid plaques for stroke risk predic-
tion and their potential clinical implications. Lancet Neurol. 2019;18(6):559–72. https://doi.
org/10.1016/S1474-4422(19)30035-3.
13. Brangsch J, Reimann C, Collettini F, Buchert R, Botnar RM, Makowski MR. Molecular
imaging of abdominal aortic aneurysms. Trends Mol Med. 2017;23(2):150–64. https://doi.
org/10.1016/j.molmed.2016.12.002.
14. Hong H, Yang Y, Liu B, Cai W. Imaging of abdominal aortic aneurysm: the present and the future.
Curr Vasc Pharmacol. 2010;8(6):808–19. https://doi.org/10.2174/157016110793563898.
15. Jalalzadeh H, Indrakusuma R, Planken RN, Legemate DA, Koelemay MJ, Balm
R. Inflammation as a predictor of abdominal aortic aneurysm growth and rupture: a systematic
6 Vascular Diseases in Geriatric Patients 149
Abbreviations
B/A Broncho-arterial
BAF Bronchial anthracofibrosis
COPD Chronic obstructive pulmonary disease
CT Computed tomography
CXR Chest radiography
DTS Digital tomosynthesis
FEV1 forced expiratory volume in 1 second
FVC Force vital capacity
HRCT High-resolution CT
HU Hounsfield unit
ILD Interstitial lung diseases
LAC Large airway collapse
TD Tracheal diverticula
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 151
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_7
152 M. Balbi et al.
7.1 Introduction
Chest radiography (CXR) remains the first-line imaging for many thoracic diseases,
showing a generally good performance as a screening evaluation [13]. If the accu-
racy of such modality is rather limited for evaluating small airways due to intrinsic
limitations (i.e., limited resolution and superimposition of overlapping structures),
large airway abnormalities can be relatively easily demonstrated on radiographic
images, regardless of age [14]. Digital chest radiography has continuously and sig-
nificantly improved over the last decades, with several processing tools being imple-
mented to support radiologists in the detection of pathological findings, including
digital tomosynthesis (DTS) and dual-energy subtraction techniques [15, 16]. DTS
has been demonstrated to overcome conventional radiography in the assessment of
central airways, improving their coronal view and thus facilitating the identification
of a suspected either ingested or aspirated foreign body, which may be of value in
geriatric patients [17]. Analogously to DTS, images obtained with dual-energy sub-
traction improve the detection of tracheal abnormalities. Tracheal stenosis and nar-
rowing due to extrinsic compression can be, in fact, more easily demonstrated on
soft-tissue-selective images [18].
Such techniques, however, are not free of limitations, including their relatively
low availability, whereas chest CT, considered the imaging modality of choice, is
much largely accessible.
Chest high-resolution CT (HRCT) is the current imaging modality of choice for the
evaluation of airways. The two consistent components of HRCT include the use of
thin sections (≤1.5 mm) coupled with a high spatial frequency reconstruction algo-
rithm [19]. Although there are no standardized HRCT protocols for the assessment
of the airways, acquisition of volumetric scanning has several advantages compared
to interspaced scanning (i.e., obtained with 10–20 mm intervals between axial
scans), which is prone to miss focal abnormalities. Volumetric HRCT allows both
the evaluation of the airways in a true cross-section using multiplanar reconstruc-
tion and application of post-processing techniques (e.g., minimum intensity projec-
tion and maximum intensity projection) as well as quantitative software [19, 20].
Technical parameters ought to be optimized to avoid motion artifacts, which
deserve particular attention in the elderly. Possible strategies include caudocranial
scanning, increased pitch, and faster rotation time [21]. The window setting used to
interpret HRCT images is of utmost importance since it may significantly affect the
apparent bronchial walls thickness, best assessed with window width centers
between −250 and −750 HU and window width >1000 HU. Other settings, particu-
larly window width <1000 HU, can lead to a substantial artificial thickening of
bronchial walls [22].
154 M. Balbi et al.
The trachea is considered to increase in size and become irregular in shape over
time. Gibellino et al. observed a significant difference in tracheal diameters and
cross-sectional area between a group of supposed healthy young men (n = 18; mean
age, 21.1; mean coronal diameter, 1.79 cm; mean sagittal diameter 2.00 cm; mean
cross-sectional area, 2.81 cm2) and middle-age men (n = 32; mean age, 52.2; mean
coronal diameter 1.91 cm; mean sagittal diameter 2.14 cm; mean cross-sectional
area, 3.22 cm2) [25]. However, neither patients greater than 61 years of age nor
women were included in the analysis. Despite the small sample size and the rela-
tively young subjects included, these results were in keeping with a previous study
that observed a nearly steady increase in sagittal and coronal tracheal diameters
from the third decade onward in 808 healthy subjects aged 10–79 years (430 males
and 378 females) measured on radiographic images [26]. Collins et al. confirmed a
significant positive correlation between age and radiographic tracheal size (r values
between 0.324 and 0.603; p < 0.01) in 165 healthy nonsmokers (79 male and 86
females; mean age, 50 years) [27]. By means of computed tomography (CT), Sakai
et al. demonstrated a significant positive correlation between tracheal area and age
(r = 0.37; p = 0.0006) in 83 healthy male volunteers aged 21–83 years (mean age,
47.7 years) [28]. The same study also demonstrated that the trachea tends to become
more irregular in shape with increasing age, as proven by a loss in its circularity
(r = −0.32; p = 0.003) [28].
A more recent study, where 81 healthy volunteers aged 25–75 years (40 female
and 41 male) underwent a chest CT at total lung capacity and during forced exhala-
tion, showed that older men (mean age, 61.8 years) had 12% greater cross-sectional
total area at total lung capacity than younger men (mean age, 32.2 years) (p = 0.02).
However, no subjects aged >75 years were enrolled [29]. Most notably, men, but not
women, showed a significant positive correlation between the percentage of airway
collapse and age (R2 = 0.40; p < 0.001) up to and exceeding the value of 50%, which
represents the most reported threshold to define large airway collapse (LAC) [30].
These results have extended knowledge in the age-related changes of the trachea,
suggesting that both age and sex should be considered in the diagnostic evaluation
of expiratory dynamic airway collapse to not mistake a normal response to expira-
tory maneuvers for a disease state.
Tracheal cartilaginous rings can become calcific as an age effect. This finding is
almost always incidental and does not have a recognized functional significance
(Fig. 7.1). In a chest radiographic series, it was reported in 37% of patients aged
75 years or older [31]. Lloyd et al. described tracheobronchial calcification in 26%
7 Airway Diseases in Geriatric Patients 155
a b
Fig. 7.1 (a, b) Coronal unenhanced CT images of a 79-year-old woman show tracheobronchial
calcification rings. In (b), the calcifications are magnified by means of maximum-intensity-
projection (MIP) reconstruction
of patients aged 40–45 years and in up to 65% of men and 40.5% of women aged
60–79 years undergoing chest CT for a wide range of suspected both mediastinal
and pulmonary disorders [32]. Of note, tracheal calcification is not an exclusive
manifestation of normal aging but has been associated with other conditions, includ-
ing adrenogenital syndrome, diastrophic dysplasia, and a history of warfarin ther-
apy [33]. When nodular in shape (8–10 mm) and adjacent to thickened tracheal
cartilage, calcifications may result from a benign disorder called tracheobroncho-
pathia osteochondroplastica. In this condition, however, the trachea appears much
more irregular in shape than normal, making the diagnosis quite straightfor-
ward [34].
association between higher age and a lower airway wall thickness at an internal
parameter of 10 mm (i.e., a standardized measurement of airway wall thickness
obtained from the regression line between the square root of the airway wall area
and the internal perimeter of the airway) in 99 healthy subjects (median age,
39 years; interquartile range, 22–54) [37]. Such measurements were performed
using an automated software, potentially more reproducible and accurate than man-
ual measurements [38]. These results are partly in keeping with those of Zach et al.,
who demonstrated a significant (p = 0.006) decrease of wall area percent (i.e., wall
area/total bronchial area) and an increase of internal lumen area with age in a cohort
of 92 healthy subjects aged 45–80 years [39].
It is worth emphasizing that no established criteria exist to define bronchial wall
thickening, which remains a subjective diagnosis. Moreover, current CT metrics to
evaluate bronchial wall thickness are affected by potential bias, such as lung vol-
ume, reconstruction parameters (i.e., see High-Resolution CT paragraph), underly-
ing airway disease, and limited capability of CT of distinguishing bronchial wall
from peribronchovascular interstitium [40, 41]. Of note, bronchial wall thickening
can be underestimated when the bronchial diameter is used for comparison in
patients with bronchial dilation (e.g., in case of bronchiectasis or suboptimal infla-
tion). In contrast, thickening of the peribronchovascular interstitium may result in
what appears to be bronchial wall thickening. Therefore, the effects of age on air-
way wall thickness are not easy to be ascertained.
The most used descriptive parameter to evaluate bronchial caliber is the broncho-
arterial (B/A) ratio, which is obtained by dividing the diameter of the bronchus
(most commonly the internal bronchial diameter) by the diameter of the adjacent
pulmonary artery. As a rule of thumb, the B/A ratio is regarded as abnormal when it
exceeds the value of 1, meeting the radiological definition of bronchiectasis [42].
There is evidence that the B/A ratio increases with age, reaching bronchiectatic
dimensions in some cases (Fig. 7.2). Matsuoka et al. found a significant correlation
between the B/A ratio and age (r = 0.768, p < 0.0001) in a group of 85 healthy sub-
jects. The B/A ratio was greater than 1 in 41% of subjects aged >65 years and in 7%
of subjects aged between 41 and 64 years but in no subjects aged between 21 and
40 years [35]. As stated above, Copley et al. observed that bronchial dilation was
more prevalent in older (>75 years) than younger (<55 years) healthy subjects (60%
vs. 6%; p < 0.001) [36]. Moreover, bronchial dilation was found to present a lesser
extent in the younger than in the older group (p < 0.001), in which it also showed a
lower lobe predominance (i.e., 80% of cases) [36].
Notably, an increase of the B/A ratio can be driven, at least to some extent, by a
diameter reduction of the pulmonary arteries rather than a true bronchial dilation. This
phenomenon can be due to a disease state (e.g., asthma) [43] but also found in healthy
subjects, including those who live at high altitudes (exposed to hypoxia) [44] and never
smokers [45]. Nevertheless, the lack of standardized age-specific reference values of
7 Airway Diseases in Geriatric Patients 157
a b
Fig. 7.2 (a) Axial unenhanced CT image of a healthy 23-year-old-man shows a broncho-arterial
ratio <1 (arrow) at the right lower lobe posterior segment. (b) At the same level, the broncho-
arterial ratio is >1 (arrow) in a 70-year-old-man with no history of cardiopulmonary disease.
Recognition of an age-related increase of broncho-arterial ratio is essential to avoid a bronchiecta-
sis overdiagnosis in otherwise healthy older individuals
airway and vessel size currently prevents understanding how they relate to each other
with increasing age. Despite these well-known limitations, recognizing a B/A ratio
greater than 1 as a possible age-related finding is essential to reduce the chance of a
spurious diagnosis of “bronchiectasis” in an otherwise healthy old individual.
aged 21–30 years, 7 of 17 (41%) aged 41–50 years, 11 of 17 (65%) aged 51–60 years,
and 13 of 17 (76%) aged 61 years or older [51]. The increase in the frequency of air
trapping with age was found statistically significant (p < 0.05), as well as the correla-
tion between air trapping extension and age (r = 0.523, p < 0.001) [51]. Thus, it was
suggested that airway occlusion or luminal narrowing might occur with normal aging,
although only a few over 75-year olds were included, and no histological sample was
obtained. Subsequent work extended these findings by exploring a larger population
of more advanced age and providing data from lung specimens. By means of paramet-
ric response mapping (i.e., a voxel-based image analysis that classifies lung by thresh-
olding Hounsfield unit, HU, values from spatially aligned inspiratory/expiratory CT
scans), Martinez et al. found that nonemphysematous air trapping increased by 2.7%
per decade in a population of 580 never- and ever-smokers free from obstruction and
respiratory symptoms (i.e., from 3.6%, if aged 40–50 years, to 12.7% when aged
70–80 years) [52]. Increasing air trapping was associated with increased force vital
capacity (FVC) (p = 0.004) but unchanged forced expiratory volume in 1 second
(FEV1) (p = 0.94), yielding lower FEV1/FVC ratios (p < 0.001) [52]. In keeping with
these results, Verleden et al. demonstrated an age-dependent loss of terminal bronchi-
oles in 32 never-smoker lung donors (age range 16–83 years) using a combination of
ex vivo CT, whole lung micro-CT, and micro-CT of extracted cores. The loss of ter-
minal bronchioles (i.e., about half of the total number of terminal bronchioles between
30 and 80 years of age) was corroborated by the association of the predicted pulmo-
nary function with the total number of terminal bronchioles, suggesting that loss of
small airways is a relevant structural component of age-related decline in pulmonary
function of healthy individuals [53].
Of note, age-related changes in CT lung density are not exclusively attributable
to a small airway involvement. Several studies have focused on the presumptive
relationship between the physiological decrease in lung density with age (i.e.,
approximately 50 HU between 20 and 70 years of age) and the progressive enlarge-
ment of the airspaces over time, a phenomenon conventionally mislabeled as “senile
emphysema” (despite the absence of destruction of alveolar walls, implicit in the
word emphysema) [54–57]. In non-smoker elderlies, lung attenuation possibly
approaches and even falls below the density thresholds commonly used to define
emphysema (i.e., 910 HU or 950 HU), demanding caution when applying densitom-
etry in aging lungs to avoid mistaking normal parenchyma for damage [57, 58].
Nevertheless, recent evidence suggests that age-related emphysematous changes
may be of limited importance because of their low extent (i.e., <5% of the whole
lung) and substantial stability over time (i.e., increase of about 0.1% per 10 years
between the 50 and 80 years of age) [52].
Chronic obstructive pulmonary disease (COPD) represents the fourth leading cause
of death worldwide, with increasing prevalence in the elderly [3]. The estimated
prevalence of COPD is more than 390 million people worldwide in 2030 [59]. To
7 Airway Diseases in Geriatric Patients 159
date, various phenotypes of COPD have been described, with chronic bronchitis and
emphysema accounting for the most common ones. The former is characterized by
predominant airway-related changes (inflammation and airway wall thickening)
with increased mucus production, whereas the latter by alveolar wall destruction
and hyperinflation, resulting in impaired gas exchange [60]. Notably, it has been
suggested that the small airway damage represents the primum movens of such het-
erogeneous disorder, even in the emphysematous phenotype [61].
Regardless of the phenotype, establishing the presence of COPD in the elderly
can be quite challenging due to the significant morphological similarities between
aged lung and COPD. The physiological airspace enlargement without the alveolar
wall destruction observed in COPD is often erroneously labeled as “senile emphy-
sema” [62, 63]. The risk of such misinterpretation is over-diagnosing COPD within
the geriatric population, potentially leading to unnecessary investigations. Having
said that, it is worth emphasizing that the diagnosis of COPD relies on both clinical
and functional evidence of expiratory airflow obstruction [64], whereas imaging
(mainly CT) normally serves the purpose of assessing the disease extent (emphyse-
matous phenotype) and exacerbations (bronchitis phenotype). Measurement of
emphysema severity by means of CT densitometry techniques has been established
in the literature [65, 66], and different quantitative approaches—whose discussion
would deserve a dedicated chapter—have been employed more recently [67–69].
Chronic bronchitis is demonstrated by a relative increase in bronchial wall thickness
as compared to the bronchial lumen and with the diameter of adjacent pulmonary
arteries, a rather subjective morphological feature, as discussed above, while poorly
defined centrilobular nodules of ground-glass attenuation represent small airway
inflammation [70].
Pulmonary cysts deserve special attention since they can be misinterpreted as
bullous emphysema, although they represent an independent entity. The depiction
of thin-walled air spaces on CT may help distinguish pulmonary cysts from emphy-
sema [71].
7.4.2 Bronchiectasis
LAC refers to an excessive inward movement of the trachea and/or main bronchi
during expiration [30]. LAC comprises two entities: tracheomalacia, where there is
softening of the cartilaginous rings, and excessive dynamic airway collapse, defined
by an exaggerated forward displacement of the posterior tracheal membrane [30].
This nomenclature suffers from limited consistency throughout the literature; hence,
the inclusive term of LAC will be used hereafter.
It is estimated that LAC affects about one out of ten patients undergoing bron-
choscopy for pulmonary complaints and as many as a third of patients with COPD
or severe asthma [30, 84, 85]. However, the true prevalence of this condition remains
difficult to ascertain due to heterogeneous populations and the diagnostic methods
of the available studies. LAC may go underrecognized because of associated respi-
ratory diseases with overlapping symptoms, such as COPD, asthma, and bronchiec-
tasis, which are regarded not only to mimic but even predispose to LAC [86, 87].
Other risk factors include prolonged intubation and longstanding extrinsic airway
compression [87, 88]. Interestingly, although the degree of airways collapse appears
to relate with age (in healthy male volunteers), a higher risk of developing LAC in
the elderly has not been proven so far [29].
7 Airway Diseases in Geriatric Patients 161
a b
Fig. 7.3 Axial unenhanced CT images of a 76-year-old man show (a) increased ratio of coronal-
to-sagittal tracheal diameters (i.e., lunate trachea) and (b) excessive collapse of the posterior tra-
cheal wall (arrow) on expiration, highly suggestive of tracheomalacia. Advanced destructive
emphysema and bronchial wall thickening, consistent with a COPD diagnosis, can also be
appreciated
162 M. Balbi et al.
7.4.4 Broncholithiasis
a b
Fig. 7.4 Axial (a) and coronal (b) unenhanced CT images of a 70-year-old man presented with
hemoptysis show a calcified nodule within a right upper lobe bronchus (empty circle), consistent
with broncholithiasis
within the bronchi [103]), and rarer conditions such as tracheobronchial amyloido-
sis and tracheobronchopathia osteochondroplastica (see above) [102].
Surgical management is normally reserved for cases complicated by airway dis-
tortion, fistula formation, and massive hemoptysis, whereas a conservative approach
seems to be an appropriate option for asymptomatic or minimally symptomatic sub-
jects [97].
Tracheal diverticula (TD) are outpouchings from the tracheal wall [104]. With the
advent of CT and its continuous technical implementations (i.e., improvements in
spatial resolution of multidetector CT and use of thin slices), the prevalence of such
anatomical abnormality has been progressively increasing, ranging from 2% to 8%
[105]. A gender predominance has not been demonstrated [104].
TD tend to involve the right posterolateral wall of the trachea at the level of T1–
T3 vertebral bodies, an unprotected space compared to the contralateral side. TD are
classified into congenital and acquired, with the latter normally encountered in the
elderly population. Acquired TD are thought to be secondary to increased
164 M. Balbi et al.
intraluminal pressure that would cause out-bulging through a weak part of the tra-
cheal wall or cystic distension of the mucous gland ducts [106, 107].
Although TD are usually asymptomatic, in some cases, they can be responsible
of recurrent respiratory tract infections, acting as a reservoir for secretions and
hemoptysis [106]. Larger diverticula (>30mm) have been reported to cause also
painful neck swelling, cervical abscess, dysphagia, cough—secondary to compres-
sion of the vagus—and dysphonia due to compression of the recurrent laryngeal
nerve [108].
CT generally shows small air bubbles connected to the tracheal lumen and sur-
rounded by a very thin wall (Fig. 7.5). The communication with the trachea, how-
ever, is clearly appreciated in half of cases only, and usually with larger diverticula.
These diverticula might also have a thicker wall and be multiloculated. It is worth
emphasizing that when there is very little air within the diverticulum, it appears as
a solid structure, often indistinguishable from a lymph node [104].
main bronchi is observed in most cases. Other CT features include collapse, con-
solidation, and mass lesions [113, 114].
7.5 Conclusions
Despite growing knowledge of cellular and molecular aging mechanisms, the bor-
derland between normal and abnormal airway imaging in the elderly is still difficult
to define. Distinguishing physiological age-related changes from a disease state
remains quite challenging in a non-negligible proportion of the geriatric population.
However, the role of imaging in the detection and stratification of airway diseases
has been well-established, with chest HRCT being increasingly employed in clini-
cal practice to support the management of patients suffering from airway disorders,
including the elderlies.
References
1. Rojas M, Mora AL, Kapetanaki M, Weathington N, Gladwin M, Eickelberg O. Aging and
lung disease. Clinical impact and cellular and molecular pathways. Ann Am Thorac Soc.
2015;12(12):S222–7.
2. Zank DC, Bueno M, Mora AL, Rojas M. Idiopathic pulmonary fibrosis: aging, mitochondrial
dysfunction, and cellular bioenergetics. Front Med (Lausanne). 2018;5:10.
3. Brandsma CA, de Vries M, Costa R, Woldhuis RR, Konigshoff M, Timens W. Lung
ageing and COPD: is there a role for ageing in abnormal tissue repair? Eur Respir Rev.
2017;26(146):170073.
4. Sharma G, Goodwin J. Effect of aging on respiratory system physiology and immunology.
Clin Interv Aging. 2006;1(3):253–60.
5. Bowdish DME. The aging lung: is lung health good health for older adults? Chest.
2019;155(2):391–400.
6. Schneider JL, Rowe JH, Garcia-de-Alba C, Kim CF, Sharpe AH, Haigis MC. The aging lung:
physiology, disease, and immunity. Cell. 2021;184(8):1990–2019.
7. Brandenberger C, Muhlfeld C. Mechanisms of lung aging. Cell Tissue Res.
2017;367(3):469–80.
8. Yoon YS, Jin M, Sin DD. Accelerated lung aging and chronic obstructive pulmonary disease.
Expert Rev Respir Med. 2019;13(4):369–80.
9. Budinger GRS, Kohanski RA, Gan W, Kobor MS, Amaral LA, Armanios M, et al. The inter-
section of aging biology and the pathobiology of lung diseases: a joint NHLBI/NIA work-
shop. J Gerontol A Biol Sci Med Sci. 2017;72(11):1492–500.
10. Weibel ERGD. Architecture of the human lung. Use of quantitative methods estab-
lishes fundamental relations between size and number of lung structures. Science.
1962;137(3530):577–85.
11. Garcia-Rio F, Dorgham A, Pino JM, Villasante C, Garcia-Quero C, Alvarez-Sala R. Lung
volume reference values for women and men 65 to 85 years of age. Am J Respir Crit Care
Med. 2009;180(11):1083–91.
12. Hansell DM. Thin-section CT of the lungs: the Hinterland of normal. Radiology.
2010;256(3):695–711.
13. Levin DC, Parker L, Rao VM. Recent trends in imaging use in hospital settings: implications
for future planning. J Am Coll Radiol. 2017;14(3):331–6.
166 M. Balbi et al.
14. Shepard JO, Flores EJ, Abbott GF. Imaging of the trachea. Ann Cardiothorac Surg.
2018;7(2):197–209.
15. Kuhlman JE, Collins J, Brooks GN, Yandow DR, Broderick LS. Dual-energy subtraction chest
radiography: what to look for beyond calcified nodules. Radiographics. 2006;26(1):79–92.
16. Chou SH, Kicska GA, Pipavath SN, Reddy GP. Digital tomosynthesis of the chest: current
and emerging applications. Radiographics. 2014;34(2):359–72.
17. Sone S, Kasuga T, Sakai F, Oguchi K, Itoh A, Li F, et al. Digital tomosynthesis imaging of the
lung. Radiat Med. 1996;14(2):53–63.
18. Kamimura R, Takashima T. Clinical application of single dual-energy subtraction technique
with digital storage-phosphor radiography. J Digit Imaging. 1995;8(1 Suppl 1):21–4.
19. Kazerooni EA. High-resolution CT of the lungs. AJR Am J Roentgenol. 2001;177(3):501–19.
20. ACR–STR practice parameter for the performance of high-resolution computed tomography
(HRCT) of the lungs in adults. 2020. www.acr.org.
21. Gossner J, Nau R. Geriatric chest imaging: when and how to image the elderly lung, age-
related changes, and common pathologies. Radiol Res Pract. 2013;2013:584793.
22. Bankier AA, Fleischmann D, Mallek R, Windisch A, Winkelbauer FW, Kontrus M, et al.
Bronchial wall thickness: appropriate window settings for thin-section CT and radiologic-
anatomic correlation. Radiology. 1996;199(3):831–6.
23. Kubo T. Vendor free basics of radiation dose reduction techniques for CT. Eur J Radiol.
2019;110:14–21.
24. Ohno Y, Koyama H, Seki S, Kishida Y, Yoshikawa T. Radiation dose reduction techniques for
chest CT: principles and clinical results. Eur J Radiol. 2019;111:93–103.
25. Gibellino F, Osmanliev DP, Watson A, Pride NB. Increase in tracheal size with age.
Implications for maximal expiratory flow. Am Rev Respir Dis. 1985;132(4):784–7.
26. Breatnach E, Abbott GC, Fraser RG. Dimensions of the normal human trachea. AJR Am J
Roentgenol. 1984;142(5):903–6.
27. Collins DV, Cutillo AG, Armstrong JD, Crapo RO, Kanner RE, Tocino I, et al. Large airway
size, lung size, and maximal expiratory flow in healthy nonsmokers. Am Rev Respir Dis.
1986;134(5):951–5.
28. Sakai H, Nakano Y, Muro S, Hirai T, Takubo Y, Oku Y, et al. Age-related changes in the tra-
chea in healthy adults. Adv Exp Med Biol. 2010;662:115–20.
29. O’Donnell CR, Litmanovich D, Loring SH, Boiselle PM. Age and sex dependence of forced
expiratory central airway collapse in healthy volunteers. Chest. 2012;142(1):168–74.
30. Mitropoulos A, Song WJ, Almaghlouth F, Kemp S, Polkey M, Hull JH. Detection and diag-
nosis of large airway collapse: a systematic review. ERJ Open Res. 2021;7(3).
31. Edge JR, Millard FJ, Reid L, Simon G. The radiographic appearances of the chest in persons
of advanced age. Br J Radiol. 1964;37:769–74.
32. Lloyd DC, Taylor PM. Calcification of the intrathoracic trachea demonstrated by computed
tomography. Br J Radiol. 1990;63(745):31–2.
33. Moncada RM, Venta LA, Venta ER, Fareed J, Walenga JM, Messmore HL. Tracheal
and bronchial cartilaginous rings: warfarin sodium-induced calcification. Radiology.
1992;184(2):437–9.
34. Webb EM, Elicker BM, Webb WR. Using CT to diagnose nonneoplastic tracheal abnormali-
ties: appearance of the tracheal wall. AJR Am J Roentgenol. 2000;174(5):1315–21.
35. Matsuoka S, Uchiyama K, Shima H, Ueno N, Oish S, Nojiri Y. Bronchoarterial ratio and
bronchial wall thickness on high-resolution CT in asymptomatic subjects: correlation with
age and smoking. AJR Am J Roentgenol. 2003;180(2):513–8.
36. Copley SJ, Wells AU, Hawtin KE, Gibson DJ, Hodson JM, Jacques AE, et al. Lung morphol-
ogy in the elderly: comparative CT study of subjects over 75 years old versus those under 55
years old. Radiology. 2009;251(2):566–73.
37. Telenga ED, Oudkerk M, van Ooijen PM, Vliegenthart R, Ten Hacken NH, Postma DS, et al.
Airway wall thickness on HRCT scans decreases with age and increases with smoking. BMC
Pulm Med. 2017;17(1):27.
7 Airway Diseases in Geriatric Patients 167
38. Reinhardt JM, D’Souza ND, Hoffman EA. Accurate measurement of intrathoracic airways.
IEEE Trans Med Imaging. 1997;16(6):820–7.
39. Zach JA, Newell JD Jr, Schroeder J, Murphy JR, Curran-Everett D, Hoffman EA, et al.
Quantitative computed tomography of the lungs and airways in healthy nonsmoking adults.
Invest Radiol. 2012;47(10):596–602.
40. Castaner E, Gallardo X, Pallardo Y, Branera J, Cabezuelo MA, Mata JM. Diseases affect-
ing the peribronchovascular interstitium: CT findings and pathologic correlation. Curr Probl
Diagn Radiol. 2005;34(2):63–75.
41. Silva M, Milanese G, Seletti V, Ariani A, Sverzellati N. Pulmonary quantitative CT imag-
ing in focal and diffuse disease: current research and clinical applications. Br J Radiol.
2018;91(1083):20170644.
42. Hansell DAP, Lynch DA, et al. Imaging of diseases of the chest. Amsterdam: Elsevier; 2005.
p. 143–81.
43. Lynch DA, Newell JD, Tschomper BA, Cink TM, Newman LS, Bethel R. Uncomplicated
asthma in adults: comparison of CT appearance of the lungs in asthmatic and healthy sub-
jects. Radiology. 1993;188(3):829–33.
44. Kim JS, Muller NL, Park CS, Lynch DA, Newman LS, Grenier P, et al. Bronchoarterial ratio
on thin section CT: comparison between high altitude and sea level. J Comput Assist Tomogr.
1997;21(2):306–11.
45. Diaz AA, Young TP, Maselli DJ, Martinez CH, Maclean ES, Yen A, et al. Bronchoarterial
ratio in never-smokers adults: implications for bronchial dilation definition. Respirology.
2017;22(1):108–13.
46. Mohamed Hoesein FA, de Jong PA. Air trapping on computed tomography: regional versus
diffuse. Eur Respir J. 2017;49(1).
47. Hansell DM, Bankier AA, MacMahon H, McLoud TC, Muller NL, Remy J. Fleischner
Society: glossary of terms for thoracic imaging. Radiology. 2008;246(3):697–722.
48. Kligerman SJ, Henry T, Lin CT, Franks TJ, Galvin JR. Mosaic attenuation: etiology, methods
of differentiation, and pitfalls. Radiographics. 2015;35(5):1360–80.
49. Mastora I, Remy-Jardin M, Sobaszek A, Boulenguez C, Remy J, Edme JL. Thin-section CT
finding in 250 volunteers: assessment of the relationship of CT findings with smoking history
and pulmonary function test results. Radiology. 2001;218(3):695–702.
50. Tanaka N, Matsumoto T, Miura G, Emoto T, Matsunaga N, Ueda K, et al. Air trapping at
CT: high prevalence in asymptomatic subjects with normal pulmonary function. Radiology.
2003;227(3):776–85.
51. Lee KW, Chung SY, Yang I, Lee Y, Ko EY, Park MJ. Correlation of aging and smoking
with air trapping at thin-section CT of the lung in asymptomatic subjects. Radiology.
2000;214(3):831–6.
52. Martinez CH, Diaz AA, Meldrum C, Curtis JL, Cooper CB, Pirozzi C, et al. Age and
small airway imaging abnormalities in subjects with and without airflow obstruction in
SPIROMICS. Am J Respir Crit Care Med. 2017;195(4):464–72.
53. Verleden SE, Kirby M, Everaerts S, Vanstapel A, McDonough JE, Verbeken EK, et al. Small
airway loss in the physiologically ageing lung: a cross-sectional study in unused donor lungs.
Lancet Respir Med. 2021;9(2):167–74.
54. Well DS, Meier JM, Mahne A, Houseni M, Hernandez-Pampaloni M, Mong A, et al.
Detection of age-related changes in thoracic structure and function by computed tomog-
raphy, magnetic resonance imaging, and positron emission tomography. Semin Nucl Med.
2007;37(2):103–19.
55. Soejima K, Yamaguchi K, Kohda E, Takeshita K, Ito Y, Mastubara H, et al. Longitudinal
follow-up study of smoking-induced lung density changes by high-resolution computed
tomography. Am J Respir Crit Care Med. 2000;161(4 Pt 1):1264–73.
56. Rosenblum LJ, Mauceri RA, Wellenstein DE, Thomas FD, Bassano DA, Raasch BN, et al.
Density patterns in the normal lung as determined by computed tomography. Radiology.
1980;137(2):409–16.
168 M. Balbi et al.
57. Copley SJ, Giannarou S, Schmid VJ, Hansell DM, Wells AU, Yang GZ. Effect of aging on
lung structure in vivo: assessment with densitometric and fractal analysis of high-resolution
computed tomography data. J Thorac Imaging. 2012;27(6):366–71.
58. Marsh S, Aldington S, Williams MV, Nowitz MR, Kingzett-Taylor A, Weatherall M,
et al. Utility of lung density measurements in the diagnosis of emphysema. Respir Med.
2007;101(7):1512–20.
59. Yang T, Chen C, Chen Z. The CT pulmonary vascular parameters and disease severity in
COPD patients on acute exacerbation: a correlation analysis. BMC Pulm Med. 2021;21(1):34.
60. Miravitlles M, Calle M, Soler-Cataluna JJ. Clinical phenotypes of COPD: identification, defi-
nition and implications for guidelines. Arch Bronconeumol. 2012;48(3):86–98.
61. Mitzner W. Emphysema—a disease of small airways or lung parenchyma? N Engl J Med.
2011;365(17):1637–9.
62. Janssens JP, Pache JC, Nicod LP. Physiological changes in respiratory function associated
with ageing. Eur Respir J. 1999;13(1):197–205.
63. Verbeken EK, Cauberghs M, Mertens I, Clement J, Lauweryns JM, Van de Woestijne KP. The
senile lung. Comparison with normal and emphysematous lungs. 2. Functional aspects.
Chest. 1992;101(3):800–9.
64. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, et al. Global strategy for the
diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD
executive summary. Am J Respir Crit Care Med. 2007;176(6):532–55.
65. Gevenois PA, De Vuyst P, Sy M, Scillia P, Chaminade L, de Maertelaer V, et al. Pulmonary
emphysema: quantitative CT during expiration. Radiology. 1996;199(3):825–9.
66. Cavigli E, Camiciottoli G, Diciotti S, Orlandi I, Spinelli C, Meoni E, et al. Whole-lung densi-
tometry versus visual assessment of emphysema. Eur Radiol. 2009;19(7):1686–92.
67. Park J, Hobbs BD, Crapo JD, Make BJ, Regan EA, Humphries S, et al. Subtyping COPD by
using visual and quantitative CT imaging features. Chest. 2020;157(1):47–60.
68. Occhipinti M, Paoletti M, Bartholmai BJ, Rajagopalan S, Karwoski RA, Nardi C, et al.
Spirometric assessment of emphysema presence and severity as measured by quantitative CT
and CT-based radiomics in COPD. Respir Res. 2019;20(1):101.
69. Gierada DS, Guniganti P, Newman BJ, Dransfield MT, Kvale PA, Lynch DA, et al.
Quantitative CT assessment of emphysema and airways in relation to lung cancer risk.
Radiology. 2011;261(3):950–9.
70. Lynch DA, Austin JH, Hogg JC, Grenier PA, Kauczor HU, Bankier AA, et al. CT-definable
subtypes of chronic obstructive pulmonary disease: a statement of the Fleischner Society.
Radiology. 2015;277(1):192–205.
71. Araki T, Nishino M, Gao W, Dupuis J, Putman RK, Washko GR, et al. Pulmonary cysts
identified on chest CT: are they part of aging change or of clinical significance? Thorax.
2015;70(12):1156–62.
72. Polverino E, Goeminne PC, McDonnell MJ, Aliberti S, Marshall SE, Loebinger MR, et al.
European Respiratory Society guidelines for the management of adult bronchiectasis. Eur
Respir J. 2017;50(3).
73. Heidinger BH, Occhipinti M, Eisenberg RL, Bankier AA. Imaging of large airways disorders.
AJR Am J Roentgenol. 2015;205(1):41–56.
74. Aliberti S, Lonni S, Dore S, McDonnell MJ, Goeminne PC, Dimakou K, et al. Clinical phe-
notypes in adult patients with bronchiectasis. Eur Respir J. 2016;47(4):1113–22.
75. Winter DH, Manzini M, Salge JM, Busse A, Jaluul O, Jacob Filho W, et al. Aging of the
lungs in asymptomatic lifelong nonsmokers: findings on HRCT. Lung. 2015;193(2):283–90.
76. Cartier Y, Kavanagh PV, Johkoh T, Mason AC, Muller NL. Bronchiectasis: accuracy of
high-resolution CT in the differentiation of specific diseases. AJR Am J Roentgenol.
1999;173(1):47–52.
77. Habesoglu MA, Ugurlu AO, Eyuboglu FO. Clinical, radiologic, and functional evaluation of
304 patients with bronchiectasis. Ann Thorac Med. 2011;6(3):131–6.
78. McGuinness G, Naidich DP. CT of airways disease and bronchiectasis. Radiol Clin North
Am. 2002;40(1):1–19.
7 Airway Diseases in Geriatric Patients 169
79. Naidich DP, McCauley DI, Khouri NF, Stitik FP, Siegelman SS. Computed tomography of
bronchiectasis. J Comput Assist Tomogr. 1982;6(3):437–44.
80. Ooi GC, Khong PL, Chan-Yeung M, Ho JC, Chan PK, Lee JC, et al. High-resolution
CT quantification of bronchiectasis: clinical and functional correlation. Radiology.
2002;225(3):663–72.
81. Reiff DB, Wells AU, Carr DH, Cole PJ, Hansell DM. CT findings in bronchiectasis: lim-
ited value in distinguishing between idiopathic and specific types. AJR Am J Roentgenol.
1995;165(2):261–7.
82. Singh A, Bhalla AS, Jana M. Bronchiectasis revisited: imaging-based pattern approach to
diagnosis. Curr Probl Diagn Radiol. 2019;48(1):53–60.
83. Reich JM, Johnson RE. Mycobacterium avium complex pulmonary disease presenting
as an isolated lingular or middle lobe pattern. The Lady Windermere syndrome. Chest.
1992;101(6):1605–9.
84. Leong P, Tran A, Rangaswamy J, Ruane LE, Fernando MW, MacDonald MI, et al. Expiratory
central airway collapse in stable COPD and during exacerbations. Respir Res. 2017;18(1):163.
85. Dal Negro RW, Tognella S, Guerriero M, Micheletto C. Prevalence of tracheobronchomalacia
and excessive dynamic airway collapse in bronchial asthma of different severity. Multidiscip
Respir Med. 2013;8(1):32.
86. Nuutinen J. Acquired tracheobronchomalacia. Eur J Respir Dis. 1982;63(5):380–7.
87. Ridge CA, O’Donnell CR, Lee EY, Majid A, Boiselle PM. Tracheobronchomalacia: current
concepts and controversies. J Thorac Imaging. 2011;26(4):278–89.
88. Feist JH, Johnson TH, Wilson RJ. Acquired tracheomalacia: etiology and differential diagno-
sis. Chest. 1975;68(3):340–5.
89. Rayl JE. Tracheobronchial collapse during cough. Radiology. 1965;85:87–92.
90. Boiselle PM, Michaud G, Roberts DH, Loring SH, Womble HM, Millett ME, et al. Dynamic
expiratory tracheal collapse in COPD: correlation with clinical and physiologic parameters.
Chest. 2012;142(6):1539–44.
91. Lomasney L, Bergin CJ, Lomasney J, Roggli V, Foster W. CT appearance of lunate trachea. J
Comput Assist Tomogr. 1989;13(3):520–2.
92. Boiselle PM, Feller-Kopman D, Ashiku S, Weeks D, Ernst A. Tracheobronchomalacia: evolv-
ing role of dynamic multislice helical CT. Radiol Clin North Am. 2003;41(3):627–36.
93. Boiselle PM, Ernst A. Tracheal morphology in patients with tracheomalacia: prevalence of
inspiratory lunate and expiratory “frown” shapes. J Thorac Imaging. 2006;21(3):190–6.
94. Leong P, Bardin PG, Lau KK. What’s in a name? Expiratory tracheal narrowing in adults
explained. Clin Radiol. 2013;68(12):1268–75.
95. Baroni RH, Feller-Kopman D, Nishino M, Hatabu H, Loring SH, Ernst A, et al.
Tracheobronchomalacia: comparison between end-expiratory and dynamic expiratory CT
for evaluation of central airway collapse. Radiology. 2005;235(2):635–41.
96. Wright CD. Tracheomalacia. Chest Surg Clin N Am. 2003;13(2):349–57, viii.
97. Alshabani K, Ghosh S, Arrossi AV, Mehta AC. Broncholithiasis: a review. Chest.
2019;156(3):445–55.
98. Jin YX, Jiang GN, Jiang L, Ding JA. Diagnosis and treatment evaluation of 48 cases of bron-
cholithiasis. Thorac Cardiovasc Surg. 2016;64(5):450–5.
99. Cerfolio RJ, Bryant AS, Maniscalco L. Rigid bronchoscopy and surgical resection for
broncholithiasis and calcified mediastinal lymph nodes. J Thorac Cardiovasc Surg.
2008;136(1):186–90.
100. Vix VA. Radiographic manifestations of broncholithiasis. Radiology. 1978;128(2):295–9.
101. Lim SY, Lee KJ, Jeon K, Koh WJ, Suh GY, Chung MP, et al. Classification of broncholiths
and clinical outcomes. Respirology. 2013;18(4):637–42.
102. Seo JB, Song KS, Lee JS, Goo JM, Kim HY, Song JW, et al. Broncholithiasis: review of the
causes with radiologic-pathologic correlation. Radiographics. 2002;22 Spec No:S199–213.
103. Ahn JM, Im JG, Seo JW, Han HS, Yoon HK, Kim WS, et al. Endobronchial hamartoma: CT
findings in three patients. AJR Am J Roentgenol. 1994;163(1):49–50.
104. Gayer G. Tracheal diverticula. Semin Ultrasound CT MR. 2016;37(3):190–5.
170 M. Balbi et al.
105. Polat AV, Elmali M, Aydin R, Ozbay A, Celenk C, Murat N. Paratracheal air cysts: prevalence
and correlation with lung diseases using multi-detector CT. J Med Imaging Radiat Oncol.
2014;58(2):144–8.
106. Shah M, Joshi JM. Tracheal diverticulum. Indian J Chest Dis Allied Sci. 2012;54(1):39–40.
107. Mackinnon D. Tracheal diverticula. J Pathol Bacteriol. 1953;65(2):513–7.
108. Caversaccio MD, Becker M, Zbaren P. Tracheal diverticulum presenting with recurrent laryn-
geal nerve paralysis. Ann Otol Rhinol Laryngol. 1998;107(4):362–4.
109. Gupta A, Shah A. Bronchial anthracofibrosis: an emerging pulmonary disease due to biomass
fuel exposure. Int J Tuberc Lung Dis. 2011;15(5):602–12.
110. Pilaniya V, Kunal S, Shah A. Occurrence of bronchial anthracofibrosis in respiratory sympto-
matics with exposure to biomass fuel smoke. Adv Respir Med. 2017;85(3):127–35.
111. Kim YJ, Jung CY, Shin HW, Lee BK. Biomass smoke induced bronchial anthracofibrosis:
presenting features and clinical course. Respir Med. 2009;103(5):757–65.
112. Shah A, Kunal S, Gothi R. Bronchial anthracofibrosis: the spectrum of radiological appear-
ances. Indian J Radiol Imaging. 2018;28(3):333–41.
113. Kahkouee S, Pourghorban R, Bitarafan M, Najafizadeh K, Makki SS. Imaging findings of
isolated bronchial anthracofibrosis: a computed tomography analysis of patients with bron-
choscopic and histologic confirmation. Arch Bronconeumol. 2015;51(7):322–7.
114. Kim HY, Im JG, Goo JM, Kim JY, Han SK, Lee JK, et al. Bronchial anthracofibrosis
(inflammatory bronchial stenosis with anthracotic pigmentation): CT findings. AJR Am J
Roentgenol. 2000;174(2):523–7.
Neoplastic Diseases of the Respiratory
System in Geriatric Patients 8
Zeno Falaschi, Francesco Filippone, Sergio Pansini,
Stefano Tricca, Paola Basile, Sara Cesano,
and Alessandro Carriero
8.1 Epidemiology
Lung cancer is the most common cancer in the world and the leading cause of
cancer-related deaths in both Western countries and the United States, with the
fastest-growing morbidity and mortality, especially in the elderly [1].
According to the latest statistics updated to 2020 of the American Cancer Society,
there will be 235,760 estimated new cases of lung cancer in 2021, of which 119,100
cases in male and 116,660 in females; on the other hand, the age-adjusted death rate
for lung cancer is higher for men (46.7 per 100,000 persons) than for women (31.9
per 100,000 persons). This means that more men are diagnosed with lung cancer
each year, but more women live with the disease. In addition, these data also con-
firm that lung cancer is mostly a disease of the elderly, with a growing incidence
from 0.6% (in the age range from 50 to 59 years), to 5.4% (from 70 years and
older) [2].
Lung cancer refers to a histologically and clinically diverse group of malignan-
cies arising in the respiratory tract, primarily but not exclusively in cells lining the
airways of the lung. The four principal types, classified by light microscopy and
special stains, are non-small-cell lung cancer (NSCLC), which is the most common
type and it constitutes between 80% and 85% of all lung cancers, adenocarcinoma,
small-cell carcinoma (SCLC), and squamous cell carcinoma. Unfortunately, at the
time of diagnosis, the majority of patients already have metastatic disease, and a
systemic, palliative treatment is the primary therapeutic option. More than 50% of
advanced NSCLCs are diagnosed in patients older than age 65 years [3].
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 171
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_8
172 Z. Falaschi et al.
The rising incidence of lung cancer through the first half of the twentieth century
prompted intensive epidemiologic investigations of the disease, resulting in the
identification of a number of causal agents. Cigarette smoking is by far the largest
cause of lung cancer, and the worldwide epidemic of lung cancer is largely attribut-
able to smoking. However, occupational exposures, including asbestos, uranium,
and coke (an important fuel in the manufacture of iron in smelters, blast furnaces,
and foundries), have placed a number of worker groups at high risk, and some of
these occupational agents are synergistic with smoking in increasing lung cancer
risks [7].
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 173
In fact, nonsmoking asbestos workers are five times more likely to develop lung
cancer than nonsmokers not exposed to asbestos; if they also smoke, the risk factor
jumps to 50 or higher [9].
Furthermore there is some evidence that both indoor and outdoor air pollution
also increases lung cancer risks, specifically exposure to radon is estimated to be the
second-leading cause of lung cancer, accounting for an estimated 21,000 lung can-
cer deaths each year (range of 8000–45,000). Radon is a tasteless, colorless, and
odorless gas that is produced by decaying uranium and occurs naturally in soil and
rock. The majority of these deaths occur among smokers since there is a greater risk
for lung cancer when smokers also are exposed to radon [7].
In addition, observational evidence showing a familial aggregation of lung can-
cer has suggested that genetic factors also may determine risks in smokers, but the
specific genes remain under active investigation [8].
who have never smoked, even after 15–20 years of not smoking. Extensive data
convincingly show how smoking cessation lowers lung cancer risks: using data
from a 1990 case-control study, [13] estimated cumulative lung cancer risks for
persons up to 75 years of age. The estimated lifetime risk of lung cancer deaths
for men who continue to smoke, absent death from another cause, was 16%.
Substantial reductions in this risk can be achieved by cessation at younger ages;
even cessation at 60 years of age lowered the cumulative risk from 16% to about
10% [14].
Since the first research reports linking smoking to lung cancer and other dis-
eases, the tobacco industry has continually changed the characteristics of the ciga-
rette. These changes have included the addition of filter tips, perforation of the filter
tips, use of reconstituted tobacco, and changes in the paper and in additives.
Nevertheless, even though during the last 50 years characteristics of cigarettes have
changed and yields of tar and nicotine have declined substantially, as assessed by
the Federal Trade Commission’s test protocol, the risk of lung cancer in smokers
has not declined and the benefits are minimal in comparison with giving up ciga-
rettes entirely [15].
The single most effective way to reduce hazards of smoking continues to be that
of quitting entirely and the general pattern of this decline is the same for men and
women, for smokers of filter-tipped and unfiltered cigarettes, and for all major his-
tologic types of lung cancer [15].
In conclusion, despite the gains in understanding respiratory carcinogenesis and
the potential of molecular and imaging techniques to screen for lung cancer, smok-
ing prevention and cessation remain the fundamental strategies for controlling the
lung cancer epidemic.
One of the biggest problems in lung cancer diagnosis is that the symptoms are
shared with many benign pulmonary diseases and that they actually are more com-
mon in these pathologies.
As a matter of fact, lung cancer is usually asymptomatic in the early stages and
this often leads to a delay in the diagnosis, that occurs when the tumor is already in
an advanced stage.
Therefore, its symptoms are present only in a minority of the patients, are non-
specific, and appear only when the tumor has already spread.
Clinical presentation is heterogeneous, and it is caused by the local tumor growth,
the intrathoracic spread, and the distant spreading. It also depends on the tumor type
and its specific location and behavior. In fact, small-cell lung cancer (SCLC) gener-
ally arises in the central part of the lung, penetrates into the mediastinum, and con-
sequently is more commonly associated with invasive symptoms. On the contrary,
non-small-cell lung cancer (NSCLC) is usually located in the peripheral lung, and
it is characterized by less specific symptoms [9].
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 175
For what concerns local growth-related lung cancer symptoms, they mainly con-
sist of cough, dyspnea, wheeze, and hemoptysis.
Specifically, cough, which is caused by the obstruction of the airways, is the
most common symptom. It is present in almost 50% of the patients, and it is more
frequently associated with SCLC, since this tumor type is mainly located on larger
bronchi. Although cough is not specific for this pathology, with no doubt a new and
persistent cough in active or past smokers should raise concern for lung cancer. In
addition, recurrent pneumonia in the same anatomic region as well as frequent exac-
erbation of chronic obstructive pulmonary disease should be considered as alarm
signs [9].
On the other hand, hemoptysis is the most specific symptom, especially when
associated with others. This also has a higher positive predictive value compared to
the other symptoms. For this reason, every patient with hemoptysis should perform
a chest X-ray.
For what concerns symptoms related to the intrathoracic spread, these are caused
by the invasion of the tumor into the mediastinal structures and mainly consist of
chest pain, vocal cord paralysis, superior vena cava syndrome (SVC), and dysphagia.
In particular, SVC syndrome is caused by the obstruction of the vena cava by the
primary tumor or by enlarged lymph nodes or thrombus. It generally presents with
edema of the upper body.
Instead, vocal cord paralysis and dysphagia are caused by the invasion of the
recurrent laryngeal nerve and of the esophagus.
On the contrary, symptoms associated with the distant spreading of lung cancer
have mostly the same frequency in SCLC and NSCLC [10].
The most common metastatic sites are brain, liver, adrenal gland, bones, and
bone marrow. The central nervous system involvement frequently manifests with
headache, seizure, altered mental status; while bone metastases present with bone
pain, and liver metastases with anemia and weight loss.
In conclusion, lung cancer can also be associated with paraneoplastic syndromes
with SCLC as the most common cause [16]. Among these conditions, the most fre-
quent ones are syndrome of inappropriate antidiuresis, Cushing’s syndrome, and
hypercalcemia. In addition, SCLC can also be associated with neurologic syn-
dromes such as Lambert-Eaton and Limbic encephalitis.
The diagnosis of lung cancer can be done through chest X-ray, computed tomogra-
phy (CT) scans, magnetic resonance (MRI), positron emission tomography (PET),
cytology sputum, and breath analysis.
All the available detection techniques of lung cancer have different detection
levels and various markers. Nevertheless, it is often impossible to radiographically
distinguish between the several histological lung cancer types.
176 Z. Falaschi et al.
with intense contrast-enhancement. It has a slow growth so can reach very large
dimensions and can spread through the extra-bronchial space. Rarely it is
peripheric.
The adenocarcinoma may show different features based on the subtypes: the pre-
invasive in situ minimally invasive adenocarcinoma and the invasive lepidic
predominant-adenocarcinoma (formerly called bronchiolo-alveolar carcinoma,
BAC) often consist of ground glass nodule or a subsolid nodule with a predominant
ground glass component, while the remaining invasive subtypes of adenocarcinoma
usually show up as a solid or subsolid nodule.
The invasive mucinous adenocarcinoma subtype (also formerly mucinous BAC)
can have a variable appearance, including consolidation, air bronchograms, or mul-
tifocal subsolid nodules or masses [26].
Nuclear Medicine
FDG-PET/CT is essential for the lung cancer staging, since it can assess for the
nodal and distant metastatic disease.
Adenocarcinoma in situ, low-grade adenocarcinomas and minimally invasive
adenocarcinoma are commonly associated with PET false-negative results.
FDG PET/CT is recommended when assessing subsolid ground glass lung
lesions that have a solid component measuring more than 8 mm [20].
Chest X-ray has a fundamental role in the diagnosis of many pulmonary diseases.
Due to its low cost, availability, and low radiation, it is usually the first exam per-
formed in order to diagnose lung pathologies. It is the first-line investigation in
patients with suspected lung cancer.
Lung cancer diagnosis may occur because of an incidental finding in an asymp-
tomatic patient that performs the exam for other reasons, in a symptomatic patient
or as an unexpected evolution from pneumonia, atelectasis, or pleural effusion.
Chest X-ray is often the first performed exam, but it is not the most sensitive
imaging technique. CT scan is considered the preferred exam to have a proper diag-
nosis and staging of the disease.
In fact chest X-ray sensitivity of lung nodule detection when it is <6 mm is very
low, but it increases when the nodule is calcified. The sensitivity is about 50% when
the nodule diameter is 6–10 mm [27].
Having an early diagnosis of lung cancer is associated with an improved sur-
vival; therefore, multiple studies have been performed to investigate the role of
chest X-ray in screening. Unfortunately, no study has demonstrated a reduction in
mortality connected to this exam. It has been shown that this technique fails to at
least initially detect lung cancer in more than 20% of patients.
178 Z. Falaschi et al.
The missed diagnosis may be related to “observer error,” to poor technique qual-
ity, but also to the dimension of the tumor. In fact, if it is less than 1 cm it can be
easily not detected. In addition, the location of the tumor may play a role, especially
when this is in the upper lobes where it can be masked by anatomical structures such
as ribs and vessels.
For these reasons, CT scan is the preferred technique to diagnose this disease [20].
Lung cancer may present in different ways on chest X-ray: as a nodule, a mass, an
enlarged mediastinum, atelectasis, and pleural effusion.
Based on the cellular type of tumor, we can have different radiological character-
istics. However the histological subtype cannot be correctly identified solely on the
basis of chest X-ray, and a confirmative biopsy is always needed.
8.2.2.1 Adenocarcinoma
Adenocarcinoma is 31% of all lung cancers. It is usually peripherally located and
measures less than 4 cm. It is associated with detection of hila and/or mediastinal
enlargement on chest X-ray in 51% of cases.
Pulmonary nodules are a very frequent finding on chest X-ray and their prevalence
ranges from 0.09% to 0.2%.
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 179
8.2.3.2 Margins
The margin characteristics may give an indication of a nodule’s nature. Regular and
smooth margins are usually considered as suggestive of benign disease but in some
cases may be present also in malignant nodules. Irregular, ill-defined, lobulated and
spiculated margins are indicative of malignancy [21].
Fig. 8.1 Two examples of large pulmonary masses in the inferior right lobe in P-A and L-L
projections
The lesion number can help us, in fact metastases are solitary only in a quarter of
cases. Metastases also have sharp margins and do not contain cavitation.
A bronchogenic cyst may also be mistaken for lung cancer. This congenital
defect is rare, and its location depends on the timing. It can be located in the medi-
astinum or intrapulmonary. On chest X-ray, it presents as a rounded opacity with
smooth margins, usually in the lower lobes and can be filled with air or fluid.
8.2.4 Atelectasis
Lung cancer may also present as an area of atelectasis. This is defined as a loss of
lung volume caused by the reduction in the content of air in the bronchi and in the
alveolar spaces. Lungs that normally appear black become white when fluids or soft
tissue substitutes air. So normally on the chest X-ray atelectasis appears as an
opaque area. In the early phases this may not present as an opacity, but indirect signs
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 181
can be visible and be suggestive of its presence, for example, the reduction in
lung volume.
Therefore in addition to the increased lung opacity, there are other signs. The first
one is the shift of the interlobar fissures toward the area of atelectasis. When this is
in the right and left upper lobes, both fissures move superiorly, as shown in Fig. 8.2.
When it is in the middle lobe the oblique one moves upward and the horizontal one
downward. If atelectasis is in the inferior lobes the oblique fissure moves downward
and posteriorly.
A second sign is the upward displacement of the hemidiaphragm which is usu-
ally more evident when atelectasis involves the inferior lobes. In addition to this, the
mobile mediastinal structures move toward the affected site. In case of upper lobes
atelectasis, there is a displacement of trachea and the superior mediastinum. Trachea,
which is normally in midline location, in correspondence with the spinous pro-
cesses of the vertebral bodies, may shift toward the area of volume loss. When
atelectasis involves the inferior lobes the heart and the inferior mediastinum usually
move. When the heart shifts toward the left, there is an overlap between the right
heart border and the spine. When it shifts toward right, the left heart border is almost
in the midline.
The hila may also dislocate: superiorly if there is atelectasis of the upper lobes,
downward if the inferior ones are affected. Of course it is important to remember
that the left hilum in the majority of the population is located superior to the
right one.
Another sign of atelectasis is the compensatory over inflation of the unaffected
ipsilateral lobes or the contralateral lung. The bigger the atelectatic area the more
evident the over inflation is.
Of course there can be atelectasis of the whole lung and in this case the medias-
tinum is shifted toward the affected site so that the contralateral lung can cross the
midline.
Atelectasis needs to be differentiated from a non-atelectatic parenchymal con-
solidation. The absence of air bronchogram may help us in doing this. In fact this
does not occur in case of atelectasis because obstructed bronchi and bronchioles fill
with secretions and appear radiopaque on the X-ray.
Compressive Atelectasis
This type of atelectasis is caused by the passive compression of the lung that can be
caused by a large pleural effusion, a pneumothorax or a space-occupying lesion
such as a lung mass.
Obstructive Atelectasis
Obstructive atelectasis is caused by the absorption of air from the alveoli through
the capillary bed distal to an obstructive lesion of the bronchial tree. This leads to
the collapse of the affected segment or lobe, and since the pleurae remain in contact
with each other, there is a pull on the mobile structures of the thorax toward the area
of atelectasis. This can be caused by various etiologies such as bronchial carcinoma,
mucus, and foreign bodies.
There are also cicatrization and band atelectasis. The first occurs due to fibrotic
changes in the parenchyma caused by chronic inflammation that lead to lung vol-
ume reduction. The second presents as fine horizontal bands of atelectasis that
occurs when the patient has decreased diaphragm mobility.
Single-sided pleural effusion should always raise the suspect of lung cancer, as
shown in Fig. 8.3. This is usually caused by the invasion of the pleural space by
malignant cells, which may both cause a reactive increase in the reactive production
of fluid and impair its reuptake by the pleura. Pleural effusion can be defined as an
excessive accumulation of fluid in the space between the visceral and parietal pleura.
The pleural space normally contains about 2–5 mL of fluid. This accumulates when
the equilibrium between fluid formation and resorption is altered. This is the most
common pleural finding documented on X-ray.
When the patient is in the upright position pleural fluid accumulates in the base
of the thoracic cavity and we can only see it if it exceeds 250 mL. When the exam
is performed in supine position, the fluid collects posteriorly and can demonstrate
effusions as small as 15–20 mL. The whole hemithorax is opacified when 2 L of
fluid is collected.
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 183
Fig. 8.3 This is an example of a patient with lung cancer that presents with pleural effusion
In some cases fluid may accumulate between the inferior surface of the lung and
the hemidiaphragm and may simulate an elevation of the diaphragm.
Lung tumors should not be confused with pseudotumors which are also called
vanishing tumors. These are fluid collections either between the layers of an inter-
lobar pulmonary fissure or beneath it. Of course these disappear when the underly-
ing condition is treated.
There are four types of pleural effusion: exudative, transudative, chylous, and
hematic. The first two are the most common ones and differ for the protein content.
Transudate contains less than 3 g/100 mL and occurs due to an increase in hydro-
static pressure or a decrease in oncotic pressure.
Exudate has more than 3 g/100 mL and is caused by an increased capillary per-
meability. The most common cause of exudative pleural effusion is malignancy.
Other causes are infections, abdominal diseases, thromboembolism, and tho-
racic trauma.
The chylous effusion contains triglycerides or cholesterol and may occur for
example due to the rupture of a great lymphatic vessel, whereas the hematic one
may be caused by lung laceration or breaking of a vessel.
The most accurate technique nowadays in lung cancer detection and staging is CT
imaging thanks to its high spatial and contrast resolution. In particular, thanks to the
axial scan plane and the possibility of multiplanar reconstructions (MPR), it elimi-
nates the overlapping of the various structures, solving the greater limit of tradi-
tional chest radiography (CXR) [23].
184 Z. Falaschi et al.
a b c
Fig. 8.4 Nodule types: (a) Solid, (b) Part solid and (c) Pure ground glass (GGO)
Table 8.1 2017 Fleischner Society Guidelines for management of incidental solid nodularities
Nodule Risk
type factors <6 mm 6–8 mm >8 mm
Solitary Low No routine CT at 6–12 months Consider CT, CT-PET or
nodule risk follow-up (consider at tissue biopsy at 3 months
18–24 months)
Solitary High Consider CT at CT at 6–12 months and at Consider CT, CT-PET or
nodule risk 12 months 18–24 months tissue biopsy at 3 months
Multiple Low No routine CT at 3–6 months CT at 3–6 months
nodules risk follow-up (consider at (consider at
18–24 months) 18–24 months)
Multiple High Consider CT at CT at 3–6 months and at CT at 3–6 months and at
nodules risk 12 months 18–24 months 18–24 months
Table 8.2 2017 Fleischner Society Guidelines for management of incidental subsolid
nodularities
Nodule type <6 mm >6 mm
Solitary GGO No routine follow-up CT at 6–12 months. If stable, CT every
nodule 2 years for a total of 5 years
Solitary part No routine follow-up CT at 3–6 months. If stable, CT every year
solid nodule for 5 years
Multiple CT at 3–6 months. If stable, CT at 3–6 months. Subsequent management
nodules consider CT at 2 and 4 years based on the most suspicious nodule(s)
–– Growth or stable size: A solid nodule that has been stable for 2 years or more on
CT does not need any further investigations. In contrast, subsolid nodules could
be attributable to a low-grade adenocarcinoma, which has slower growth average
and an elevated VDT (volume doubling time), so it is considered likely to be
benign only when stable for 5 or more years by CT.
186 Z. Falaschi et al.
The 2004 World Health Organization (WHO) classification divides lung cancers
into two main histological categories: non-SCLC (NSCLC, 85% of all lung cancers)
and small-cell lung carcinoma (SCLC, 15% of all lung cancers) [26, 30, 31].
The distinction between the various subtypes of lung cancer can help to evaluate
the subsequent diagnostic and therapeutic process and can give the clinician infor-
mation about a patient’s prognosis.
However, although some morphological and metabolic characteristics may sug-
gest the histotype of the detected lung cancer, the diagnosis of certainty is still his-
tological and requires biopsy sampling.
SCC accounts for at least 20% of all bronchogenic cancers and is strongly
associated with cigarette smoking.
In two-thirds of cases SCC has central location with intraluminal obstruction
of the main, lobar, or segmental bronchus resulting in obstruction or atelectasis
of the downstream parenchyma. Centrally located tumors therefore manifest
themselves with symptoms such as chronic cough, superimposed infectious
pneumonia, or hemoptysis and are usually reachable by endobronchial examina-
tions. In the initial stages they therefore produce a thickening of the bronchial
wall, subsequently they can manifest themselves with hilar or peri-hilar masses
or they can be more subtle, manifesting mainly with indirect signs such as atel-
ectasis [28].
SCC can also have peripheral localization usually manifesting itself as a mass
with irregular borders. Peripheral cancer occurs later and is therefore diagnosed
when it is larger with possible involvement of the chest wall or when it is meta-
static. SCC is the most common histotype of Pancoast tumor, an example can be
seen in Fig. 8.5.
Another feature frequently associated with SCC is cavitation, both in the pri-
mary lesion and in metastases with irregular and thick margins. Figure 8.6 shows
an example of a big cavitated lesion, which has been proven to be a peripheral
SCC [30, 32–34].
–– Adenocarcinoma:
Histologically the term adenocarcinoma refers to an epithelial neoplasm with
glandular differentiation or intracytoplasmic mucin production.
Adenocarcinoma is the most common histologic type of lung cancer, account-
ing for nearly 40% of lung cancers. Furthermore, adenocarcinoma is also the
most common histologic group seen in women and nonsmokers, although there
is a minimal association with cigarette smoking.
In 2011, the International Association for the Study of Lung Cancer (IASLC),
American Thoracic Society (ATS), and European Respiratory Society (ERS)
introduced a new classification of adenocarcinoma, which is now divided into
four main categories: adenocarcinoma in situ (AIS), minimally invasive
a b
Fig. 8.5 (a) Axial CT scan of a 69-year-old man showing a Pancoast tumor in the right upper lobe.
(b) Coronal view of the same CT
188 Z. Falaschi et al.
a b
Fig. 8.6 (a) Axial CT scan of a 75-year-old man showing a cavitated mass in the right lower lobe.
Histological examination proved to be an SCC. (b) Coronal view of the same CT
a b
Fig. 8.8 (a) Axial CT scan of a 67-year-old man showing a peri-hilar mass in the right lower lobe.
Histological examination proved to be an SCLC. (b) The same CT after administration of contrast
medium that shows an indissociable mass from the bronchial branches and the vessels for the
lower lobe. The mass has a central necrotic component. Some enlarged lymph nodes in the medi-
astinal area may also be observed
T Component
The T component can be classified into five categories, as shown in Table 8.3. The
tumor’s greater dimension is the main variable when assessing this parameter,
although the invasion of nearby anatomical structure plays a fundamental role. In
particular, the invasion of mediastinal structures such as trachea, carina, or pericar-
dium immediately leads to an increment of the T component. The infiltration of
peripheral tissues such as the pleura or the chest wall also comports a T increment.
If more than one neoplastic nodule is present, the location of such nodule respec-
tively to the main mass determines T staging.
IASLC recommends that all tumors are measured in centimeters and to use the
lung window in order to determine the greatest dimension, since using the medias-
tinal window can lead to an understaging.
N Component
Table 8.4 summarizes the N classification in the eighth edition of the TNM staging
in lung cancer. Lymph nodes with a short axis >1 cm are to be considered
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 191
Table 8.3 T stage definition in the 8th edition of the TNM in lung cancer
T: Primary tumor
Tx Primary tumor cannot be assessed or tumor proven by presence of malignant cells in
sputum or bronchial washings but not visualized by imaging or bronchoscopy
T0 No evidence of primary tumor
Tis Carcinoma in situ
T1 Tumor ≤3 cm in greatest dimension surrounded by lung or visceral pleura without
bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in
the main bronchus)
T1a Minimally invasive adenocarcinoma
(mi)
T1a Tumor ≤1 cm in greatest dimension
T1b Tumor >1 cm but ≤2 cm in greatest dimension
T1c Tumor >2 cm but ≤3 cm in greatest dimension
T2 Tumor >3 cm but ≤5 cm or tumor with any of the following features:
– Involves main bronchus regardless of distance from the carina but without
involvement of the carina
– Invades visceral pleura
– Associated with atelectasis or obstructive pneumonitis that extends to the hilar
region, involving part or all of the lung
T2a Tumor >3 cm but ≤4 cm in greatest dimension
T2b Tumor >4 cm but ≤5 cm in greatest dimension
T3 Tumor >5 cm but ≤7 cm in greatest dimension or associated with separate tumor
nodule(s) in the same lobe as the primary tumor or directly invades any of the following
structures: Chest wall (including the parietal pleura and superior sulcus tumors), phrenic
nerve, parietal pericardium
T4 Tumor >7 cm in greatest dimension or associated with separate tumor nodule(s) in a
different ipsilateral lobe than that of the primary tumor or invades any of the following
structures: Diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal
nerve, esophagus, vertebral body, and carina
Source: Goldstraw et al., The IASLC Lung Cancer Staging Project: Proposals for Revision of the
TNM Stage Groupings in the Forthcoming (Eighth) Edition of the TNM Classification for Lung
Cancer. https://doi.org/10.1016/j.jtho.2015.09.009. PMID: 26762738
M Component
Table 8.4 summarizes the M component. Nodules located in the pleura or in the
pericardium, as well as pleural or pericardial effusion and contralateral or bilateral
metastases in the lung parenchyma, are considered M1a. The M1b category identi-
fies a tumor with a single extrathoracic metastasis, while the M1c category encom-
passes tumors with multiple extrathoracic metastases.
Table 8.4 N and M stage definitions in the eighth edition of the TNM in lung cancer
N: Regional lymph node involvement
Nx Regional lymph nodes cannot be assessed
N0 No regional lymph node metastasis
N1 Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and
intrapulmonary nodes, including involvement by direct extension
N2 Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s)
N3 Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral
scalene, or supraclavicular lymph node(s)
M: Distant metastasis
M0 No distant metastasis
M1 Distant metastasis present
M1a Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial
nodule(s) or malignant pleural or pericardial effusion
M1b Single extrathoracic metastasis
M1c Multiple extrathoracic metastases in one or more organs
Source: Goldstraw et al., The IASLC Lung Cancer Staging Project: Proposals for Revision of the
TNM Stage Groupings in the Forthcoming (Eighth) Edition of the TNM Classification for Lung
Cancer. https://doi.org/10.1016/j.jtho.2015.09.009. PMID: 26762738
Table 8.5 Correspondence between lung cancer stages and TNM categories
T/M Label N0 N1 N2 N3
T1a ≤ 1 IA1 IIB IIIA IIIB
T1 T1b > 1-2 IA2 IIB IIIA IIIB
T1c > 2-3 IA3 IIB IIIA IIIB
T2a Cent, Pl IB IIB IIIA IIIB
T2 T2 a > 3-4 IB IIB IIIA IIIB
T2b > 4-5 IIA IIB IIIA IIIB
T3 > 5-7 IIB IIIA IIIB IIIC
T3 T3 Inv IIB IIIA IIIB IIIC
T3 Saltell IIB IIIA IIIB IIIC
T4 > 7 IIIA IIIA IIIB IIIC
T4 T4 Inv IIIA IIIA IIIB IIIC
T4 Ipsi Nod IIIA IIIA IIIB IIIC
M1a Contr nod IVA IVA IVA IVA
M1a Pl dissem IVA IVA IVA IVA
M1
M1b Single IVA IVA IVA IVA
M1c Multi IVB IVB IVB IVB
Source: Detterbeck et al., The Eighth Edition Lung Cancer Stage Classification.
https://doi.org/10.1016/j.chest.2016.10.010
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 193
Type IA1 IA2 IA3 IB IIA IIB IIIA IIIB IIIC IVA IVB
Clinical 92 83 77 68 60 53 36 26 13 10 0
Pathologic 90 85 80 73 65 56 41 24 12 – –
Source: Detterbeck et al., The Eighth Edition Lung Cancer Stage Classification. https://doi.
org/10.1016/j.chest.2016.10.010
Stage I: In this case the tumor is limited to the lungs and there is no nodal involvement.
Stage II: The neoplastic tissue is located in the lung and in the ipsilateral peribron-
chial or hilar nodes.
Stage III: The tumor is locally advanced and invades mediastinal nodes. In particular:
Stage IIIa: The neoplastic extension is only in ipsilateral mediastinal nodes.
Stage IIIb: The neoplastic extension is into contralateral mediastinal nodes or above
the clavicle.
Stage IV: The tumor is diffused into both lungs, to the pleural fluid or any other
body part [26].
In a global perspective, the improvement of general life conditions and the develop-
ment of medical science have led to a stable increase in life expectancy worldwide.
Therefore, the age profile of the society is changing; due to the longer life of the
population and the combined reduction in fertility, we have seen a reduction of
people of working age and an increase in the proportion of the elderly population,
commonly referred to as people >65 years old, a phenomenon that is often referred
to as “demographic aging.” This trend, which first started in the richest countries in
the world, has involved all developing countries and is expected to continue in the
next couple of decades. These developments are likely to have profound implica-
tions, among others, in health and social care systems [44].
As we age, the human body is subjected to progressive decay due to physiologi-
cal metabolic, hormonal, and anatomical changes. The respiratory system and the
parenchymal, vascular, and osteo-cartilage structures that compose it are character-
ized by numerous physiological and pathological factors that determine a progres-
sive remodeling over the years. It is mandatory for the radiologist to know the most
important anatomical and physiological evolutionary characteristics of old age, first
of all of the chest, so as to be able to establish the real limit between changes in the
chest compatible with age and proper pathological pictures.
The constant aging of the general population, the condition of fragility and
greater predisposition to the disease associated with the aging of the body, the con-
stant improvement of the imaging technologies available to the radiologist, and the
194 Z. Falaschi et al.
The thoracic cage represents the anatomical structure responsible for carrying out
the function of “container” of the viscera of the thorax. It consists of the ribs, the
sternum, the dorsal spine, the diaphragm, and the muscles of the anterior and poste-
rior thoracic wall.
With aging, the skeleton of the rib cage undergoes a progressive stiffening, a
consequence of the calcification of the costal cartilages and the arthritic-degenerative
processes affecting the costo-vertebral joints. These events, which are associated
with the progressive rarefaction of the bone matrix due to pathological condition
commonly present in the elder (i.e., osteoporosis) and the physiological reduction
of the tone of the parietal muscles, result in an increase in the fragility of the chest
wall, which results in a greater predisposition to fractures and deformation of the
bone components. Reduced thickness of intervertebral determinate worsening dor-
sal kyphosis and cause the onset of dorsal-lumbar scoliosis, which together with the
factors listed above determine deformation and reduction of compliance of the rib
cage. Other errors may be caused by frequent findings in the elderly such as rib
compact islands, vertebral osteophytic bridges, costo-transverse arthritic hypertro-
phy, all situations that can simulate parenchymal nodular opacity [46].
Even the involutional aspects affecting the muscle tissue are a source of interpre-
tation errors for the radiologist. In the elderly subject, there is a progressive replace-
ment of muscle cells with fat cells, resulting in the progressive atrophy of the
muscles of the chest wall. The atrophy of the muscle component, while not neces-
sarily altering the thickness of the chest wall, in any case determines a significant
alteration of the constitution. Such modifications may go unnoticed on a careful
physical examination, but are always apparent on the examination performed using
diagnostic imaging. On a standard chest radiographic examination, it is possible to
observe widespread radiolucency of the thoracic resulting in the lower radio-
attenuating capacity of the adipose component. This finding becomes even more
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 195
evident with the aid of the tomographic examination (CT) which allows a better
definition through the acquisition of multiplanar sequences.
Fig. 8.9 Axial and coronal CT scan of a 83-year-old man with cardiomegalia. The volumetric
increase in the heart chambers is appreciable, in particular in the left ventricle
196 Z. Falaschi et al.
Fig. 8.10 Samples of standard chest radiograph in pulmonary emphysema with “saber sheath”
trachea. It represents degeneration and ossification of the tracheal cartilage due to elevated intra-
thoracic pressure
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 197
Fig. 8.11 Aortic ectasia in the ascendant and aortic arch in an 88-year-old male, observed as a
prominent aortic knob
198 Z. Falaschi et al.
The findings of emphysema and chronic bronchitis in old age are the same as
those found in younger subjects, but more difficult to interpret correctly given the
frequent coexistence of alterations in other systems (cardiovascular, musculoskele-
tal) that can simulate them or mask the radiological manifestations. It is important
for the radiologist to know the pathophysiological basis of the pathologies of the
cardio-pulmonary circulation to avoid any misinterpretation of the image. For
example, a common mistake for the radiologist is to confuse the radiolucency of the
pulmonary fields resulting from the already described physiological involution of
the muscular structures for a picture of senile emphysema, in the absence of those
signs necessary to confirm a picture of hyperinsufflation, oligoemia, or of blisters
that must be present to confirm the diagnosis of emphysema [50].
Furthermore, in the elderly patients, the worsening of the pathological state is
often accompanied by nonspecific clinical manifestations such as asthenia, retroster-
nal pain, and deterioration of cognitive functions, thus leading to a late or inade-
quate diagnosis.
Both traditional radiology and computed tomography (CT) can provide an
important help in giving an initial diagnostic address to the appearance of the first
symptoms.
There are two classic radiological-clinical patterns that guide the physician in the
diagnosis of emphysema/chronic bronchitis in the elderly: the first pattern, arterial
deficiency, characterized by the rarefaction of the vascular pattern, corresponding to
the prevalence of the emphysematous picture; the second pattern, increased mark-
ings, sees the accentuation of vascular landmarks characteristic of the prevalence of
chronic bronchitis which is associated with secondary pulmonary arterial
hypertension.
Among the radiographic alterations observable in COPD reported in the litera-
ture and may concern alterations of the diaphragm (widening of the costo-phrenic
angles, lowering of the diaphragmatic dome, widening of the clear retrosternal
space), parenchymal (presence of emphysematous bubbles, thickening of the bron-
chial walls, areas of opacity), cardiovascular alterations (heart drop, dilation of the
pulmonary arteries with peripheral barrage), and alterations of the trachea (“saber
sheath” trachea).
The principle can be stated that, even in the presence of multiple concomitant
radiographic alterations, the standard X-ray is not very sensitive in the mild-
moderate severity forms of COPD, it may also appear negative in patients with
chronic bronchitis, while it acquires greater usefulness in the most advanced forms,
although it is not possible to correctly quantify the extent or extent of functional or
anatomical damage. It is generally used in the presence of acute onset of symptoms
with fever to confirm the clinical suspicion of a bronchopneumonic outbreak, deter-
mining the presence of any complications, such as pleural effusion and bronchial
obstructions. Moreover, it is often technically inadequate since it is carried out in
nonoptimal conditions, with the patient in a semi-sitting position, insufficient inspi-
ration, with portable equipment, which further reduces its already limited diagnostic
value [51].
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 199
Finally, the chest X-ray can also detect, in asthmatic forms, nonspecific radio-
logical signs, such as thickening of the bronchial walls and bronchopneumonic foci,
or recognize the acute complications of an asthmatic event, such as pneumothorax
or pneumomediastinum.
CT is more useful in this regard, which allows us to quantify with sufficient pre-
cision the extent of emphysema in HRCT and the severity of emphysematous dam-
age even in the mildest forms; it is possible to demonstrate with HRCT the presence
of emphysema in patients with impaired respiratory function and completely nor-
mal chest radiograph. In the pattern, increased markings are often observed in
HRCT, in addition to the reduced parenchymal density, oligoemia, indicative of
emphysema, thickening of the bronchial and bronchiolar walls and areas of ground
glass opacity.
Finally, HRCT allows precise documentation of the type of prevailing
emphysema:
• Centrilobular emphysema, the most common type usually associated with smok-
ing, affects the centrilobular portion of the lung. Usually the upper lobes of the
lungs are affected.
• Panlobular emphysema, also called panacinar emphysema, can involve the
whole lung or mainly the lower lobes. Is commonly associated with alpha-1 anti-
trypsin deficiency (A1AD or AATD).
• Paraseptal emphysema, also called distal acinar emphysema, relates to emphy-
sematous change next to a pleural surface. The cystic spaces known as blebs or
bullae that form in paraseptal emphysema typically occur in just one layer
beneath the pleura (Fig. 8.12).
In the forms of severe chronic asthma, HRCT, much better than the thoracic
radiogram, is able to demonstrate and quantify the thickening of the bronchial walls
due to the structural remodeling of the walls and can detect the frequent presence of
bronchiolitis highlighting direct signs (branched opacities from commitment of the
a b c d
Fig. 8.12 Comparative images in axial computed tomography. (a) Absence of emphysema; (b)
Centrilobular emphysema; (c) Paraseptal emphysema; (d) Panlobular emphysema
200 Z. Falaschi et al.
8.4.4 Pneumonia
Pneumonia represents the main causes of death from infection, especially in the
elderly population where the immune system is often compromised due to an age-
related decrease in immune activity, chronic use of medications altering immune
function as the use of systemic corticosteroids in rheumatic disease.
Pneumonia can be divided into typical or atypical presentation, and in accor-
dance to history in community acquired, nosocomial, or infections in the
immunocompromised.
Conventional chest radiography plays a major role in diagnosing pneumonia,
being able to detect or exclude infiltrates, show the extent of the disease, estimate
possible complications, and show response to treatment. However, it is not indicated
in the suspicion of pneumonia in an immunocompromised patient, where the image
obtained is often normal.
Supported by clinical history and clinical laboratory data, chest radiography
allows to limit the spectrum of possible pathogens and will guide the calculated use
of antibiotics.
It is important to know the possible differential diagnoses in the presence of
certain signs, such as the presence of persistent infiltrations, which can lead to a
diagnosis of bronchoalveolar carcinoma. In the elderly patients, elimination of pul-
monary infiltration usually takes longer. It was shown that 15% of elderly patients
still showed radiographic abnormalities beyond 3 months. Delayed clearance may
be related to existing comorbidity. For this reason, a minimum interval of 3 months
for follow-up radiograph appears to be indicated to rule out previous malignant
changes.
8.4.5 Conclusion
In the elderly, conventionally defined as individuals >65 years of age, it is often dif-
ficult to establish what is normal, due to the numerous anatomical and physiological
changes that occur during the physiological process of aging. Knowing how to dis-
tinguish normal pictures from strictly pathological pictures is an important chal-
lenge for the radiologist today. Diagnostic imaging often offers borderline results to
chest imaging, and it is essential to know how to distinguish a picture that is only
apparently pathological from an image that may underlie the principle of pulmonary
pathologies for which the elderly patient, due to the condition of fragility associated
with aging, is particularly vulnerable.
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 201
During the COVID-19 pandemic, clinicians have been forced to balance the risk
of delaying potentially necessary evaluation and management against the risks of
exposing patients to the virus in hospital settings, or exposing healthcare workers to
patients who may be asymptomatic carriers of the disease [57].
In this scenario, consensus statements were developed to guide clinicians man-
aging lung cancer screening programs and patients with lung nodules during the
COVID-19 pandemic, concluding that it is [58, 59] appropriate to defer enrollment
in lung cancer screening and modify the evaluation of lung nodules due to the added
risks from potential exposure. In particular more than 95% of the clinicians involved
in the “CHEST expert panel” agreed to delay the evaluation of pulmonary nodules
detected incidentally or by screening that have a low probability of cancer or are
likely to be an indolent cancer [58, 60–67].
8.6.1.1 Introduction
Lung cancers which fit into stage I (cT1N0 and cT2N0) or stage II (cT1N1, cT2N1
and cT3N0) can be eligible for surgery. Patients with a Stage IIIA neoplasia (cT3N1
and cT1–3N2) have a low probability of disease eradication by surgery alone, but
they may be considered eligible after adjuvant therapy.
Up to 76% of lung cancer patients undergo some kind of surgery during their
treatment journey. The commonly performed thoracic intervention on patients with
lung cancer is the pulmonary resection. There are two main types of pulmonary
resection: anatomical and non-anatomical. Anatomical resections are those which
respect the scissures and/or the lung anatomical divisions, such as pneumonectomy,
lobectomy, segmentectomy, or segmental resection. Two lobes in the right lung can
also be removed in an upper lobectomy (excision of the upper and middle lobes) or
in a lower lobectomy (excision of the lower and middle lobes). On the contrary, non-
anatomical resections, which are also called atypical, usually remove a wedge-
shaped portion of the lung parenchyma without respecting the anatomical boundaries.
Each procedure is subject to distinct postoperative complications, both early and
delayed, which radiologists usually encounter in day-to-day practice.
a b
c d
Fig. 8.13 (a) Chest X-ray obtained the first day after left pneumonectomy. (b) Chest X-ray
obtained on the third day after the intervention in the same patient. (c, d) Postero-anterior and
latero-lateral projections on the eighth day after the pneumonectomy. The images display how the
left chest cavity is gradually filled with fluid and the mediastinum is shifted toward the oper-
ated side
cavity gradually filling with fluid. On the contrary, an increase in air component
may indicate the presence of a perforation or a bronchopleural fistula. After the first
month the residual lung tends to compensatory hyperinflation, while the mediastinal
structures become furtherly shifted toward the operated side.
When an anatomical or an atypical partial resection is performed, volume loss is
obviously expected. Pneumothorax may be present immediately after the surgery, as
shown in Fig. 8.14. Surgical clips may be visible in the intervention site, where
atelectasis or bleeding may also be found in the days after surgery. Muscle flaps are
often used in order to obstruct the bronchi and prevent pneumothorax after lobec-
tomy or pneumonectomy. Muscle tissue is commonly collected from the intercostal
and from the serratus anterior; bone tissue may be present in the flap if periosteal
tissue from the adjacent rib is included.
After the median longitudinal sternotomy, cerclage wires surrounding the sternal
body are visible. If a traditional thoracotomy was performed a rib transection will
be seen, while in case of an “en bloc” chest resection several consecutive ribs will
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 205
a b
Fig. 8.14 (a) Chest X-ray performed the first day after a superior right lobectomy. The right dia-
phragm is elevated, subcutaneous edema and apical right pneumothorax are present. There is a
basal drainage to collect pleural fluids and an apical drainage to collect air. (b) Chest X-ray per-
formed on the same patient 3 days after the intervention. Resolution of the pneumothorax, no
pleural effusion
8.6.1.4 Chest CT
Patients with stage I or II lung cancer who were treated primarily with surgery
should undergo a chest CT with intravenous contrast administration every 6 month
for 2–3 years after the intervention according to the National Comprehensive Cancer
Network (NCNN) guidelines. After that period of time, a noncontrast enhanced
low-dose CT should be performed every year. On the other hand, patients with stage
III lung cancer treated for curative and not palliative intent should undergo a con-
trast enhanced chest CT every 3–6 month for 3 years, followed by a contrast
enhanced CT every 6 months for the subsequent 2 years and finally by a noncontrast
enhanced low-dose CT performed annually.
space may be demonstrated on chest CT, but not in every case. This complication
often requires surgical intervention. Figure 8.15 shows an example of this
complication.
Pneumonia: Aspiration, poor pain control, and mechanical ventilation may lead
to pneumonia in the postoperative days. Radiological findings include ground glass
opacities, consolidations, or cavitation that may be both peribronchial or subpleu-
ral. Pleural effusion may be present; an empyema may form following surgical
contamination or preexisting infections. Pneumonectomy patients are the most
affected.
Adult respiratory distress syndrome (ARDS): From 2 to 15% of patients undergo-
ing thoracotomy develop a diffuse damage of the alveolar-capillary barrier resulting
in acute lung injury (ALI). Clinically, respiratory failure and decreased PaO2/FiO2
ratio may be present. Findings on chest X-ray are nonspecific and are similar to
those of typical pulmonary edema and pulmonary hemorrhage. On chest CT, the
typical pattern involves bilateral dishomogeneous pulmonary opacifications that
usually form a gravitational pattern, with lung consolidations in the most dependent
areas and ground glass opacities in the superior regions. Additionally, bronchial
dilatation in the ground glass areas and pulmonary cysts may be present. The prog-
nosis is poor, with mortality that can be as high as 50%.
Pulmonary edema: This complication is more common after pneumonectomy. It
is caused by augmented hydrostatic pressure and by altered alveolo-capillary bar-
rier. Pulmonary edema is more common in patients who underwent abundant peri-
operative fluid resuscitations or plasma transfusions; even patients who suffer from
arrhythmias are more affected. On chest X-ray common findings include interlobu-
lar septal thickening, Kerley B lines, and diffuse alveolar opacities. The most impor-
tant differential diagnoses include pneumonia and ARDS.
Hemothorax: A damage to pulmonary or systemic vasculature can lead to a pleu-
ral space hematoma in up to 1.3% of operated patients. On X-rays a rapidly increas-
ing pleural effusion may generate the suspicion of pleural hematoma and lead to
further investigations. On chest CT, the pleural collection demonstrates diffusely
high density values ranging from 40 to 90 Hounsfield units. A surgical reinterven-
tion to evacuate the collection is often needed.
Lung torsion: This is an infrequent complication with modern surgery tech-
niques. However, a pulmonary lobe can undergo a torsion in the setting of pleural
effusion and pneumothorax. The middle lobe is most commonly affected. On chest
radiographs, the lobe appears diffusely radiopaque and reduced in dimensions, and
also the lobar position can be unusual. On chest CT, the lobe appears diffusely
hyperdense because of atelectasis and venous congestion; the airways and the ves-
sels appear to be distorted and possibly occluded.
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 207
b c
Fig. 8.15 (a) Chest X ray obtained the first day after right inferior lobectomy shows a large right
pneumothorax. (b, c) The control X-ray obtained 30 days after the intervention shows how the
pneumothorax fails to resolve. This raised the suspicion of bronchopleural fistula, which was sub-
sequently confirmed via bronchoscopy
stage IA disease, while a limited survival benefit was demonstrated in patients with
stage IB.
Neoadjuvant chemotherapy has not been so extensively evaluated; its use may
however be beneficial since downstaging can be achieved, resulting in a less exten-
sive resection.
Patients with locally advanced lung cancer (stage III) should undergo a contrast-
enhanced total body CT to rule out distant metastases, followed by a PET/CT. A
contrast enhanced brain MRI should also be performed to exclude intracranial
localizations of the disease. Platinum based chemotherapy has proved to be effec-
tive in improving survival in stage III lung cancers, both in resectable and in unre-
sectable tumors. Targeted immunotherapy agents have proven useful in the treatment
of stage IV patients with specific mutations, such as EGFR and ALK. The role of
immunotherapeutic agents in patients with stage I, II, and III lung cancer has not yet
been evaluated properly, although several clinical trials are under way to assess the
feasibility of targeted treatment in both the adjuvant and the neoadjuvant settings.
biggest diameter should be used for all the other target lesions. This parameter is
known as baseline sum diameters.
Target lesions response criteria:
• Complete response (CR): All target lesions are no longer visible, all pathological
lymph nodes have a short axis <10 mm.
• Partial response (PR): A minimum decrease of 30% in the overall diameter of
target lesions compared to the baseline sum diameters.
• Progressive disease (PD): An increase of the sum of the diameters of target lesion
equal or superior to 20%, or the presentation of one or more new lesions.
• Stable disease (SD): Insufficient diameter reduction or increase to qualify either
for PR or for PD.
• Complete response: all nontarget lesions are no longer visible, tumor markers
are normal.
• Non-CR/non-PD: At least one nontarget lesion is still present or tumor markers
levels are superior to the norm.
• Progressive disease: Categorical progression of preexisting nontarget lesions.
The response criteria of target and nontarget lesions are to be evaluated together,
identifying the best overall response as shown in Table 8.8. Figures 8.16 and 8.17
show two clinical examples of partial response and progressive disease, respectively.
8.6.2.3 iRECIST
In recent years a new category of antineoplastic drugs has been developed, and it
evolved into one of the most important in the treatment of aggressive tumors: immu-
nomodulators. These highly specific pharmaceuticals act on specific intracellular
pathways, such as CTLA 4, PD-1, and PD-L1, and agents active on those pathways
have been marketed since 2011. Neoplastic diseases commonly treated with these
agents include melanoma, bladder, kidney, lung, and head and neck cancers.
Fig. 8.16 Consecutive CT scans in a 71-year-old male patients. The images in the upper row are
from the initial CT scan, while the lower pictures are from the follow-up CT scan obtained
3 months later, after 4 cycles of chemotherapy. The pulmonary mass shrinked from 8.7 to 6.1 cm;
the pathological node’s short axis reduced from 10 to 4 mm. This can be classified as partial
response
Patients treated with these drugs can be a challenge for the radiologists, since
immunomodulators can provoke an unusual pattern of response which resembles
progressive disease. This concept has been known as pseudoprogression. From a
histopathological point of view, the phenomenon of pseudoprogression seems to be
correlated to the immune response stimulated by the drugs. The neoplastic tissue
becomes infiltrated by T CD4+ and T CD8+ lymphocytes, and the inflammatory
response may be accompanied by edema and hemorrhage. All these alterations can
and often do result in a perceived increase of the tumoral mass, while the patient is
actually responding to the therapy.
Given this response pattern, RECIST 1.1 felt inadequate to evaluate the therapy
response in patients treated with immunomodulators, and new criteria had to be
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 211
Fig. 8.17 Consecutive CT scans in a 74-year-old male patients. The images in the upper row are
from the initial CT scan, while the lower pictures are from the follow-up CT scan obtained
9 months later, after chemotherapy treatment. A new lung nodule has appeared in the right lower
lobe and the pathological node in the aorto-pulmonary window’s short axis increased from 2.7 to
3.4 cm. This is progressive disease according to RECIST 1.1
developed by the RECIST working group itself. The new criteria, apart from adding
the prefix “i” in front of the previous response evaluation classes (e.g., iCR, iPR),
defined two new key concepts: unconfirmed progressive disease, iUPD, and con-
firmed progressive disease, iCPD.
iUPD is defined just like the progressive disease in RECIST 1.1, but it needs to
be confirmed with a subsequent imaging evaluation obtained from 4 to 8 weeks after
the first examination. The progression is confirmed only if there is a further incre-
ment of the previously reported lesions, or if there is dimensional expansion of
previously stable localizations. If the progression is not confirmed, but instead the
lesions show a volume decrease in comparison with baseline values compatible
with iCR, iPR, or iSD, the bar is reset. In this case, iUPD has to manifest again and
then to be confirmed in order to have a confirmed progressive disease (iCPD). As a
212 Z. Falaschi et al.
consequence iUPD can be assigned several times, as long as the progression is not
confirmed in the following assessment. The appearance of new lesions results in
iUPD, but the progression is confirmed only if other new lesions are present in the
confirmatory evaluation or if the new lesions show dimensional increase. If iUPD
was given only on the basis of target or nontarget lesions, then the progression of the
other category in the confirmatory scan also brings to iCPD.
The principles for assigning the best overall response are similar to those
described in RECIST 1.1, but the presence of iUPD makes things a little more intri-
cate. In general, the best overall response in iRECIST (iBOR) is the best response
to therapy recorded from the treatment start to its end, and all imaging evaluation
must be taken into account. The assignment of an iUPD category will not nullify the
following iBOR of iCR, IPR or iSD, as long as the requirements for iCPD are not
met. A complete list of examples can be found in the iRECIST presentation article
in the bibliography [51, 52, 68–77].
References
1. Parkin DM. Global cancer statistics in the year 2000. Lancet Oncol. 2001;2:533–43.
2. National Cancer Institute. Data sources: surveillance, epidemiology, and end results (SEER) 9
registries. National Cancer Institute; 2020.
3. Bravo-Iñiguez C, Perez Martinez M, Armstrong KW, et al. Surgical resection of lung cancer in
the elderly. Thorac Surg Clin. 2014;24:371.
4. Kung HC, Hoyert DL, Xu J, Vincent GK, Velkoff VA. The next four decades-the older popula-
tion in the United States: 2010 to 2050 Deaths: final data for 2005.
5. Mehta HJ, Ross C, Silvestri GA, et al. Evaluation and treatment of high-risk patients with
early-stage lung cancer. Clin Chest Med. 2011;32:783.
6. Extermann M. Measuring comorbidity in older cancer patients. Eur J Cancer. 2000;36:453.
7. Smith PW, Jones DR. Biology and epidemiology of lung cancer. In: Patterson GA, Cooper JD,
Deslauriers J, et al., editors. Pearson’s thoracic and esophageal surgery. 3rd ed. Philadelphia,
PA: Churchill Livingstone Elsevier; 2008.
8. Wingo PA, Cardinez CJ, Landis SH, et al. Long term trends in cancer mortality in the United
States; 2003.
9. Hamilton W, Peters TJ, Round A, Sharp D. What are the clinical features of lung cancer before
the diagnosis is made? A population based case-control study. Thorax. 2005;60(12):1059–65.
https://doi.org/10.1136/thx.2005.045880.
10. Nasim F, Sabath BF, Eapen GA. Lung cancer. Med Clin N Am. 2019;103(3):463–73. https://
doi.org/10.1016/j.mcna.2018.12.006.
11. Gridelli C, Perrone F, Gallo C, et al. Chemotherapy for elderly patients with advanced non
small-cell lung cancer: the multicenter Italian lung cancer in the elderly study (MILES) phase
III randomized trial. J Natl Cancer Inst. 2003;95:362–72.
12. National Center for Health Statistics: Health, United States, 7982. DHHS Pub. No. (PHS)
83–1232. Public Health Service. Washington: U.S. Government Printing Office; 1982.
13. Peto R, Darby S, Deo H, Silcocks P, Whitley E, Doll R. Smoking, smoking cessation, and lung
cancer in the UK since 1950: combination of national statistics with two case-control studies.
BMJ. 2000;321(7257):323–9.
14. Alberg AJ, Samet JM. Epidemiology of lung cancer. Chest. 2003;123:21S.
15. Ries LAG, Eisner MP, Kosary CL, et al. SEER Cancer Statistics Review 1975–2000.
16. Van Meerbeeck JP, Fennell DA, De Ruysscher DKM. Small-cell lung cancer. Lancet.
2011;378(9804):1741–55. https://doi.org/10.1016/S0140-6736(11)60165-7.
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 213
17. Bradley SH, Grice A, Neal RD, Abraham S, Rodriguez Lopez R, Shinkins B, Callister
MEJ, Hamilton WT. Sensitivity of chest X-ray for detecting lung cancer in people present-
ing with symptoms: a systematic review. Br J Gen Pract. 2019;69(689):E827–35. https://doi.
org/10.3399/bjgp19X706853.
18. Missrie I, Hochhegger B, Zanon M, Capobianco J, de Macedo C, Neto A, Maciel RP, Antunes
VB, de Figueiredo CM, Szarf G, Meirelles G. Small low-risk pulmonary nodules on chest digi-
tal radiography: can we predict whether the nodule is benign? Clin Radiol. 2018;73(10):902–6.
https://doi.org/10.1016/j.crad.2018.06.002.
19. William H. Learning radiology; n.d.
20. Vogl TJ, Reith W, Rummeny EJ. Diagnostic and interventional radiology. In: Diagnostic and
interventional radiology; 2016. https://doi.org/10.1007/978-3-662-44037-7.
21. Hansell DM, Bankier AA, MacMahon H, McLoud TC, Müller NL, Remy J. Fleischner soci-
ety: glossary of terms for thoracic imaging. Radiology. 2008;246(3):697–722. Epub 2008 Jan
14. PMID: 18195376. https://doi.org/10.1148/radiol.2462070712.
22. MacMahon H, Naidich DP, Goo JM, Lee KS, Leung ANC, Mayo JR, Mehta AC, Ohno Y,
Powell CA, Prokop M, Rubin GD, Schaefer-Prokop CM, Travis WD, Van Schil PE, Bankier
AA. Guidelines for management of incidental pulmonary nodules detected on CT images:
from the Fleischner Society 2017. Radiology. 2017;284(1):228–43. Epub 2017 Feb 23. PMID:
28240562. https://doi.org/10.1148/radiol.2017161659.
23. Gruden JF, Ouanounou S, Tigges S, Norris SD, Klausner TS. Incremental benefit of maximum-
intensity-projection images on observer detection of small pulmonary nodules revealed by
multidetector CT. AJR Am J Roentgenol. 2002;179(1):149–57. PMID: 12076925. https://doi.
org/10.2214/ajr.179.1.1790149.
24. Bueno J, Landeras L, Chung JH. Updated Fleischner society guidelines for managing inci-
dental pulmonary nodules: common questions and challenging scenarios. Radiographics.
2018;38(5):1337–50. PMID: 30207935. https://doi.org/10.1148/rg.2018180017.
25. Hasegawa M, Sone S, Takashima S, Li F, Yang ZG, Maruyama Y, Watanabe T. Growth rate of
small lung cancers detected on mass CT screening. Br J Radiol. 2000;73(876):1252–9. PMID:
11205667. https://doi.org/10.1259/bjr.73.876.11205667.
26. Detterbeck FC, Boffa DJ, Kim AW, Tanoue LT. The eighth edition lung cancer stage clas-
sification. Chest. 2017;151(1):193–203. Epub 2016 Oct 22. PMID: 27780786. https://doi.
org/10.1016/j.chest.2016.10.010.
27. Cittadini G, Cittadini G, Sardarelli F. Cittadini—Diagnostica per immagini e radioterapia; n.d.
28. Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WE, Nicholson
AG, Groome P, Mitchell A, Bolejack V, International Association for the Study of Lung Cancer
Staging and Prognostic Factors Committee, Advisory Boards, and Participating Institutions;
International Association for the Study of Lung Cancer Staging and Prognostic Factors
Committee Advisory Boards and Participating Institutions. The IASLC Lung Cancer Staging
Project: Proposals for Revision of the TNM Stage Groupings in the Forthcoming (Eighth)
Edition of the TNM Classification for Lung Cancer. J Thorac Oncol. 2016;11(1):39–51.
26762738. https://doi.org/10.1016/j.jtho.2015.09.009.
29. Carter BW, Lichtenberger JP 3rd, Benveniste MK, et al. Revisions to the TNM staging of lung
cancer: rationale, significance, and clinical application. Radiographics. 2018;38(2):374–91.
https://doi.org/10.1148/rg.2018170081.
30. Beasley MB, Brambilla E, Travis WD. The 2004 World Health Organization classification of
lung tumors. Semin Roentgenol. 2005;40(2):90–7. PMID: 15898407. https://doi.org/10.1053/j.
ro.2005.01.001.
31. Govindan R, Page N, Morgensztern D, Read W, Tierney R, Vlahiotis A, Spitznagel EL,
Piccirillo J. Changing epidemiology of small-cell lung cancer in the United States over the last
30 years: analysis of the surveillance, epidemiologic, and end results database. J Clin Oncol.
2006;24(28):4539–44. PMID: 17008692. https://doi.org/10.1200/JCO.2005.04.4859.
32. Zhao YR, Heuvelmans MA, Dorrius MD, van Ooijen PM, Wang Y, de Bock GH, Oudkerk M,
Vliegenthart R. Features of resolving and nonresolving indeterminate pulmonary nodules at
214 Z. Falaschi et al.
follow-up CT: the NELSON study. Radiology. 2014;270(3):872–9. Epub 2013 Nov 18. PMID:
24475806. https://doi.org/10.1148/radiol.13130332.
33. Rosado-de-Christenson ML, Templeton PA, Moran CA. Bronchogenic carcinoma: radiologic-
pathologic correlation. Radiographics. 1994;14(2):429–46. quiz 447–8. PMID: 8190965.
https://doi.org/10.1148/radiographics.14.2.8190965.
34. Hollings N, Shaw P. Diagnostic imaging of lung cancer. Eur Respir J. 2002;19(4):722–42.
PMID: 11999004. https://doi.org/10.1183/09031936.02.00280002.
35. Austin JH, Garg K, Aberle D, Yankelevitz D, Kuriyama K, Lee HJ, Brambilla E, Travis
WD. Radiologic implications of the 2011 classification of adenocarcinoma of the lung.
Radiology. 2013;266(1):62–71. Epub 2012 Oct 15. PMID: 23070271. https://doi.org/10.1148/
radiol.12120240.
36. Mets OM, Chung K, Scholten ET, Veldhuis WB, Prokop M, van Ginneken B, Schaefer-Prokop
CM, de Jong PA. Incidental perifissural nodules on routine chest computed tomography:
lung cancer or not? Eur Radiol. 2018;28(3):1095–101. Epub 2017 Oct 6. PMID: 28986629;
PMCID: PMC5811588. https://doi.org/10.1007/s00330-017-5055-x.
37. Lambe G, Durand M, Buckley A, Nicholson S, McDermott R. Adenocarcinoma of the
lung: from BAC to the future. Insights Imaging. 2020;11(1):69. PMID: 32430670; PMCID:
PMC7237554. https://doi.org/10.1186/s13244-020-00875-6.
38. Aoki T, Nakata H, Watanabe H, Nakamura K, Kasai T, Hashimoto H, Yasumoto K, Kido
M. Evolution of peripheral lung adenocarcinomas: CT findings correlated with histology and
tumor doubling time. AJR Am J Roentgenol. 2000;174(3):763–8. PMID: 10701622. https://
doi.org/10.2214/ajr.174.3.1740763.
39. Altmayer S, Verma N, Francisco MZ, Almeida RF, Mohammed TL, Hochhegger
B. Classification and imaging findings of lung neoplasms. Semin Roentgenol.
2020;55(1):41–50. Epub 2019 Oct 25. PMID: 31964479. https://doi.org/10.1053/j.
ro.2019.10.002.
40. Sholl LM. Large-cell carcinoma of the lung: a diagnostic category redefined by immunohis-
tochemistry and genomics. Curr Opin Pulm Med. 2014;20(4):324–31. . PMID: 24811836.
https://doi.org/10.1097/MCP.0000000000000068.
41. Carter BW, Glisson BS, Truong MT, Erasmus JJ. Small cell lung carcinoma: staging, imaging,
and treatment considerations. Radiographics. 2014;34(6):1707–21. PMID: 25310425. https://
doi.org/10.1148/rg.346140178.
42. Benson RE, Rosado-de-Christenson ML, Martínez-Jiménez S, Kunin JR, Pettavel
PP. Spectrum of pulmonary neuroendocrine proliferations and neoplasms. Radiographics.
2013;33(6):1631–49. PMID: 24108555. https://doi.org/10.1148/rg.336135506.
43. Lee D, Rho JY, Kang S, Yoo KJ, Choi HJ. CT findings of small cell lung carcinoma: can rec-
ognizable features be found? Medicine (Baltimore). 2016;95(47):e5426. PMID: 27893684;
PMCID: PMC5134877. https://doi.org/10.1097/MD.0000000000005426.
44. Sharma G, Goodwin J. Effect of aging on respiratory system physiology and immunology.
Clin Interv Aging. 2006;1(3):253–60. https://doi.org/10.2147/ciia.2006.1.3.253.
45. Gossner J, Nau R. Geriatric chest imaging: when and how to image the elderly lung, age-
related changes, and common pathologies. Radiol Res Pract. 2013;2013:584793. https://doi.
org/10.1155/2013/584793.
46. Brandsma C-A, de Vries M, Costa R, Woldhuis RR, Königshoff M. Wim Timens lung
ageing and COPD: is there a role for ageing in abnormal tissue repair? Eur Respir Rev.
2017;26(146):170073. https://doi.org/10.1183/16000617.0073-2017.
47. Groenink M, Langerak SE, Vanbavel E, van der Wall EE, Mulder BJ, van der Wal AC, Spaan
JA. The influence of aging and aortic stiffness on permanent dilation and breaking stress of
the thoracic descending aorta. Cardiovasc Res. 1999;43(2):471–80. https://doi.org/10.1016/
S0008-6363(99)00095-4.
48. Freeman LM, Stein EG, Sprayregen S, Chamarthy M, Haramati LB. The current and continu-
ing important role of ventilation-perfusion scintigraphy in evaluating patients with suspected
pulmonary embolism. Semin Nucl Med. 2008;38(6):432–40.
8 Neoplastic Diseases of the Respiratory System in Geriatric Patients 215
49. Lowery EM, Brubaker AL, Kuhlmann E, Kovacs EJ. The aging lung. Clin Interv Aging.
2013;8:1489–96. https://doi.org/10.2147/CIA.S51152.
50. Copley SJ, Wells AU, Hawtin KE, et al. Lung morphology in the elderly: comparative CT study
of subjects over 75 years old versus those under 55 years old. Radiology. 2009;251(2):566–73.
51. British Thoracic Society; Society of Cardiothoracic Surgeons of Great Britain and Ireland
Working Party. BTS guidelines: guidelines on the selection of patients with lung cancer for
surgery. Thorax. 2001;56(2):89–108. PMID: 11209097; PMCID: PMC1745996. https://doi.
org/10.1136/thorax.56.2.89.
52. Cassidy A, Myles JP, et al. The LLP risk model: an individual risk prediction model for lung
cancer. Br J Cancer. 2008;98:270–6.
53. de Koning HJ, van der Aalst CM, et al. Reduced lung-cancer mortality with volume CT screen-
ing in a randomized trial. N Engl J Med. 2020;382(6):503–13.
54. Goffin JR. Biennial lung cancer screening in Canada with smoking cessation-outcomes and
cost-effectiveness. Lung Cancer. 2016;101:98–103.
55. Becker N, Motsch E, Trotter A, et al. Lung cancer mortality reduction by LDCT screening-
results from the randomized German LUSI trial. Int J Cancer. 2020;146(6):1503–13. https://
doi.org/10.1002/ijc.32486.
56. Heuvelmans MA. Appropriate screening intervals in low-dose CT lung cancer screening.
Transl Lung Cancer Res. 2018;7(3):281–7.
57. Mazzone PJ. Management of Lung Nodules and Lung Cancer Screening during the COVID-19
pandemic: CHEST expert panel report. Chest. 2020;158(1):406–15.
58. Ten Haaf KT, Bastani M, et al. A comparative modeling analysis of risk-based lung cancer
screening strategies. J Natl Cancer Inst. 2020;112:466–79.
59. US Preventive Services Task Force. Screening for Lung Cancer: US Preventive Services Task
Force Recommendation Statement. JAMA. 2021;325:962–70.
60. National Lung Screening Trial Research Team. Reduced lung-cancer mortality with low-dose
computed tomographic screening. N Engl J Med. 2011;365(5):395–409.
61. Oudkerk M, et al. European position statement on lung cancer screening. Lancet Oncol.
2017;18:e754–66.
62. Oudkerk ML, et al. Lung cancer LDCT screening and mortality reduction—evidence, pitfalls
and future perspectives. Nat Rev Clin Oncol. 2021;18:135–51.
63. Pastorino US. Prolonged lung cancer screening reduced 10-year mortality in the MILD trial:
new confirmation of lung cancer screening efficacy. Ann Oncol. 2019;30(7):1162–9.
64. Pham D, Bhandari S, et al. Lung cancer screening registry reveals low-dose CT screening
remains heavily underutilized. Clin Lung Cancer. 2020;21:e206–11.
65. Radiology AC. Lung RADS version 1.1. 2019.
66. Rampinelli C, De Marco P, et al. Exposure to low-dose computed tomography for lung can-
cer screening and risk of cancer: secondary analysis of trial data and risk-benefit analysis.
BMJ. 2017;356:j347.
67. Tammemägi MC, Haaf KT, et al. Development and validation of a multivariable lung can-
cer risk prediction model that includes low-dose computed tomography screening results:
a secondary analysis of data from the National Lung Screening Trial. JAMA Netw Open.
2019;2:e190204.
68. Brunelli A, Charloux A, Bolliger CT, Rocco G, Sculier J-P, Varela G, Licker M, Ferguson
MK, Faivre-Finn C, Huber RM, Clini EM, Win T, De Ruysscher D, Goldman L. ERS/ESTS
clinical guidelines on fitness for radical therapy in lung cancer patients (surgery and chemo-
radiotherapy). Eur Respir J. 2009;34(1):17–41. https://doi.org/10.1183/09031936.00184308.
69. American Cancer Society. Cancer treatment and survivorship facts & figures 2014–2015.
Atlanta: American Cancer Society; 2014.
70. Cardinale L, Priola AM, Priola SM, Boccuzzi F, Dervishi N, Lisi E, Veltri A, Ardissone
F. Radiological contribution to the diagnosis of early postoperative complications after lung
resection for primary tumor: a revisional study. J Thorac Dis. 2016;8(8):E643–52. PMID:
27621893; PMCID: PMC4999725. https://doi.org/10.21037/jtd.2016.07.02.
AL GRAWANY
216 Z. Falaschi et al.
71. de Groot PM, Truong MT, Godoy MCB. Postoperative imaging and complications in resec-
tion of lung cancer. Semin Ultrasound CT MR. 2018;39(3):289–96. Epub 2018 Mar 1. PMID:
29807639. https://doi.org/10.1053/j.sult.2018.02.008.
72. Chae EJ, Seo JB, Kim SY, Do KH, Heo JN, Lee JS, Song KS, Song JW, Lim TH. Radiographic
and CT findings of thoracic complications after pneumonectomy. Radiographics.
2006;26(5):1449–68. PMID: 16973775. https://doi.org/10.1148/rg.265055156.
73. Ettinger DS, Wood DE, Akerley W, Bazhenova LA, Borghaei H, Camidge DR, Cheney RT,
Chirieac LR, D’Amico TA, Dilling TJ, Dobelbower MC, Govindan R, Hennon M, Horn L,
Jahan TM, Komaki R, Lackner RP, Lanuti M, Lilenbaum R, Lin J, Loo BW Jr, Martins R,
Otterson GA, Patel JD, Pisters KM, Reckamp K, Riely GJ, Schild SE, Shapiro TA, Sharma N,
Stevenson J, Swanson SJ, Tauer K, Yang SC, Gregory K, Hughes M. NCCN Guidelines Insights:
Non-Small Cell Lung Cancer, Version 4.2016. J Natl Compr Canc Netw. 2016;14(3):255–64.
PMID: 26957612. https://doi.org/10.6004/jnccn.2016.0031.
74. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J,
Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe
D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline
(version 1.1). Eur J Cancer. 2009;45(2):228–47. PMID: 19097774. https://doi.org/10.1016/j.
ejca.2008.10.026.
75. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, Lin NU, Litière S,
Dancey J, Chen A, Hodi FS, Therasse P, Hoekstra OS, Shankar LK, Wolchok JD, Ballinger
M, Caramella C, de Vries EGE, RECIST Working Group. iRECIST: guidelines for response
criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18(3):e143–52. Epub
2017 Mar 2. Erratum in: Lancet Oncol. 2019 May;20(5):e242. PMID: 28271869; PMCID:
PMC5648544. https://doi.org/10.1016/S1470-2045(17)30074-8.
76. Postmus PE, Kerr KM, Oudkerk M, Senan S, Waller DA, Vansteenkiste J, Escriu C, Peters
S, ESMO Guidelines Committee. Early and locally advanced non-small-cell lung can-
cer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-
up. Ann Oncol. 2017;28(suppl_4):iv1–iv21. PMID: 28881918. https://doi.org/10.1093/
annonc/mdx222.
77. McWilliams A, Tammemagi MC, Mayo JR, Roberts H, Liu G, Soghrati K, Yasufuku K, Martel
S, Laberge F, Gingras M, Atkar-Khattra S, Berg CD, Evans K, Finley R, Yee J, English J,
Nasute P, Goffin J, Puksa S, Stewart L, Tsai S, Johnston MR, Manos D, Nicholas G, Goss
GD, Seely JM, Amjadi K, Tremblay A, Burrowes P, MacEachern P, Bhatia R, Tsao MS, Lam
S. Probability of cancer in pulmonary nodules detected on first screening CT. N Engl J Med.
2013;369(10):910–9. PMID: 24004118; PMCID: PMC3951177. https://doi.org/10.1056/
NEJMoa1214726.
The Gastrointestinal System in Geriatric
Patients 9
Damiano Caruso, Domenico De Santis,
Francesco Pucciarelli, and Andrea Laghi
9.1 Introduction
All organs and physiological processes of the human organism, including gastroin-
testinal system, are affected by aging [1]. Effect of aging of gastrointestinal system
includes a reduction in sensory perceptions, salivation, oral health, the absorption of
nutrients, and lactose tolerance. Although many of these age-related changes are
primarily functional, there are other changes that can be detected with imaging tech-
niques (e.g., pancreatic atrophy, lobulation, and fatty degeneration [2]). Functional
changes can then result in organic alterations and therefore become visible by imag-
ing, both directly and indirectly [3]. In this type of patient, diagnostic imaging
acquires an increasingly important value, and constant technological innovations
allow for a more accurate and early diagnosis.
One of the main problems of geriatric imaging is that patients might have multiple
comorbidities, making it difficult for them to collaborate during the examination [4].
In fact, from the simplest of exams (such as X-ray) up to technically more complex
examinations (such as MR-cholangiography), patient collaboration is a fundamental
requirement in order to reduce artifacts and achieve reliable diagnostic quality.
This chapter will be focused on the main pathologies and problems related to the
imaging of the elderly and the main strategies to solve them will be illustrated in the
following paragraphs.
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 217
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_9
218 D. Caruso et al.
9.2 Esophagus
9.2.1 Achalasia
Hiatal hernia is a condition in which elements of the abdominal cavity herniates through
the esophageal hiatus into the mediastinum; prevalence of hiatal hernia increases with
age, with a slight female predilection. The organ most commonly involved is the stom-
ach, and the most comprehensive classification of hiatal hernias divides them into four
types: sliding (type I, the most common, >90%) and paraesophageal (type II, III, and
IV). In sliding hernia, the GEJ is usually displaced >2 cm above the esophageal hiatus,
due to a widening of hiatus itself. In paraesophageal hernia, the GEJ remains in its
a b
Fig. 9.1 Axial unenhanced CT (a) and portal venous phase (b) CT images of an 87-year-old
woman with achalasia depicts a dilated and thin-walled esophagus containing air-fluid level
9 The Gastrointestinal System in Geriatric Patients 219
a b c
Fig. 9.2 Coronal (a) and sagittal reformatted (b and c) portal venous phase CT images of the same
patient showing marked dilation of the entire esophagus, characterized by thin-wall and containing
endoluminal air-fluid level
normal location while the fundus of the stomach herniates above the diaphragm through
a defect in the phrenoesophageal membrane. Mixed hernias have also been described
(type III), which simultaneously include components of type I and II. Type IV consists
of mixed type with the association of herniation of other abdominal viscera. Hiatal
hernia may be asymptomatic or cause nonspecific symptoms, such as chest pain or
epigastric pain, nausea, and vomiting [10].
a b
Fig. 9.3 Chest X-ray in posterior-anterior (a) and lateral view (b) of a 73-year-old woman with
chest pain and dysphagia; a fluid-air level is present (arrows)
a c
Fig. 9.4 MRI axial T2-weighted image without oral contrast (a), and axial and coronal
T2-weighted images acquired after ingestion of oral contrast medium (b and c) of a 73-year-old
woman with chest pain and dysphagia. A voluminous sliding hiatal hernia is confirmed
9.2.3 Ulcers
a b
Fig. 9.5 Axial (a) and coronal (b) portal venous phase CT images of a 71-year-old man with distal
esophagus cancer, showing circumferential irregular wall thickening (>5 mm; arrows)
Peptic ulcers are the consequence of increased gastric acid secretion. Helicobacter
pylori infection is the main cause of peptic ulcers, although other etiologies have
been also recognized, such as stress and the use of nonsteroidal anti-inflammatory
drugs and corticosteroids [18]. Although its incidence is showing a trend of decrease,
peptic ulcer disease still represent a global problem, with a lifetime risk of develop-
ment ranging from 5% to 10% [19]. Duodenal ulcers are four times more common
than gastric ulcers; affected patients are often asymptomatic or present with nonspe-
cific symptoms (epigastric pain, nausea, etc.) or with signs and symptoms related to
complications (bleeding, perforation, etc.) [18].
Duodenal diverticula are outpouching from the duodenal wall. They are usually
located near the ampulla of Vater and often asymptomatic. If present, symptoms
9 The Gastrointestinal System in Geriatric Patients 223
Gastric cancer is the fifth most commonly diagnosed cancer in the world, and the
seventh most prevalent [26]; the cumulative risk of developing gastric cancer from
birth to 74 years of age is 1.87% in males and 0.79% in females worldwide. The
most common gastric malignancy is adenocarcinoma (95% of malignant tumors of
the stomach); there is a strong association with Helicobacter pylori infection; and
other risk factors are represented by smoking, pernicious anemia, anthropic gastri-
tis, and adenomatous polyposis [27].
a c
Fig. 9.6 Axial (a, b) and coronal (c) portal venous phase CT images of a 79-year-old man with
duodenal cancer. Note the diffuse duodenal wall thickening (arrows) causing dilation of the main
biliary duct (asterisk) and the intrahepatic biliary tree (arrowhead)
a c
Fig. 9.7 Axial (a, b) and coronal (c) portal venous phase CT images of a 72-year-old woman with
gastric cancer demonstrates extensive, circumferential, and irregular gastric wall thickening
(arrow) and enlarged nodes in the lesser sac (arrowhead)
9 The Gastrointestinal System in Geriatric Patients 225
Primary neoplasms of the small bowel are rare, 60% of these tumors are malignant.
Peak incidence occurs in the fifth and sixth decades, clinical manifestations are
nonspecific and can include nausea, vomiting, abdominal pain, weight loss, and
melena [31].
a b
c d
Fig. 9.8 Axial unenhanced phase (a), axial (b, c), and coronal reformatted (d) portal venous phase
CT images of a 66-year-old man with acute diverticulitis, complicated with perforation. Note the
pericolic stranding (arrowheads) and the segmental bowel wall thickening (arrows). Perforation is
demonstrated by the presence of extra-intestinal air bubbles (asterisks)
9.5.2 Polyps
Polyps are wall protrusions and can be sessile or pedunculated. Mostly asymptom-
atic, they can cause symptoms in case of malignant transformation or in case of
excessive growth [37].
Colorectal cancer (CRC) is the third deadliest and fourth most commonly diagnosed
cancer in the world [39]. Incidence of CRC is expected to rise worldwide, since the
9 The Gastrointestinal System in Geriatric Patients 227
risk of developing CRC increases with age, and most countries have an ever-growing
aging population [40]. Adenocarcinoma is the most common type (98%) and, in the
vast majority of cases, arises from pre-existing colonic adenomas (neoplastic pol-
yps), which progressively undergo a malignant transformation [41].
CRC is usually asymptomatic at early stages, eventually causing symptoms due
to excessive growth or complications (bleeding, perforation, and obstruction).
a c
Fig. 9.9 Axial (a, b) and reformatted coronal (c) portal venous phase CT images of a 78-year-old
man with adenocarcinoma of the ascending colon. Note the irregular wall thickening with adjacent
fat stranding (arrows) and enlarged locoregional nodes (arrowhead)
228 D. Caruso et al.
a b
Fig. 9.10 Axial unenhanced CT (a), axial (b), and coronal (c) portal venous phase CT of a
73-year-old man with acute cholecystitis. Note the thickened and enhancing gallbladder wall sur-
rounded by fluid and adjacent fat stranding (arrows). Unenhanced CT depicts also hyperdense
gallbladder stones in the infundibulum (arrowhead)
Surgical adhesions and hernias are the main causes of small bowel obstruction,
while malignancy is the most frequent cause of large bowel obstruction.
a b
Fig. 9.11 Axial T2-weighted (a, b) and T2-weighted fat saturated (c) MR images of a 73-year-old
man with acute cholecystitis. MR images confirm the presence of gallstones in the gallbladder
infundibulum and show dilation of in the main biliary duct (arrows)
a b c
Fig. 9.12 CT scout image (a), coronal (b), and axial (c and d) portal venous phase CT images of
a 79-year-old man with volvulus. Note the “whirlpool sign” (arrows) and the dilated proximal
bowel loops filled with air
9 The Gastrointestinal System in Geriatric Patients 231
a b
Fig. 9.13 Axial unenhanced CT (a, b) of a 71-year-old man with acute pancreatitis. Indistinct
pancreatic margins, surrounding retroperitoneal fat stranding (arrowhead), and diffuse pancreatic
calcifications (arrows) are showed
232 D. Caruso et al.
a b
Fig. 9.14 Axial portal venous phase CT images (a, b) of a 67-year-old woman with mesenteric
ischemia. Note the intramural bowel gas and adjacent fat stranding adjacent to bowel loops in left
hypochondrium (arrows)
a b
Fig. 9.15 Multiple axial arterial phase images (a) and coronal maximum intensity projection CT
images (b) of a 67-year-old woman with mesenteric ischemia, depicting obstructed superior mes-
enteric artery (arrows)
9 The Gastrointestinal System in Geriatric Patients 233
References
1. Rémond D, et al. Understanding the gastrointestinal tract of the elderly to develop dietary solu-
tions that prevent malnutrition. Oncotarget. 2015;6(16):13858–98.
2. Sato T, et al. Age-related changes in normal adult pancreas: MR imaging evaluation. Eur J
Radiol. 2012;81(9):2093–8.
3. Dumic I, et al. Gastrointestinal tract disorders in older age. Can J Gastroenterol Hepatol.
2019;2019:6757524.
4. Mohammed S, Rosenkrantz AB. Providing compassionate care for the elderly patient in radi-
ology. Curr Probl Diagn Radiol. 2020;49(2):67–9.
5. Achem SR, Gerson LB. Distal esophageal spasm: an update. Curr Gastroenterol Rep.
2013;15(9):325.
6. Woodfield CA, et al. Diagnosis of primary versus secondary achalasia: reassessment of clinical
and radiographic criteria. AJR Am J Roentgenol. 2000;175(3):727–31.
7. Gupta P, et al. Primary versus secondary achalasia: new signs on barium esophagogram. Indian
J Radiol Imaging. 2015;25(3):288–95.
8. Akaishi T, et al. Clinical usefulness of endoscopy, barium fluoroscopy, and chest computed
tomography for the correct diagnosis of achalasia. Intern Med. 2020;59(3):323–8.
9. Rabushka LS, Fishman EK, Kuhlman JE. CT evaluation of achalasia. J Comput Assist Tomogr.
1991;15(3):434–9.
10. Kahrilas PJ, Kim HC, Pandolfino JE. Approaches to the diagnosis and grading of hiatal hernia.
Best Pract Res Clin Gastroenterol. 2008;22(4):601–16.
11. Cakmakci E, et al. Accuracy of ultrasonography in the diagnosis of sliding hiatal hernias. Acad
Radiol. 2013;20(4):453–6.
12. Chaturvedi A, et al. Imaging of thoracic hernias: types and complications. Insights Imaging.
2018;9(6):989–1005.
13. Levine MS. Radiologic imaging of gastroesophageal reflux disease. Berlin: Springer.
p. 235–53.
14. Adanir H, et al. Endoscopic findings of gastro-esophageal reflux disease in elderly and younger
age groups. Front Med (Lausanne). 2021;8:606205.
15. Berkovich GY, Levine MS, Miller WT. CT findings in patients with esophagitis. AJR Am J
Roentgenol. 2000;175(5):1431–4.
16. Liang H, Fan JH, Qiao YL. Epidemiology, etiology, and prevention of esophageal squamous
cell carcinoma in China. Cancer Biol Med. 2017;14(1):33–41.
17. Charles TR, Jobe BA, Hunter JG, et al. Esophageal cancer: principles and practice. New York:
Demos Medical.
18. Malik TF, Gnanapandithan K, Singh K. Peptic ulcer disease. In: StatPearls [Internet]. Treasure
Island, FL: StatPearls; 2022. https://www.ncbi.nlm.nih.gov/books/NBK534792/. Accessed 29
Jul 2021.
19. Snowden FM. Emerging and reemerging diseases: a historical perspective. Immunol Rev.
2008;225:9–26.
20. Baghdanian AH, et al. Imaging manifestations of peptic ulcer disease on computed tomogra-
phy. Semin Ultrasound CT MR. 2018;39(2):183–92.
21. Guniganti P, et al. CT of gastric emergencies. Radiographics. 2015;35(7):1909–21.
22. Moysidis M, et al. The challenging diagnosis and treatment of duodenal diverticulum perfora-
tion: a report of two cases. BMC Gastroenterol. 2020;20(1):5.
23. Castilho Netto JM, Speranzini MB. Ampullary duodenal diverticulum and cholangitis. Sao
Paulo Med J. 2003;121(4):173–5.
24. Tsitouridis I, et al. MR cholangiography in the evaluation of patients with duodenal periampul-
lary diverticulum. Eur J Radiol. 2003;47(2):154–60.
25. Bittle MM, et al. Imaging of duodenal diverticula and their complications. Curr Probl Diagn
Radiol. 2012;41(1):20–9.
26. Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and preven-
tion. Prz Gastroenterol. 2019;14(1):26–38.
234 D. Caruso et al.
Population aging is taking place throughout the world, and about 13% of the 76 mil-
lion persons in the USA were aged 65 years and older [1]. In Europe, there is the
oldest population in the world, with almost 25% of European projected to be aged
65 years or older by 2030. In particular, Italy and Germany are estimated to have the
oldest population in Europe. The progressive increase in the proportion of a popula-
tion that is elderly depends on changes in the survival of older persons and in the
birth rate [1]. The increasingly greater life expectancy of the population has been
mainly determined by reduced mortality at older ages. The five leading causes of
death, including heart disease, cancer, stroke, chronic lower respiratory tract dis-
ease, and Alzheimer’s disease, account for 69.5% of all death [1]. The renal causes
of death account only for 2% of all deaths and for 4% of chronic conditions in per-
sons aged >65 years, even though these represent an important cause of disability
and comorbidity in older patients.
Due to the progressive increase in the mean age of the population, it is very
important to know the morphologic changes of the kidney according to aging. As a
matter of fact, older individuals, often with a compromised renal reserve and sub-
stantial comorbidities, are the norm in the hospitalized population [2]. The func-
tional alterations of the aged kidney are characterized principally by a progressive
reduction of renal blood flow from about 600 to 300 mL/min/1.73 m2 and of glo-
merular filtration rate (GFR) from 130 to 60–80 mL/min. An accurate quantitation
of the GFR should always be performed in elderly patients before injection of iodin-
ated and gadolinium-based contrast agents. Moreover, when exposed to iodinated
E. Quaia (*)
Institute of Radiology, Padova University Hospital, Padova, Italy
Department of Medicine-DIMED, University of Padova, Padova, Italy
e-mail: [email protected]
F. Crimí
Institute of Radiology, Padova University Hospital, Padova, Italy
e-mail: [email protected]
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 235
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_10
236 E. Quaia and F. Crimí
afferent and efferent arterioles in the juxtamedullary glomeruli, whereas in the cor-
tical glomeruli the vessels become obliterated. The significance of these findings is
unclear, as it is difficult to separate out changes which may have been engendered
by hypertension and those due to aging alone [4].
BPH is an “age-dependent” disease with a reported prevalence in patients older
than 60 years of 50% that increases to 90% in patients older than 85 years. Half of
these patients show lower urinary tract symptoms and, therefore, should undergo
medical or surgical therapy. Imaging by US and MRI plays a fundamental role in
the detection of this condition and in the clinical decision-making for treatment.
Prostate cancer is the second most common cancer affecting men worldwide and the
risk of prostate cancer increases with age, it has been reported that the median age
of symptoms onset is 72 years. US was the most used technique to identify prostate
cancer but in the last years, thanks to the PI-RADS standardized reporting system,
MRI has progressively become the gold standard for the detection of prostate can-
cer. Prostatitis is also quite common in old men, especially chronic prostatitis, and
is pivotal to correctly identify this infection that in some cases can be misdiagnosed
as prostate cancer both at US and MRI.
Aging induces in the kidney a progressive, functional, and anatomic decay that does
not have a particular clinical impact. The fundamental alterations of renal morphol-
ogy in elderly patients include size reduction, parenchymal thickness reduction,
margin irregularities, and increased corticomedullary differentiation. In particular,
the most important morphologic alteration of the aged kidney is the volume reduc-
tion, approximately 20–30% in 80-year-old men, and a loss of weight that decreases
from 250–270 to 180–200 g after age 65 [5].
In elderly patients, the kidneys appear frequently reduced in their largest dimen-
sion (within the range of 9–9.5 cm) on grayscale US, with reduction in the renal
parenchymal thickness due to chronic reduction of the renal parenchymal perfusion
from nephroangiosclerosis. The renal capsule becomes thicker and there is an
increase of the renal sinus fatty tissue, in particular at the level of the renal hilum
(Fig. 10.1). Typically, there is evidence of irregular margins (Fig. 10.2a), frequently
with a pseudolobular appearance (Fig. 10.2b) and/or coexisting with renal paren-
chymal scars due to previous renal cortical infarctions (Fig. 10.2b). The corticome-
dullary differentiation appears usually increased due to the relative higher
echogenicity of the renal cortex compared to the medulla due to nephroangiosclero-
sis. Anyway, less frequently, the corticomedullary differentiation may also be
reduced. On color/power Doppler US, the renal peripheral vessels are usually not
visualized in the subcapsular renal parenchyma (Fig. 10.3).
238 E. Quaia and F. Crimí
a b
Fig. 10.2 (a, b) Different grades of renal margin irregularities. Longitudinal grayscale US scan.
(a) Reduction of the renal parenchymal thickness with renal contour irregularities (arrows) and
increased corticomedullary differentiation. (b) Diffuse renal margin irregularities also with evi-
dence of renal parenchymal scars (arrows) due to previous regional infarctions. The interposed
renal parenchyma presents a pseudolobar appearance
a b
Fig. 10.3 (a, b) Fundamental morphologic alterations of the kidney in the elderly patient. (a)
Grayscale US. Increased echogenicity of the renal parenchyma with reduced corticomedullary
differentiation. (b) Power Doppler US. Reduced renal parenchymal vascularization at the level of
the subcapsular region (arrows)
a b
Fig. 10.4 (a, b) Fundamental morphologic alterations of the kidney in the elderly patient.
Contrast-enhanced CT, excretory phase. Coronal reformation. Diffuse reduction of the renal corti-
cal thickness with overt focal irregularities of renal margins (arrows)
240 E. Quaia and F. Crimí
a b
Fig. 10.5 (a, b) Contrast-enhanced CT showing a large renal parenchymal retention cysts (a) and
renal sinus cysts (b)
10.2 Nephrosclerosis
Nephrosclerosis is the term used for the renal pathology associated with sclerosis of
renal arterioles and small arteries due to medial and intimal thickening as a response
to hemodynamic changes, aging, genetic defects, or some combination of these and
to hyaline deposition in arterioles [7]. The resultant effect is focal ischemia of the
renal parenchyma supplied by vessels with thickened walls and consequent nar-
rowed lumen [7]. Some degree of nephrosclerosis is present at autopsy with increas-
ing age preceding or in the absence of hypertension. Hypertension and diabetes
mellitus increase the incidence and severity of the lesions. In nephrosclerosis, there
is a general hardening of the kidney due to overgrowth and contraction of interstitial
connective tissue. Nephrosclerosis may be compared to the arteriosclerosis of the
small renal arteries, and it is due to renovascular disease, mainly chronic hyperten-
sion. The renal vascular alterations of hypertension depend on the severity of the
blood pressure elevation and whether the process accelerates to malignant hyperten-
sion. Arteriolosclerosis of small cortical renal arteries, interlobar, arcuate, and inter-
lobular is a common feature of the kidney in patients with systemic arterial
hypertension and particularly in elderly patients [8].
In nephroangiosclerosis, both kidneys are usually symmetrically reduced in their
diameters with cortical scars. Cortical echogenicity is increased with increased or
reduced corticomedullary differentiation according to the grade of echogenicity of
the renal medulla. Color and power Doppler US reveal nonspecific reduction of
vascularization (Fig. 10.6). If compared to the younger population, renal RIs are
typically increased (>0.7 and frequently around 0.8) (Fig. 10.7). Renal perforating
arteries and veins [9] are much more visible in the kidneys of nephroangiosclerotic
patients in comparison with normal subjects since they enlarge in nephroangioscle-
rosis and present normally directed flows from the kidney toward the renal capsule.
10 The Male Urogenital System in Geriatric Patients 241
a b
c d
Fig. 10.6 (a–d) Fundamental morphologic alterations of the kidney in the elderly patient.
Reduction of the renal parenchymal thickness, margin irregularities, and increased corticomedul-
lary differentiation are evident on grayscale US (a). (b) Color Doppler US, longitudinal scan.
Reduction of renal cortical vascularization. (c, d) Doppler interrogation of the intrarenal segmental
arteries with increased arterial resistive indices
Fig. 10.7 Fundamental morphologic alterations of the kidney in the elderly patient. Color Doppler
US with Doppler interrogation of renal segmental artery. Increased arterial resistive index mea-
sured at the level of one renal segmental artery of the lower renal pole
242 E. Quaia and F. Crimí
The geriatric population is affected by many vascular diseases since the incidence
of atherosclerosis increases with age. Generally, the imaging of vascular diseases
in the elderly is complicated by the presence of coexisting diseases, while the
image quality is degraded due to obesity and limited patient compliance.
Renovascular hypertension accounts for 0.5–5% of patients who have hyperten-
sion. The renovascular disease may manifest as asymptomatic renal artery steno-
sis, intractable or uncontrollable hypertension requiring multiple medications, or
ischemic nephropathy with progressive loss of renal function [10]. In young
patients, the most common cause of renovascular hypertension is fibromuscular
dysplasia, while in the elderly the most common cause is atherosclerosis mainly
localized in the ostial or proximal tract of the renal artery. Color Doppler US, heli-
cal computed tomographic (CT) angiography, angiotensin converting enzyme
(ACE) inhibitor scintigraphy with captopril, and magnetic resonance (MR) angiog-
raphy have been assessed in the diagnosis of renal artery stenosis. Digital subtrac-
tion angiography remains the gold standard for the diagnosis of renal artery
stenosis, and it is part of any endovascular intervention. Contrast-enhanced CT and
MR imaging angiographic techniques have improved in their detection of renal
artery stenosis, and MR angiography is generally considered more sensitive for
renal artery stenosis than US [11]. Anyway, kidney disease limits the use of con-
trast agents during CT and MR imaging examinations, and this is particularly true
in elderly patients. Moreover, coexistent cardiopulmonary diseases, such as con-
gestive heart failure, arrhythmias, and chronic obstructive lung diseases, limit the
ability of the elderly to hold breath during image acquisition [10]. Additionally,
cardiopulmonary diseases may preclude the use of some of the imaging modalities
because of the inherent contraindications, such as pacemakers in MR examina-
tions. Consequently, color Doppler US is the principal imaging technique employed
in the elderly for renovascular disease diagnosis.
The velocimetric analysis of Doppler trace derived from renal arteries is of pri-
mary importance to identify renal artery stenosis. Direct Doppler criteria have been
proposed for the detection of renal artery stenosis, including an increased peak sys-
tolic velocity (>150–180 cm/s) (Fig. 10.8) and end-diastolic velocity at the level of
the stenosis [12, 13], a poststenotic flow disturbance resulting in spectral broaden-
ing and reversed flow [12], an increased ratio (≥3.5) of peak systolic velocity in the
renal artery and aorta (renal-aortic ratio), and the presence of turbulence within the
renal artery [14, 15]. Although this technique is easy to perform, its accuracy is
questionable because the lack of an early systolic peak has a low sensitivity for
10 The Male Urogenital System in Geriatric Patients 243
a b c
d e f
Fig. 10.8 (a–f) Renal artery stenosis in a 75-year-old male patient. Worsening renal function was
precipitated by the treatment of hypertension with angiotensin converting enzyme (ACE) inhibi-
tors. (a) Doppler interrogation revealed aliasing at the level of the proximal tract of the right renal
artery with spectral broadening of the Doppler trace and increase of the peak systolic velocity. (b)
“tardus et parvus” profile of the waveform at intrarenal arteries. (c, d) CT angiography (CTA).
Severe stenosis of the right renal artery is confirmed (arrow) and a moderate stenosis of the left
renal artery (arrowhead). (e, f) angiography confirmed the stenosis that was treated with a stent
components of the arterial waveform. This information allows to identify those con-
ditions, which may produce false-positive or false-negative results when the tardus
phenomenon is used to predict hemodynamically significant upstream stenosis [16].
This is the case of the loss of vascular compliance in severe diffuse atherosclerosis
of elderly patients, which may prevent the tardus-parvus phenomenon decreasing
the sensitivity of color Doppler US [13]. Other findings that may be observed in the
intraparenchymal arteries in the presence of renal artery stenosis are decreased
resistive indices in interlobar-arcuate renal cortical arteries with increased side dif-
ference higher than 10% [12].
Contrast-enhanced CT angiography (CTA) and MR imaging angiography
(MRA) are also very sensitive and specific for the demonstration of renal artery
occlusion. Additional views provided by CTA allow for display of the renal arteries
in multiple planes and projections, often necessary for the depiction of stenosis
(Fig. 10.8). Calcified plaques limit the CT evaluation of luminal narrowing. In par-
ticular, in cases with extensive calcification, as is frequently observed in elderly
patients, renal artery stenosis can be obscured by MIP technique and requires care-
ful evaluation of the volume-rendered images. CTA can also depict secondary signs
of renal artery stenosis, including poststenotic dilatation and renal parenchymal
changes of atrophy and decreased cortical enhancement. CTA is also very helpful in
the post-treatment evaluation of renal stent grafts and can usually delineate between
the highly attenuating graft material and the intraluminal contrast material.
MRA is well suited for the evaluation of renal artery stenosis in the elderly.
Calcified atheromatous plaques do not hamper the assessment of the arterial lumen.
MR angiography provides information about the size of the kidney, collateral ves-
sels, and poststenotic dilatation. Contrast-enhanced axial MR imaging can directly
show the narrowing of the stenosis, and reformatted multiplanar imaging is often
used. Both MIP and volume rendering are useful and complimentary in the evalua-
tion of renal artery stenosis. Axial images alone are not sufficient for the evaluation
of renal artery stenosis because the renal arteries often have a tortuous course, espe-
cially in elderly patients. Multiplanar reformations are very useful, in particular to
show renal artery occlusion.
Nontraumatic acute renal infarction is quite common in elderly patients, and it may
present the same symptoms of stone colic or acute pyelonephritis. Renal infarction
may be caused by tight stenosis or occlusion of segmental or of the main renal artery
or by renal artery embolization due to renal angioplasty, atrial fibrillation, and car-
diac valvular defects. Other causes of renal infarction are vasculitis, systemic lupus
erythematosus, drug-induced vasculitis, paraneoplastic syndrome, hypercoagulable
state, or acute venous occlusion [19]. Both CT and angiography are reference imag-
ing techniques in renal infarct detection, whereas US presents a lower sensitivity.
Even though large renal infarcts may be hypoechoic in comparison with the viable
renal parenchyma, segmental renal infarcts are usually isoechoic or rarely hyper-
echoic if hemorrhagic component is present.
10 The Male Urogenital System in Geriatric Patients 245
Renal infarcts often reveal a wedge shape with capsular base. Even though base-
line color Doppler US and power Doppler US present overt limitations to detect
renal perfusion defects due to the low sensitivity to low-velocity and low-amplitude
flow states, they may increase diagnostic capabilities of US in detecting renal
infarcts, especially in elderly or obese patients and in patients with renal diseases.
In renal infarct, color Doppler US and power Doppler US reveal absolute absence
of renal cortical flows, even though it is very difficult to differentiate renal segmen-
tal infarct from areas which appear poorly perfused due to underlying parenchymal
disease, deep renal position, and artifacts. Moreover, color Doppler US presents a
low accuracy in the detection of small renal infarcts in the subcapsular region for
limited spatial resolution and in the superior renal pole for the high Doppler angle
and for the depth position [20].
Recent advances in microbubble-based contrast agents, and dedicated contrast-
specific modes, have determined the achievement of increased image contrast in
tissues. By transmitting at the fundamental frequency and receiving selectively har-
monic frequencies, the background signal from stationary tissues is markedly sup-
pressed resulting in a greater signal-to-noise ratio and a better visibility of renal
infarcts. Blooming and flash artifacts are eliminated, shadowing artifacts are less-
ened, both spatial and temporal resolutions are improved, and the brightness of
grayscale pixel does not depend on angle-dependent frequency shift estimates.
Differently from iodinated contrast agent and gadolinium-based contrast agents,
microbubbles are pure intravascular agents which are not excreted in renal tubules
and may be safely employed in patients with advanced chronic renal failure, which
is frequently observed in the elderly. Microbubble-based contrast agents and
contrast-specific imaging techniques improve significantly the diagnostic confi-
dence level in identifying nonperfused renal parenchymal zones and allow a reliable
depiction of renal perfusion defects (Fig. 10.9). Renal perfusion defects due to renal
parenchymal infarction appear as single or multiple focal wedge-shaped areas of
absent, diminished, or delayed contrast enhancement in comparison to the adjacent
renal parenchyma after microbubble injection [21].
a b
Fig. 10.9 (a) Contrast-enhanced US after sulfur hexafluoride-filled microbubble injection. (b)
Contrast-enhanced CTA, corticomedullary phase. The left kidney shows partial parenchymal
infarction (arrow) in a 72-year-old woman with atrial fibrillation
246 E. Quaia and F. Crimí
a b
Fig. 10.10 (a, b) Renal artery thrombosis in a 75-year-old male patient. (a) Contrast-enhanced
CTA. Coronal reformation. The left renal artery is occluded by a complex thrombus (arrow) with
relative avascularity of the left kidney. (b) Contrast-enhanced CT. Corticomedullary phase shows
a complete renal infarction
a b
Fig. 10.12 (a, b) Contrast-enhanced CT. Multiple renal parenchymal perfusion defects (arrows)
due to diffuse septic embolization are evident on both kidneys of an 85-year-old patient
248 E. Quaia and F. Crimí
a b
c d
e f
Fig. 10.13 (a–h) Renal artery embolization in an 82-year-old male patient presenting at the emer-
gency unit with acute flank pain on the right side. (a–d) Contrast-enhanced US after sulfur
hexafluoride-filled microbubble injection. (e–h) Contrast-enhanced CT, nephrographic phase.
Multiple bilateral renal parenchymal perfusion defects (arrows), involving mainly the right kidney,
due to embolization of an ulcerated plaque of the thoracic aorta
10 The Male Urogenital System in Geriatric Patients 249
g h
Renal vein thrombosis in elderly patients, as in adults and differently from infants,
is typically of insidious onset and is almost always overimposed on an established
disease [22]. Causes of renal vein thrombosis in elderly patients include idiopathic
nephrotic syndrome, especially that due to membranous glomerulonephritis, vol-
ume loss due to dehydration (often aggravated by diuretic therapy) with altered
renal blood flow, hypercoagulable states (malignancy), renal cell carcinoma, or
extrinsic compression of the renal vein (retroperitoneal fibrosis, lymphoma, etc.).
The process may progress without any clinical sign. Mild abdominal or back pain
may be present, but severe pain is uncommon. Pulmonary emboli occur during the
course of approximately 50% of patients with chronic renal vein thrombosis and are
frequently the first manifestation of this condition [22].
Diagnosis of renal vein thrombosis relies on the visualization of an echogenic thrombus
within a dilated renal vein devoid of flow signals on CD corticomedullary differentiation
on grayscale US. Doppler spectral analysis of renal arteries may reveal slightly increased
RIs and normal parenchymal venous flows, since collateral venous supplies open after
renal vein thrombosis. Absent or reversed end-diastolic flow in renal interlobar–arcuate
arteries has been described in transplanted kidney which lacks collateral venous supply.
US contrast agents facilitate identification of renal vein patency and thrombosis in cases of
technical failure and enhance detection of collateral venous blood supply. A mass is evi-
dent in the renal vein with renal enlargement and delayed renal function.
CTA and MRA show complete occlusion of the renal vein [19] which appears
dilated and heterogeneous, while the infracted kidney appears enlarged and with a
diffuse alteration of the nephrographic phase (Fig. 10.14). Renal vein involvement
by tumor (Fig. 10.15) is frequently identified in elderly patients and it is crucial in
the determination of surgical options for removing a renal tumor. The renal veins
are well depicted on CT during the corticomedullary or nephrographic phase of
contrast enhancement.
250 E. Quaia and F. Crimí
a b
Fig. 10.15 (a, b) Thrombosis of the left renal vein due to infiltrating papillary renal cell carci-
noma. (a) Unenhanced CT. (b) Contrast-enhanced CT. Contrast-enhanced CT. Nephrographic
phase. The renal vein appears dilated and heterogeneous (small arrow), while the left kidney (large
arrow) appears enlarged and with diffuse alteration of the nephrographic phase. The inferior vena
cava (IVC) is also involved and appears occluded by a tumoral thrombus
In the elderly, the kidneys are more vulnerable when other pathologies occur, in
particular, atherosclerosis, arterial hypertension, diabetes mellitus, bacterial infec-
tions, and malnutrition. Most cases of acute renal failure in elderly patients are
caused by drugs or are secondary to dehydration, especially in patients with hyper-
tensive intrarenal nephrosclerosis. In elderly patients, the differentiation between
renal and prerenal cause of acute renal failure may be difficult because the RIs are
usually elevated for the preexisting renal parenchymal disease. Moreover, an elderly
patient with severe and prolonged prerenal acute renal failure leading to acute tubu-
lar necrosis may present increased RIs.
10 The Male Urogenital System in Geriatric Patients 251
a b
Fig. 10.16 (a–c) Renal acute cortical necrosis. An 80-year-old patient with aortic endoprosthesis
was admitted to the emergency unit with acute renal failure. The absence of contrast enhancement
in the superficial cortex of the left kidney (arrow) is identified after microbubble injection (a). (b,
c) Contrast-enhanced CT confirmed the existence of diffuse renal cortical necrosis in the left kid-
ney (arrow)
a b
c d
Fig. 10.17 (a–d) Cholesteric renal embolization in a 70-year-old female patient presenting with
acute renal failure. Baseline color Doppler US (a, b) does not allow the identification of renal
perfusion defects. Contrast-enhanced US (c, d) allows a reliable depiction of renal perfusion
defect (arrow)
10 The Male Urogenital System in Geriatric Patients 253
renal infarcts in every old patient presenting with a renal colic-like pain in the
flank region.
The proportion of elderly individuals is growing rapidly in all societies, and the
incidence of chronic kidney disease among elderly people increases constantly [28].
Therefore, the accurate monitoring of kidney function, that is GFR, in elderly peo-
ple is of considerable clinical interest in order to detect individuals who are at risk
for developing chronic kidney disease. The management of end-stage renal failure
in the elderly should not be significantly different from that in younger patients and
should be based on the capacity for rehabilitation.
Chronic kidney disease is an important problem in the elderly and is associated
with a high risk of kidney failure, cardiovascular disease, and death [29]. The disor-
der is indicated either by a GFR of less than 60 mL/min/1.73 m2 of body surface
area or by the presence of kidney damage, assessed most commonly by the finding
of albuminuria for 3 or more consecutive months [30–32]. In persons 70 years of
age or older, the percentage of people with a chronic kidney disease is around
30% [29].
Risk factors for chronic kidney disease include an age of more than 60 years,
hypertension, diabetes, cardiovascular disease, and a family history of the disease.
According to a recent series, diabetic nephropathy, obstructive uropathy, and hyper-
tensive nephrosclerosis were the major causes of chronic renal failure and accounted
for 80% of total chronic renal failure in the elderly [33].
Recommendations for evaluating people at increased risk are to measure urine
albumin to assess kidney damage and to estimate the GFR with an equation based
on the level of serum creatinine [32]. Older adults who suffer an acute injury to the
kidneys—from trauma, surgery, or illness—are at dramatically increased risk of
later end-stage renal disease.
Special care should be used in patients with chronic renal failure when the IV
injection of iodinated or gadolinium-based agents is planned. Iodinated contrast
agents should be employed in patients with chronic renal failure only before and
after proper hydration, while gadolinium-based contrast agents should not be
employed in patients with a GFR value below 30 mL/min. Differently from iodin-
ated contrast agent and gadolinium-based contrast agents, microbubbles may be
safely employed in patients with advanced chronic renal failure, especially in the
evaluation of renal masses and perfusion defects.
US reveals reduced renal length and cortical thickness and a hyperechoic renal
parenchyma with a poor visibility of renal pyramids and of renal sinus. Doppler US
reveals a reduced parenchymal perfusion and increased resistive index (RI) values.
In elderly patients with mild chronic renal failure, acute renal failure represents a
254 E. Quaia and F. Crimí
In elderly patients, acute urinary tract obstruction can occur anywhere in the urinary
tract from the renal papilla to the urethral meatus and may be determined by a plenty
of causes. As in some patients, obstruction may be completely asymptomatic even
though, most frequently, it manifests with clear clinical symptoms.
Hydronephrosis may also be absent in the acute obstruction of the urinary tract
principally due to hypovolemia, dehydration, or nephrosclerosis. The most impor-
tant causes of urinary tract obstruction in elderly patients are urinary stones, tumors
of the urinary tract and ureter, and benign prostatic hyperplasia (Fig. 10.18).
The obstruction of the urinary tract, if not treated, usually determines a progres-
sive atrophy of the renal parenchyma which is frequently observed in elderly
patients.
Chronic obstructive uropathy (Fig. 10.19) may be determined by the tumoral
infiltration of the ureteral wall or by chronic incomplete obstruction of the ureter,
which may be suddenly complicated by an acute event such as infection.
10 The Male Urogenital System in Geriatric Patients 255
a b
Fig. 10.19 (a, b) Contrast-enhanced CT, excretory phase. Chronic urinary tract obstruction of the
right kidney (arrow) due to tissue scarring of the lower ureter. The kidney appears small and with-
out any sign of function (contrast excretion). Perirenal strands with dilatation and wall thickening
of the renal pelvis are also evident on the right kidney
256 E. Quaia and F. Crimí
a b
Fig. 10.20 Pyelonephritis with diffuse abscessual evolution in a 70-year-old diabetic woman pre-
senting with septic shock (a) Grayscale US. Longitudinal scan. The left kidney appears increased
in dimension with multiple cystic lesions (arrows). (b) Contrast-enhanced CT during the nephro-
graphic phase after iodinated contrast injection. Both kidneys appear involved by multiple absces-
sual lesions (arrows). (c) Gross autopsy specimen confirming multiple renal abscesses (arrows)
10 The Male Urogenital System in Geriatric Patients 257
a b
c d
e f
Fig. 10.21 (a–f) Pyonephrosis in an 82-year-old woman presenting with acute right flank pain.
Grayscale US, longitudinal (a) and transverse scan (b). The right kidney presents dilatation of the
intrarenal urinary tract (white arrow) with diffuse corpuscular echogenic content and evidence of
renal stones (black arrow) lying in the renal pelvis with posterior acoustic shadowing. (c–f) Contrast-
enhanced CT, nephrographic phase. The right kidney (large arrow) presents increased dimensions,
multiple renal stones lying in the renal pelvis, and dilatation and diffuse thickening of the renal pel-
vis. Renal parenchyma presents also some abscesses (small arrows) due to infection diffusion
258 E. Quaia and F. Crimí
Frequently, urologists are confronted with an elderly patient (≥75 years of age) with
a renal mass seeking treatment. As the population ages, comorbidities become more
confounding in predicting patient outcome to therapy and may influence the appli-
cation of surgical therapy with curative intent to elderly patients [35]. Epidemiological
studies show an increasing incidence of renal cell carcinoma over the past two
decades, and interestingly, this increase has included a larger proportion of elderly
people. The presentation of renal cancer has evolved. There has been an increase in
the incidence of cases in the USA and several European countries and, at the same
time, a shift to incidentally diagnosed, smaller, localized tumors in a slightly older
population [36].
Generally, in elderly patients there is an increase in neoplastic disorders includ-
ing clear cell-type renal carcinoma and transitional cell carcinoma (TCC). The
median age of presentation of renal cell carcinoma is in the sixth decade of life.
Conversely, transitional cell carcinoma of the upper urinary tract is commonly seen
in older patients, usually between the sixth and eighth decade of life. This increased
incidence is mainly due to the more widespread use of imaging technology [37].
Most renal tumors are completely asymptomatic and are found incidentally in
elderly patients during imaging of the upper abdomen mainly by US (Fig. 10.22).
There is a great variance of growth rate with the majority of small renal tumors
(≤3 cm in diameter) in the elderly, with a prevalence of low growth rate (0.35 cm/
year with a median range of 0–10 cm), and a low incidence of distant metastases
[38]. Conversely, the majority of larger renal tumors usually present local invasive-
ness (Figs. 10.23 and 10.24) and distant metastases (Fig. 10.25). A “wait and see”
observational approach for renal masses 1.5 cm or smaller in the elderly can be
suggested [39].
TCCs are relatively rare tumors of the kidney, while they are commonly seen in
older patients usually between the sixth and eighth decade of life with a mean age
a b
Fig. 10.22 (a, b) Small renal tumor incidentally found in a 75-year-old male patient during US
examination of the abdomen. Unenhanced CT scan (a) showed a solid exophytic mass (arrow)
with enhancement after contrast injection in arterial phase (b). Clear cell-type renal cell carcinoma
is identified after partial nephrectomy
10 The Male Urogenital System in Geriatric Patients 259
a b
c d
Fig. 10.23 (a–d) Clear cell-type renal cell carcinoma in a 75-year-old man with hematuria. Local
tumoral invasiveness. (a) Grayscale US. A solid renal mass (arrow) is identified on the right kid-
ney. (b, c) Contrast-enhanced CT. Nephrographic phase shows a renal mass (arrow) on the right
kidney with invasion of the renal pelvis. (d) Photograph of gross specimen. Evidence of invasion
of the renal pelvis which justified the presenting symptom hematuria
of 65 years. TCCs of the renal pelvis or calices present an incidence of 5–15% of all
malignant tumors of the kidney [40, 41]. The incidence in men exceeds that in
women and the usual sex ratio is between 2:1 and 4:1 [41]. Over 85–90% of upper
urinary tract tumors are TCCs, with the renal pelvis (Fig. 10.26) being more com-
monly involved than the ureter [42]. Renal lymphoma occurs in all age groups, even
though the disease usually affects adults (average age, 60 years) and frequently
elderly patients. Renal involvement with lymphoma occurs much more commonly
with non-Hodgkin disease, the majority of patients having intermediate- or
260 E. Quaia and F. Crimí
a b
Fig. 10.24 (a, b) Clear cell-type renal cell carcinoma in an 82-year-old man. Local tumoral inva-
siveness. Contrast-enhanced CT. (a) Transverse plane. (b) Sagittal plane. Corticomedullary phase
shows a large solid renal mass of the right kidney invading the adjacent liver parenchyma (arrow)
high-grade lymphomas including Burkitt and histiocytic types [43]. Lymphoma that
is isolated to the kidney as a primary site of involvement is quite rare, whereas addi-
tional sites of extranodal involvement are common and are seen in most patients at
the time of diagnosis. Lymphoma typically involves the kidney in one of the several
recognizable patterns including multiple renal masses, solitary masses, diffuse renal
infiltration, renal invasion from contiguous retroperitoneal disease (Fig. 10.27),
perirenal disease, or atypical patterns of renal involvement with invasion of the
renal pelvis.
10 The Male Urogenital System in Geriatric Patients 261
a b
c d
Fig. 10.25 (a–d) Clear cell-type renal cell carcinoma in a 77-year-old man. (a) Contrast-enhanced
CT. Nephrographic phase shows a heterogeneous large renal mass on the lower pole of the right
kidney. (a, b) Multiple enlarged lymph nodes (small white arrow) are identified in the retrocaval
nodal site. (c) Floating thrombus (large white arrow) in the inferior vena cava is also present. (d)
Distant bone metastasis is visualized on the right acetabulum (large arrow)
262 E. Quaia and F. Crimí
a b
Fig. 10.26 (a, b) Transitional renal cell carcinoma in a 70-year-old man with hematuria. (a)
Contrast-enhanced CT. Coronal (a) and sagittal reformations (b). A solid endoluminal tumor
(arrow) in the left kidney pelvis
a b
Fig. 10.27 (a, b) Renal lymphoma in a 67-year-old male patient with a known non-Hodgkin
disease retroperitoneal disease. (a) Contrast-enhanced CT, transverse plane. Direct and extensive
renal parenchymal invasion from contiguous retroperitoneal disease. (b) Photograph of gross spec-
imen from autopsy. Gross pathologic examination reveals yellow/gray tumor with extensive renal
parenchymal invasion
10 The Male Urogenital System in Geriatric Patients 263
10.11 Prostate
a b
Fig. 10.28 (a, b) Benign prostatic hyperplasia (BPH) at ultrasound in sagittal (a) and transverse
plane (b). The calculated volume of the prostate is 65 cm3. Multiple isoechoic nodules on a
hypoechoic background are detected in the transition zone
a b
Fig. 10.29 (a, b) Benign prostatic hyperplasia (BPH) at MRI in transverse (a) and sagittal plane
(b). The calculated volume of the prostate is 52 cm3. Multiple typical encapsulated nodules of the
BPH can be appreciated in the transition zone
10 The Male Urogenital System in Geriatric Patients 265
Prostate cancer is the second most common cancer affecting men worldwide, with
an estimated number in 2020 of 1,400,000 new cases and 375,000 deaths [54]. The
vast majority of tumors arising from the prostate are carcinomas of epithelial origin
[55]. Prostate cancer originates mainly from the peripheral zone that is located pos-
teriorly and constitutes the main glandular component, nevertheless among one-
fourth of the tumors originate from the transition zone that is located more anteriorly
[56]. The risk of prostate cancer increases with age: foci of prostate cancer have
been identified in 30–40% of men aged 60 years or older and the median age of
symptoms onset is 72 years [57, 58]. Hence, it is fundamental to have screening
procedures performed, especially in elderly patients. The screening tests are two:
the first is the physical examination with the digital rectal examination (DRE) and
the second one is the serum prostate-specific antigen (PSA) measurement that is
more reliable and widely used [59]. Anomalous findings in DRE or increased PSA
levels bring the suspect of prostate cancer and, therefore, imaging and eventually
biopsies are required to confirm the diagnosis. Since a few years ago, endorectal US
was the main imaging modality to verify the presence of prostate cancer [60]. At US
prostate cancer in around 60–70% of cases is detected as a hypoechoic nodule in the
gland, although the remaining 30–40% are iso or hyperechoic; anyway, the reported
accuracy of this technique is low, around 50–60% [60]. The accuracy of the tran-
srectal US has been reported to improve with the use of color/power Doppler tech-
niques, with the use of microbubble contrast agents and with elastography [60].
Nevertheless, in the last 10 years MRI has increasingly been used for the detection
of prostate cancer and its risk stratification. The first standardized system to report
MRI results was published in 2013, the Prostate Imaging Reporting and Data
System (PI-RADS), that was later updated in 2015 and now is currently used with
the 2.1 version published in 2019 [61–63]. The PI-RADS system assigns a score to
the lesions found by MRI in the prostate from PI-RADS 1 (very low risk of prostate
cancer) to PI-RADS 5 (very high risk of prostate cancer). The MRI characterization
of the lesion changes in the peripheral zone compared to the transition zone, in the
first one is mainly based on diffusion weighted imaging (DWI) signal of the nodule,
while in the second one on T2-weighed signal of the lesion [63]. For the peripheral
zone, the characteristics of the lesions are the following that are based mainly on
diffusion weighted imaging (DWI): PI-RADS 1 normal signal in DWI without
alteration at the apparent diffusion coefficient (ADC) map; PI-RADS 2 mild hypoin-
tensity in the ADC map but not focal hyperintensity in high b value DWI images;
PI-RADS 3 focal mild or moderate hypointensity in ADC map and mild hyperinten-
sity in high b value DWI images; PI-RADS 4 focal and marked hypointensity on the
ADC map with marked hyperintensity in high b value DWI images. <1.5 cm;
PI-RADS 5 same as 4 but ≥1.5 cm in greatest dimension or definite extraprostatic
extension/invasive behavior [63]. The contrast enhancement pattern plays a role in
this scale since PI-RADS 3 lesions with hypervascularization in arterial phase
should be classified as PI-RADS 4 [63] (Fig. 10.30).
266 E. Quaia and F. Crimí
a b
c d
Fig. 10.30 (a–d) Prostate MRI in a 72 years-old patient with PSA elevation. (a) T2-weighted
image showing a hypointense lesion (arrow) of the peripheral zone with a maximum diameter of
14 mm and without extracapsular extension; (b) focal and marked hypointensity of the lesion on
the ADC map (arrow); (c) marked hyperintensity of the lesion (arrow) in 1500 b value DWI
images; (d) contrast enhancement of the nodule after contrast injection (arrow). The lesion was
scored as a PI-RADS 4 nodule and targeted biopsies revealed a Gleason 7 prostate
adenocarcinoma
In the transition zone, the grading is based on the T2-wighted signal of the lesion:
PI-RADS 1 normal appearing transition zone (rare) or a round, completely encap-
sulated nodule; PI-RADS 2 a mostly encapsulated nodule or a homogeneous cir-
cumscribed nodule without encapsulation (“atypical nodule”), or a homogeneous
mildly hypointense area between nodules; PI-RADS 3 heterogeneous signal inten-
sity with obscured margins; includes others that do not qualify as PI-RADS 2, 4, or
5; PI-RADS 4 lenticular or non-circumscribed, homogeneous, moderately hypoin-
tense, and <1.5 cm in greatest dimension; PI-RADS 5 same as 4 but ≥1.5 cm in
greatest dimension or definite extraprostatic extension/invasive behavior [63]. If a
10 The Male Urogenital System in Geriatric Patients 267
PI-RADS 3 lesion ≥1.5 cm shows marked hypointensity on the ADC map and
marked hyperintensity in high b value DWI images, it should be classified as
PI-RADS 4 [63].
The PI-RADS system divides the prostate into 39 sectors/regions for three parts
of the prostate, the apex, the mid prostate, and the base of the prostate [63].
Multi-parametric MRI results allow to identify the suspicious lesions in the pros-
tate and to target the biopsies on them [64]. There are three techniques of MRI guid-
ance to perform a targeted prostate biopsy: the first is the cognitive fusion in which
the US operator performs the biopsy aiming in the prostate area where the MRI
results showed a lesion, the second is the direct MRI-guided biopsy that is a biopsy
performed directly in the MRI tube and guided by the images acquired during the
procedure and finally using device for images fusion where there is a co-registration
of stored MR images with real-time US scan [64].
MRI and contrast-enhanced CT can also identify loco-regional lymph node
metastases, at MRI or CT nodes with a short axis ≥1 cm, or ≥0.8 cm if round
shaped, are suspect for metastatic involvement [65]. A better accuracy for nodal
metastases detection has been reported for choline-PET/CT and prostate-specific
membrane antigen (PSMA)-PET/CT compared to CT and MRI [66].
For distant metastases contrast-enhanced CT is routinely used but MRI, choline-
PET/CT, PSMA-PET, and NaF-PET/CT showed a better accuracy, especially in
detection of bone metastases [66].
10.11.3 Prostatitis
References
1. Ferrucci L, Giallauria F, Guralnik JM. Epidemiology of aging. Radiol Clin North Am.
2008;46:643–52.
2. Dagher PR, Herget-Rosenthal S, Ruehm SG, et al. Newly developed techniques to study and
diagnose acute renal failure. J Am Soc Nephrol. 2003;14:2188–98.
3. Faubert PF, Porush JG. Renal disease in the elderly. 2nd ed. Basel: Marcel Decker; 1998.
4. Davison AM. Renal disease in the elderly. Nephron. 1998;80:6–16.
5. Mulder WJ, Hillen HF. Renal function and renal disease in the elderly: part I. Eur J Intern Med.
2001;12(4):327–33.
6. Pozzi Mucelli R, Faccioli N, Manfredi R. Imaging findings of genitourinary tumors in the
elderly. Radiol Clin North Am. 2008;46:773–84.
7. Alpers CE. The kidney. In: Kumar V, Abbas AK, Fausto N, editors. Robbins and Cotran patho-
logic basis of disease. Philadelphia: Elsevier Saunders; 2005. p. 955–1021.
8. Quaia E, Bertolotto M. Renal parenchymal diseases: is characterization feasible with ultra-
sound? Eur Radiol. 2002;12:2006–20.
9. Bertolotto M, Quaia E, Galli G, et al. Color Doppler sonographic appearance of renal per-
forating vessels in subjects with normal and impaired renal function. J Clin Ultrasound.
2000;28:267–76.
10. Kalva SP, Mueller PR. Vascular imaging in the elderly. Radiol Clin North Am. 2008;46:663–83.
11. Zhang H, Prince MR. Renal MR angiography. Magn Reson Imaging Clin N Am.
2004;12:487–503.
12. Correas JM, Helenon O, Moreau JF. Contrast enhanced ultrasonography of native and trans-
plant kidney diseases. Eur Radiol. 1999;9(Suppl 3):394–400.
13. Grant EG, Melany ML. Ultrasound contrast agents in the evaluation of the renal arteries. In:
Goldberg BB, Raichlen JS, Forsberg F, editors. Ultrasound contrast agents. Basic principles
and clinical applications. 2nd ed. London: Martin Dunitz; 2001. p. 289–95.
14. Desberg AL, Paushter DM, Lammert GK, et al. Renal artery stenosis: evaluation with color
Doppler flow imaging. Radiology. 1990;177:749–53.
15. Helenon O, Rody EL, Correas JM, et al. Color Doppler US of renovascular disease in native
kidneys. Radiographics. 1995;15:833–54.
16. Bude RO, Rubin JM, Platt JF, et al. Pulsus tardus: its cause and potential limitations in detec-
tion of arterial stenosis. Radiology. 1994;190:779–84.
17. Bude RO, Rubin JM. Detection of renal artery stenosis with Doppler sonography: it is more
complicated than originally thought (editorial). Radiology. 1995;196:612–3.
18. Stavros AT, Parker SH, Yakes WF, et al. Segmental stenosis of the renal artery: pattern recogni-
tion of tardus and parvus abnormalities with duplex sonography. Radiology. 1992;184:487–92.
19. Kawashima A, Sandler CM, Ernst RD, et al. CT evaluation of renovascular disease.
Radiographics. 2000;20:1321–40.
20. Correas JM, Claudon M, Tranquart F, et al. Contrast-enhanced ultrasonography: renal applica-
tions. J Radiol. 2003;84:2041–54.
21. Bertolotto M, Martegani A, Aiani L, et al. Value of contrast-enhanced ultrasonography for
detecting renal infarcts proven by contrast-enhanced CT. A feasibility study. Eur Radiol.
2008;18(2):376–83.
22. Cohen JJ. Vascular disorders of the kidney. In: Wyngaarden JB, Smith LH, Bennett JC, editors.
Cecil textbook of medicine. Philadelphia: Saunders; 1992. p. 598–9.
23. Taylor GA, Ecklund K, Dunning PS. Renal cortical perfusion in rabbits: visualization
with color amplitude imaging and an experimental microbubble-based US contrast agent.
Radiology. 1996;201:125–9.
24. Coley BD, Mattrey RF, Roberts A, et al. Potential role of PFOB enhanced sonography of the
kidney. II. Detection of partial infarction. Kidney Int. 1991;39:740–5.
25. Pozzi Mucelli R, Bertolotto M, Quaia E. Imaging techniques in acute renal failure. In: Ronco
C, Bellomo R, La Greca G, editors. Blood purification in intensive care. Basel: Karger; 2001.
p. 76–91.
10 The Male Urogenital System in Geriatric Patients 269
26. Jordan J, Low R, Jeffrey RB. CT findings in acute renal cortical necrosis. J Comput Assist
Tomogr. 1990;14(1):155–6.
27. Cohan RH, Cowan NC, Ellis JH. Unknown ESUR cases 2004. Abdom Imaging.
2006;31:141–53.
28. Fliser D. Assessment of renal function in elderly patients. Curr Opin Nephrol Hypertens.
2008;17(6):604–8.
29. Stevens LA, Levey AS. Chronic kidney disease in the elderly—how to assess risk. N Engl J
Med. 2005;352:2122–4.
30. National Kidney Foundation. K/DOQI clinical practice guide-lines for chronic kidney disease:
evaluation, classification, and stratification. Am J Kidney Dis. 2002;39(Suppl 1):S1–S266.
31. Levey AS, Eckardt KU, Tsukamoto Y, et al. Definition and classification of chronic kidney dis-
ease: a position statement from kidney disease: improving global outcomes (KDIGO). Kidney
Int. 2005;67:2089–100.
32. Stevens LA, Coresh J, Greene T, et al. Assessing kidney function—measured and estimated
glomerular filtration rate. N Engl J Med. 2006;354:2473–83.
33. Prakash J, Saxena RK, Sharma OP. Spectrum of renal disease in the elderly: single center
experience from a developing country. Int Urol Nephrol. 2001;33(2):227–33.
34. Gleckman R, Blagg N, Hibert D, et al. Acute pyelonephritis in the elderly. South Med
J. 1982;75(5):551–4.
35. Berdjis N, Hakenberg OW, Novotny V, et al. Treating renal cancer in the elderly. BJU Int.
2006;97:703–5.
36. Linehan JA, Nguyen MM. Kidney cancer: the new landscape. Curr Opin Urol.
2009;19(2):133–7.
37. Jayson M, Sanders H. Increased incidence of serendipitously discovered renal cell carcinoma.
Urology. 1998;51:203–5.
38. Bosniak MA, Birnbaum BA, Krinsky GA, et al. Small renal parenchymal neoplasms: further
observations on growth. Radiology. 1995;197(3):589–97.
39. Silverman SG, Israel GM, Herts BR, et al. Management of the incidental renal mass. Radiology.
2008;249:16–31.
40. Grabstald H, Whitmore WF, Melamed MR. Renal pelvic tumors. JAMA. 1971;218:845–54.
41. Nocks BN, Heney NM, Daly JJ, et al. Transitional cell carcinoma of the renal pelvis. Urology.
1982;19:472–7.
42. Wong-You-Cheong JJ, Wagner BJ, Davis CJ. Transitional cell carcinoma of the urinary tract:
radiologic-pathologic correlation. Radiographics. 1998;18:123–42.
43. Urban BA, Fishman EK. Renal lymphoma: CT patterns with emphasis on helical
CT. Radiographics. 2000;20:197–212.
44. Wasserman NF. Benign prostatic hyperplasia: a review and ultrasound classification. Radiol
Clin North Am. 2006;44(5):689–710, viii. https://doi.org/10.1016/j.rcl.2006.07.005.
45. Guneyli S, Ward E, Peng Y, Nehal Yousuf A, Trilisky I, Westin C, Antic T, Oto A. MRI evalu-
ation of benign prostatic hyperplasia: correlation with international prostate symptom score.
J Magn Reson Imaging. 2017;45(3):917–25. Epub 2016 Aug 3. https://doi.org/10.1002/
jmri.25418.
46. Grossfeld GD, Coakley FV. Benign prostatic hyperplasia: clinical overview and value of diag-
nostic imaging. Radiol Clin North Am. 2000;38:31–47.
47. Park YJ, Bae KH, Jin BS, Jung HJ, Park JS. Is increased prostatic urethral angle related to
lower urinary tract symptoms in males with benign prostatic hyperplasia/lower urinary tract
symptoms? Korean J Urol. 2012;53:410–3.
48. Wasserman NF, Spilseth B, Golzarian J, Metzger GJ. Use of MRI for lobar classification of
benign prostatic hyperplasia: potential phenotypic biomarkers for research on treatment strate-
gies. AJR Am J Roentgenol. 2015;20:564–71.
49. McNeal JE. Normal histology of the prostate. Am J Surg Pathol. 1988;12:619–33.
50. Marks LS, Treiger B, Dorey FJ, et al. Morphology of the prostate: distribution of tissue com-
ponents in hyperplastic glands. Urology. 1996;44:486–92.
270 E. Quaia and F. Crimí
The female genital system includes the ovaries, fallopian tubes, uterus and cervix,
and vagina. The imaging appearance of the female genital system changes signifi-
cantly during a woman’s lifespan, reflecting the influence of hormones. After meno-
pause, hormonal levels diminish, leading to a progressive involution of the uterus,
cervix, ovaries, and vagina. The observed genital diseases also change in the elder-
lies, with increased incidence of neoplastic processes and of organ prolapse due to
laxity of the pelvic floor musculature and less frequent ovarian functional disor-
ders [1, 2].
Common indications for imaging the pelvis of a post-menopausal patient include
post-menopausal bleeding, pelvic pain or pressure, history of ovarian cysts, increas-
ing abdominal girth, or adnexal masses. Knowing the anatomy and normal imaging
appearance of the female pelvis in the post-menopausal woman is fundamental
since findings that can be normal in the reproductive years can be pathological when
M. A. Cova (*)
Department of Medicine, Surgery and Health Sciences, University of Trieste, Azienda
Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
Department of Radiology, Azienda Sanitaria Universitaria Giuliano Isontina, Cattinara
Hospital, Trieste, Italy
e-mail: [email protected]
L. Bottaro
Department of Radiology, Azienda Sanitaria Universitaria Giuliano Isontina, Cattinara
Hospital, Trieste, Italy
e-mail: [email protected]
C. Marrocchio · A. M. Bozzato
Department of Medicine, Surgery and Health Sciences, University of Trieste, Azienda
Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
e-mail: [email protected]; [email protected]
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 271
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_11
272 M. A. Cova et al.
a b
c d
Fig. 11.1 Normal anatomy. (a) Normal US anatomy of the ovaries after menopause, appearing
small and homogeneously hypoechoic due to the lack of follicles (between calipers). (b) MR
appearance of the involuted ovaries, which have a decreased volume and a homogeneous interme-
diate signal intensity on T2-weighted images (arrows). (c) US appearance of the uterus after meno-
pause, decreased in size (between calipers). (d) Sagittal T2-weighted imaging showing an involuted
uterus, decreased in dimension and with less defined anatomical layers
11.1.1 Ultrasound
approach does not allow an adequate assessment because of its smaller field of view,
e.g., in case of large uterine size or ovaries or other lesions located high in the pel-
vis. Transvaginal ultrasound (TVUS) is performed after bladder voiding and using
high-frequency probes. This approach has higher resolution in assessing the uterus,
the cervix, and the adnexa [3]. This is particularly true in older patients, in whom
decreased urinary bladder capacity, increased body habitus, and involution of the
organs to be studied may decrease the diagnostic accuracy of the transabdominal
approach [3].
The ovaries are generally identified by knowing their location with respect to the
uterus and recognizing the broad ligaments on whom posterior aspect they are
attached; however, they can be difficult to assess after menopause for their reduction
in volume and reduced number or absence of follicles [4]. The normal ovarian vol-
ume starts decreasing after 30 years, passing from 6.6 cm3 in women younger than
30 years to 2.6 cm3 in women 50–59 years old, and can continue decreasing during
menopause [5]. The mean post-menopausal volumes range from 1.2 to 5.8 cm3, and
a volume greater than 8 cm3 is always considered abnormal [3, 6, 7]. The post-
menopausal ovaries appear more hypoechoic and homogeneous because of the
fewer or absent follicles. Small echogenic foci, 1–3 mm in size, with no associated
soft-tissue component, may be recognized, generally at the periphery. These may be
related to dystrophic calcifications in atretic follicles, epithelial inclusion cysts, or
millimetric cysts causing reverberation artifacts [3]. The fallopian tubes are not nor-
mally seen unless abnormal or surrounded by fluid. When recognized, the normal
tubes appear as elongated echogenic structures, directed posterolaterally from the
uterine horns, with echogenic fingerlike projections (fimbriae), and about 10–12 cm
in length and 1–4 mm in diameter [8]. The appearance and the size of the uterus
vary depending on the woman’s age [3, 9]. The uterine size in young women ranges
from 5 to 9 cm, while it decreases after menopause, varying from 3.5 to 7.5 cm in
length and from 1.2 to 3.3 cm in the anteroposterior diameter. On TVUS, a hyper-
echoic thin endometrium and a myometrium with coarse, speckled echotexture can
often be seen [3]. Free peritoneal fluid, when small and simple, can be normal in
early menopause, but in late menopause its presence is always abnormal and can be
related to gynecological and non-gynecological diseases [3].
Magnetic resonance imaging (MRI) is a panoramic imaging modality with high con-
trast resolution. It provides an excellent assessment of the female pelvis as a second-
line imaging modality after US or as a primary imaging modality when US is not
feasible [3]. MRI can be performed on both 1.5 T and 3 T and pelvic phased array
coils are recommended at both 1.5 T and 3.0 T to increase signal-to-noise ratio (SNR),
with anterior and superior saturation bands. Antiperistaltic agents can be optionally
used to minimize artifact caused by bowel movement or contraction. The exam is usu-
ally performed with the patient in the supine position. The acquisition protocol will
depend on the specific pathology and organ to study; the standard protocol should
274 M. A. Cova et al.
collagen and elastic fibers, and the muscular layer, composed of smooth muscle
cells organized in an inner circular and outer longitudinal layer. Most externally,
there is the adventitia layer, in which a serpiginous T2-high-signal intensity can be
recognized, corresponding to the vaginal venous plexus [20, 21].
The vulva includes the mons pubis, labia majora and labia minora, clitoris, and
vestibule. It has low-to-intermediate signal intensity on T1-weighted images and
slightly high signal intensity on T2-weighted images [20].
The role of computed tomography (CT) in studying the female pelvis is limited and
is generally reserved to acute settings or for pelvic malignancies systemic staging.
When recognizable, the ovaries appear as small, roughly triangular structures of
soft-tissue density, often near the iliac vessels or uterus [3]. The gonadal vessels
may be an important anatomical landmark for their identification. On non-contrast
CT, the uterus appears as a uniform hypoattenuating formation, with a central zone
of lower attenuation representing the endometrial canal [9]. The vaginal mucosa,
which in fertile women is hyper-enhancing, becomes of similar density to that of the
vaginal wall in the post-menopausal age. The vaginal wall shows poor enhancement
after contrast administration [20]. Vaginal pathologies may be difficult to assess at
CT because of the similar density with the adjacent soft-tissue structures. The vulva
is identified as a triangular soft-tissue density structure within the perineum, poste-
rior to the symphysis pubis and anterior to the anal sphincter [23].
11.2 Ovaries
11.2.1 Endometriosis
Endometriosis results from the presence of aberrant endometrial tissue outside the
uterine cavity [24]. It is a common occurrence in the female population, with an
estimated 5 to 10% of women in the reproductive age being affected [25].
The pathogenesis of endometriosis is complex, particularly after menopause,
when it is unclear if it is a continuation or a reactivation of a previously existing
disease or a de novo condition. Estrogen exposure appears to have a key role;
indeed, due to the decreased estrogen levels after menopause, endometriotic lesions
in most cases regress in this age group [26] (Fig. 11.2).
Although endometriosis in post-menopausal patients is relatively uncommon, it
is estimated that about 2 to 5% of post-menopausal women are affected [27], and it
should be considered as a possible diagnosis. Hormone replacement therapy, espe-
cially estrogen-only treatments without progestin, and Tamoxifen use have been
associated with post-menopausal endometriosis [28–30]. Other risk factors include
a history of symptoms before menopause suggestive of endometriosis and condi-
tions that may raise the level of serum estrogens, such as obesity [28].
276 M. A. Cova et al.
a b
e f
Fig. 11.2 Endometriosis. (a) TVUS showing a unilocular lesion in the right ovary, with a homo-
geneous hypoechoic content and diffuse low-level internal echoes, the so-called ground glass
appearance (between calipers). (b–f) MRI of a different patient showing an endometrioid cyst in
the left ovary. Axial T2-weighted image (b) showing an ovoid mass with regular margins (white
arrow). The mass is characterized by a mostly cystic T2-hyperintense (b) and T1-hypointense (c)
content, and a hematic component in the dependent regions, separated by a fluid-fluid level. The
hematic component is in the subacute phase, showing T1-hyperintensity and T2-hypointensity
(black arrows), with restricted diffusion on DWI (d), as confirmed on the ADC map (e). In the
presence of a T1-hyperintense cystic content, it is always important to obtain fat-suppressed
T1-weighted images (f) to exclude the presence of fat. Note the large simple cystic mass in the
right ovary (asterisk, b)
11 The Female Urogenital System in Geriatric Patients 277
US Findings
US is the most common modality used in the suspect of endometriosis. Particular
attention should be paid to the evaluation of the ovaries and the cul-de-sac, common
sites involved [39].
The US appearance of endometrial cysts is highly variable. The most common
one is a unilocular cystic lesion with a homogeneous hypoechoic content, with dif-
fuse low-level internal echoes, sometimes referred to as “ground glass” [39, 42].
Rarely, they may be completely anechoic, similar to a functional cyst [39].
Endometriomas may also appear as multilocular complex lesions, with thick
walls and septa, wall nodularity, and echogenic foci within the cyst wall [43, 44].
These echogenic foci are thought to be related to cholesterol deposits in the endo-
metrial wall and should be differentiated from wall nodules, which usually appear
278 M. A. Cova et al.
larger and less echogenic [39]. Also, the multilocularity may in some cases be due
to multiple adjacent separate cysts [39]. Malignancy should always be suspected in
the presence of a solid mural nodule; an increasing size of the cyst is another less
reliable signs of degeneration [45].
The heterogeneous appearance of the endometrioid cysts results in a broad range
of differential diagnoses, including functional cysts, tubo-ovarian abscesses, der-
moid cysts, and benign or malignant lesions [41]. The stability or minimal growth
at follow-up represents an important distinguishing feature from functional cysts, in
particular hemorrhagic ones, which may be very similar in appearance but generally
have a more acute onset and resolve in 4–6 weeks [39].
MRI Findings
Endometrial cysts have two typical patterns of presentation. In the early subacute
bleeding phase, they will be T1-hyperintense and T2-hypointense. In the later sub-
acute phase, they will be hyperintense on both T1 and T2 weighted images [39, 46,
47]. The shading sign, i.e., loss of signal within the lesion that can be seen on
T2-weighted images, is an important sign of endometriomas. This is due to the pres-
ence of hematic products in different stages of degeneration because of repeated
bleedings [39]. After contrast administration, subtraction images help detect any
enhancing tissue if there is a concern of malignant degeneration [37]. The differen-
tial diagnosis of endometriomas includes dermoid cysts, hemorrhagic cysts, muci-
nous cystic neoplasms, and an ovarian carcinoma with internal hemorrhage. The
absence of signal loss on fat-suppressed T1-weighted images confirms the hematic
content and rules out fat-containing lesions such as dermoid cysts [39, 48].
T1-weighted images help in the differential with mucinous cystic neoplasms, which
will show a high signal intensity but less than fat or blood. Differentiating an endo-
metrioma from a hemorrhagic corpus luteum can be more difficult; hemorrhagic
cysts are usually unilocular (while endometriomas are often multilocular and bilat-
eral), do not show the T2-shading sign, and mostly disappear at follow-up. An ovar-
ian carcinoma with internal hemorrhage will have features suggestive of malignancies
such as solid components, larger dimensions, and septations [39].
Endometrial implants will have variable signal intensities. They may have low
T1 signal intensity and high T2 signal intensity, similar to the normal endometrium,
or they can be hyperintense or hypointense on both T1- and T2-weighted images [39].
Adherences will appear as spiculated hypointense bands between organs, with or
without anatomical distortion, that in advanced disease may result in the so-called
kissing ovaries configuration, i.e., the ovaries displaced posteriorly and medially
toward one another [49].
Imaging features of endometriosis-associated malignancy are similar to the other
malignancies not related to endometriosis. On MRI, it will often have an intermedi-
ate T2 signal intensity, with avid enhancement and restricted diffusion. Enhancing
mural nodules and septations will be best appreciated on post-contrast T1-weighted
images with fat suppression and subtraction [30]. Restricted diffusion may also
occur in benign endometriomas due to the presence of blood products [50]. Another
non-specific sign is the loss of T2 shading [51]. Extra-ovarian malignancy
11 The Female Urogenital System in Geriatric Patients 279
associated with endometriosis may have an infiltrative appearance, and benign vari-
ants, e.g., polypoid endometriosis, may mimic features of malignancy [30].
be clearly assessed [63]. Features that indicate a uterine origin are the presence of a
pedicle between the lesion and the uterus; the “bridging vessel sign” on contrast-
enhanced T1-weighted images (i.e., the presence of vascular structures going from
the uterus to the lesion as it receives its blood supply from uterine vessels); and, in
case of uterine leiomyomas, the normal uterine tissue may be draped around the
lesion like a “claw” [46, 63]. Suggestive of an ovarian origin is the “ovarian beak
sign,” i.e., the presence of sharp angles between the lesion and the ovary [64]; also,
an ovarian fibroma will be separate from the uterus [63]. According to current
guidelines, the MRI protocol should include a sagittal T2-weighted sequence of the
pelvis, a T1- and T2-weighted sequences in the same orthogonal plane (axial or
coronal) and with the same slice thickness covering the mass, a DWI sequence, and
dynamic contrast-enhanced T1-weighted sequences [63]. If the lesion shows high
signal intensity on T1-weighted images, an axial fast spin-echo (FSE) T1-weighted
sequence with fat suppression needs to be acquired. If doubt exists on whether the
lesion belongs to the uterus or the ovary, 3D T1-weighted or FSE T1-weighted
sequences with fat suppression or FSE T2-weighted sequences may be acquired on
the axial plane of the ovary, which corresponds to the parallel plane of the endome-
trial cavity [10].
Epithelial Tumors
Epithelial tumors constitute 60% of all ovarian tumors and 85% of malignant ones
[67]. Their incidence increases with age, peaking in the sixth to seventh decade
[68]. They include serous, mucinous, seromucinous, endometrioid, clear cell,
Brenner tumors, and undifferentiated carcinoma [69].
a b
Fig. 11.3 Ovarian cystadenoma with features of malignancy. Coronal (a) and sagittal (b) CT scan
reconstructions showing a large abdominal multilocular cystic lesion (*) with the presence of
thick, irregular walls and septa, papillary projections within the lumen (arrow) and enhancing soft-
tissue components with necrotic foci
(Fig. 11.3). The solid tissue will have intermediate signal intensity on T1- and inter-
mediate on T2-weighted images [70, 71]. The signal intensity of the papillary veg-
etations reflects their tissue architecture, composed of a stromal core lined by
neoplastic cells. They will appear as structures of intermediate signal intensity on
T1-weighted images and with a hypointense core lined by a hyperintense neoplastic
epithelium on T2-weighted images [67, 72]. Malignant lesions tend to have a faster
and more intense enhancement than benign lesions after contrast administration
[73]. Malignancy is also associated with ascites, lymphadenopathies, invasion of
pelvic organs, and peritoneal and omental implants, whose identification can be
helped by the DWI sequence, especially if small in dimensions [70, 71].
Serous cystadenomas appear as mostly unilocular cystic mass, with a thin wall or
septum and no vegetations. They have a homogeneous low signal intensity on
T1-weighted images and high signal intensity on T2-weighted images, with no
enhancement or enhancement of the thin walls alone after contrast administration
[67, 74, 75]. Serous cystadenocarcinoma can have discrete dimensions, are bilateral
in two-thirds of cases, and have malignant features at imaging [68].
Mucinous cystadenomas are multilocular cystic masses with thin walls or septa
and no vegetations, usually larger than serous cystadenomas [67, 71]. The signal
intensity will depend on mucin concentration, with T1 hypointensity and T2 hyperin-
tensity with higher water content, and T1 hyperintensity and T2 hypointensity with
thicker mucin [70]. Sometimes, multiple locules with variable signal intensities can be
observed, in an appearance referred to as “stained-glass” [76] (Figs. 11.4 and 11.5).
282 M. A. Cova et al.
a b
c d
e f
Fig. 11.4 Ovarian cystadenoma. (a, b) TVUS showing an ovoid mass with regular margins and
minimally thickened wall (3 mm) (between calipers, a), with a finely corpuscular content. At the
color Doppler analysis, neither the mass nor its thickened wall exhibit any significant vasculariza-
tion (b). (c–f) MRI exam of the same patient showing a mass in the left ovary with minimally
thickened wall, well appreciated on the coronal T2-weighted sequence (arrow, c), and a content
with high signal intensity on T2-weighted images and slightly low signal intensity on T1-weighted
images (arrow, d). There is no restricted diffusion (e) and only the wall shows enhancement (f).
The mass has no aggressive features
11 The Female Urogenital System in Geriatric Patients 283
a b
Fig. 11.5 Mucinous cystadenoma. Axial contrast-enhanced CT scan (a, b) and sagittal recon-
structions (c) showing a cystic mass of significant dimensions within the abdomen, with thin, mini-
mally enhancing wall and septa (arrows), and no vegetations. The mass dislocates the abdominal
organs posteriorly
a b
Fig. 11.6 Endometrioid carcinoma. (a, b) Axial contrast-enhanced CT scan at different levels of
a patient admitted to the emergency room for left flank pain. In the abdomen, there is a hypodense
voluminous solid mass (white asterisks, a, b), slightly heterogeneous in density and with no
enhancement after contrast administration (unenhanced CT not shown). A small amount of fluid in
its most caudal region is also present (black asterisk, b). The patient underwent surgery, and this
was confirmed to be an endometrioid carcinoma
hyperplasia or synchronous endometrial carcinoma are also present [24, 63, 79].
The tumor appears as a complex cyst with solid components, with the solid compo-
nent having an intermediate or heterogeneous signal intensity and enhancement
after contrast administration [67, 80]. The involvement is bilateral in about 40% of
cases, and this frequently indicates the spread of the disease beyond the genital
tract [24].
Clear cell carcinomas (Fig. 11.7) represent about 5% of ovarian tumors and may
be solid or cystic [67, 68]. The usual MR appearance is a unilocular cyst with solid
protrusions into the lumen, with very variable T1 signal intensity [67].
Brenner tumors are rarely malignant and constitute 2–3% of ovarian tumors.
They are associated with other ovarian tumors in 30% of cases, and the large major-
ity are unilateral [67]. There is a cystic and a solid variant [24]. They appear as
multilocular cystic masses with a solid component that is T2-hypointense and has at
least a moderate enhancement after contrast administration. Amorphous calcifica-
tions may be present [46, 67].
b c
Fig. 11.7 Clear cell ovarian carcinoma. (a) Color Doppler US showing a unilocular cyst (*) with
solid protrusions (arrows) into the lumen, demonstrating high signal. (b, c) Axial contrast-
enhanced CT showing enhancing nodules (arrows, b) and the infiltration of the sigma (arrow, c)
• Benign cystic mature teratomas are the most common germ cell tumor of the
ovary, composed of mature tissue of ectodermal, mesodermal, and endodermal
origin [48]. Therefore, hairs, skin glands, bone, cartilage, fat, and muscles may be
present within the mass. Monodermal tumors of the ovary, such as struma ovarii
or carcinoid tumors, also exist [48]. The US appearance is usually non-specific,
appearing as a predominantly cystic, solid, or complex mass with reflections and
shadowing. Findings vary from the classic presence of a so-called Rokitansky
nodule (cystic lesion with a densely echogenic tubercle projecting into the cyst
lumen), to an atypical diffusely or partially echogenic mass with sound attenua-
tion, due to the presence of sebaceous material and hair within the cavity, to mul-
tiple thin echogenic bands due to hairs. Fluid-fluid levels can result from the
separation of the sebum, which appears more hypoechoic than the fluid layer.
Shadowing may be present due to calcific or tooth components [48, 85]. At MRI,
286 M. A. Cova et al.
• Granulosa cell tumors are the most common ovarian tumors to produce estrogens
and the most common malignant sex cord-stromal tumor [90]. The adult form
represents 95% of them and occurs mostly in perimenopausal and post-
menopausal patients [89], with an incidence peaking at 50–55 years [91]. The
imaging characteristics are non-specific, as they may appear as a solid mass, a
tumor with hemorrhagic or fibrous components, or may be multilocular cystic or
entirely cystic tumors [67]. The multilocular cystic pattern with solid compo-
nents is the most common one, often resulting in a typical sponge-like appear-
ance on T2-weighted sequences [89]. For their hormonal production, endometrial
hyperplasia, endometrial polyps, or endometrial carcinoma can co-occur in
3–25% of cases [90]. Peritoneal dissemination is not frequent [67].
• Fibromas, fibrothecomas, and thecomas are a spectrum of benign tumors ranging
from purely fibrotic to lipid-rich tumors generally occurring in peri- and post-
menopausal patients [89, 92]. Due to their fibrotic component, fibromas have a
11 The Female Urogenital System in Geriatric Patients 287
a b
c d
e f
Fig. 11.8 Teratoma. On T1-weighted (a) and T2-weighted images (b), the tumor (*) appears
heterogeneously hyperintense, with a signal intensity similar to retroperitoneal fat, and it contains
T2-hyperintense and T1-hypointense septa (black arrows). The lesion shows loss of signal on fat-
saturated T1-weighted imaging (c). On the T1-weighted (d) and T2-weighted images (e) on a dif-
ferent plane, the so-called Rokitansky nodule (white arrows) can be appreciated. It appears
T1-hypointense and T2-hyperintense, with mild enhancement on fat-saturated contrast-enhanced
T1-weighted images (f, coronal plane)
288 M. A. Cova et al.
Adnexal torsion is the twisting of an ovary, and often the fallopian tube, on their
ligamentous support, that may result in compromised blood flow [94]. It constitutes
a gynecological emergency requiring surgery, presenting with acute onset, intense,
and progressive pain, which in post-menopausal women may be continuous and
dull rather than acute and sharp [95, 96]. Adnexal torsion frequently is a complica-
tion of ovarian cysts and tumors [97], and most cases occur in pre-menopausal
patients because of the increased frequency of benign cysts and teratomas. It may
occur after menopause, although the incidence is low [95]. While the rate of malig-
nancy in younger patients is believed to be low, in post-menopausal patients, torsion
is more frequently associated with a malignant mass, but this occurs in a relative
minority of cases [95, 98].
11 The Female Urogenital System in Geriatric Patients 289
a b
c d
Fig. 11.10 Ovarian fibroma. (a–c) MRI showing a roundish mass in the left ovary, with very low
signal intensity on T2-weighted image (arrow, a) and low signal intensity on T1-weighted image
(arrow, b), with only mild enhancement after contrast administration (c), compatible with an ovar-
ian fibroma. (d) CT scan of a different patient showing a large solid abdominal mass with mild
enhancement and heterogeneous density (*), confirmed to be a fibroma after surgical excision
A nabothian cyst is a mucus-filled cyst located usually on the surface of the cervix
[3]. On TVUS, it usually appears as an anechoic well-defined cystic lesion, with no
signal on color Doppler imaging [108], whereas, on non-contrast CT, it may appear
as a low attenuation formation. It is usually hyperintense on T2-weighted images,
while on T1-weighted images it appears as an intermediate or hyperintense lesion
because of its proteinaceous component and does not show enhancement after con-
trast administration [109, 110] (Fig. 11.11).
11 The Female Urogenital System in Geriatric Patients 291
a b
Fig. 11.11 Nabothian cyst (arrows). On gray-scale TVUS (a), a Nabothian cyst appears as an
anechoic well-defined cystic lesion, whereas it appears hyperintense on the axial T2-weighted MR
image (b)
11.3.2 Adenomyosis
On MRI, the diffuse form and the focal form of adenomyosis can be also evalu-
ated. The former presents itself with increased uterine dimensions (Fig. 11.12a). On
T2-weighted images, ill-defined hypointense foci, because of muscular hyperplasia
292 M. A. Cova et al.
a b
Fig. 11.12 Diffuse form of adenomyosis in a bicornuate uterus (a) and focal form of adenomyo-
sis (b, c). (a) Axial T2-weighted image showing increased uterine dimensions, ill-defined hypoin-
tense areas, due to muscular hyperplasia and hypertrophy, mildly hyperintense foci consisting of
ectopic endometrium, and markedly hyperintense cystic foci. Less than 25% of the lesion is sur-
rounded by normal myometrium. (b, c) In a different patient, axial (b) and sagittal (c) T2-weighted
images showing a hypointense mass-like formation (arrows) in the myometrium with hyperintense
cystic area (arrowheads). More than 25% of the lesion is surrounded by normal myometrium
11.3.3 Polyps
a b
Fig. 11.13 Cervical (a) and endometrial (b) polyps. (a) Gray-scale TVUS showing a hypoechoic
pedunculated cervical polyp (between calipers). (b) Gray-scale TVUS showing a mildly
hypoechoic endometrial polyp with hyperechoic periphery (between calipers)
11.3.4 Leiomyomas
Leiomyomas are the most frequent uterine benign disease, occurring in about
20–30% of young women, usually regressing in post-menopausal women [64, 123].
Leiomyomas, generally affecting the body of the uterus and more rarely the cervix,
are uterine smooth muscle benign neoplasms that may also contain connective tis-
sue and have a pseudocapsule [119, 123]. They can be divided depending on their
location into: submucosal (in the subendometrial zone), intramural (within the myo-
metrium), subserosal (in the subserosal zone), or cervical (in the cervix) [110, 123].
On TVUS, leiomyomas may appear as well-defined lesions hyperechoic or
hypoechoic compared to the myometrium; they may show shadowing near and
within the formation, and calcifications with distal shadowing may also be present
(Fig. 11.14a). On MRI, leiomyomas can be appreciated as solid roundish forma-
tions with sharp borders [124]. Besides standard T1-weighted and T2-weighted
sequences of the female pelvis, additional coronal and axial oblique perpendicular
to the long axis of the uterus T2-weighted sequences are helpful to accurately local-
ize the lesions and to confirm their uterine origin. On T1-weighted sequences, they
can be hardly distinguished because they show intermediate signal intensity similar
to myometrium [119, 123], whereas, on T2-weighted images, they usually show
low signal intensity or slightly high signal intensity in case of high cellularity and
may have a hyperintense pseudocapsule [123] (Fig. 11.14b). When fat is present,
leiomyomas are called lipoleiomyomas, and they appear as high signal intensity
lesions on both T1 and T2-weighted images [110] (Fig. 11.15). After contrast
294 M. A. Cova et al.
a b
Fig. 11.14 Leiomyoma. (a) Gray-scale TVUS showing a large, well-defined subserosal leiomy-
oma (arrow). (b) Sagittal T2-weighted MR image in the same patient confirming the leiomyoma
(arrow), showing the typical hypointense signal
a b
Fig. 11.15 Lipoleiomyoma (*). It appears hyperintense on T2-weighted image (a) and on
T1-weighted image (not shown). The lesion shows loss of signal on fat-saturated T1-weighted
image (b) and no enhancement on fat-saturated T1-weighted image after contrast administration (c)
11 The Female Urogenital System in Geriatric Patients 295
a b
Fig. 11.16 Calcific leiomyoma. Gray-scale TVUS showing an end-stage leiomyoma (black
arrow, a and between calipers, b) with calcific degeneration (arrowheads) and distal shadowing
(white arrows)
296 M. A. Cova et al.
a b
Fig. 11.17 Large degenerated leiomyoma. Coronal (a) and sagittal (b) T2-weighted images
showing a large degenerated intramural leiomyoma. Some components show high signal on DWI
(arrow, c). In this case, it is not possible to certainly differentiate this large degenerated leiomyoma
from leiomyosarcoma based on MR imaging. Nevertheless, the absence of nodular borders, hem-
orrhage, T2-weighted dark areas, and central foci without enhancement is suggestive of a large
degenerated leiomyoma
TVUS should be the first imaging choice in older women with abnormal bleed-
ing to measure the endometrial thickness. An upper threshold of 5 or 4 mm is con-
sidered as the cut-off of normality in these patients [133].
Although the staging of EC, which follows the International Federation of
Gynecology and Obstetrics (FIGO) criteria [132], depends on surgical and histo-
pathological findings, MRI, thanks to its contrast resolution, can be an important
tool in the preoperative staging of the EC [122, 136].
According to current guidelines, the dedicated MRI protocol should include an axial
oblique perpendicular to uterus corpus T2-weighted sequence for an accurate evaluation
of the depth of myometrial invasion, an axial oblique DWI sequence to match the
T2-weighted sequence, and a T1- or T2-weighted sequence up to the renal hilum for
lymph nodes and hydronephrosis. In case of grade 3 endometrioid adenocarcinoma or
non-endometrioid carcinomas, an axial DWI sequence to match the sequence for lymph
nodes and hydronephrosis should be added. In addition, contrast-enhanced images
acquired after two and a half minutes should be usually performed [10].
EC is stage IA (Fig. 11.18) in case of a tumor confined to less than half of the
myometrium, whereas it becomes stage IB (Fig. 11.19) in case of involvement of
a b
Fig. 11.18 Endometrial cancer, FIGO stage IA: A tumor (arrow) confined to less than half of the
myometrium can be appreciated on the sagittal T2-weighted image (a), DWI image (b), and fat-
suppressed T1-weighted contrast-enhanced image, on which it appears hypointense, being hypo-
vascular compared to the normal myometrium (c)
298 M. A. Cova et al.
a b
c d
Fig. 11.19 Endometrial cancer, FIGO stage IB. A tumor (asterisk) within the uterine lumen,
hyperintense on T2-weighted image (a) and hypointense being hypovascular compared to the
myometrium on the contrast-enhanced image with fat suppression (b), involving more than half of
the myometrium (arrow, b), can be appreciated. The lesion shows restricted diffusion, with high
signal intensity on DWI (c) and low signal on the ADC map (d)
more than half of the myometrium [136, 137]. Stage II EC is a tumor involving the
hypointense stroma of the cervix. A dynamic contrast-enhanced T1-weighted
sequence may be useful in the evaluation of difficult cases; indeed, if the enhance-
ment of the cervical mucosa is conserved in the delayed phase, the presence of
stromal infiltration can be excluded [136]. EC is stage III if the lesion interrupts the
outer contour of the uterus; EC in this stage remains confined to the true pelvis [136,
137]. EC is stage IVA in case of invasion of the vesical or rectal mucosa. This can
be excluded with high accuracy in case of preservation of the fat planes between the
lesion and bladder or rectum. When distant metastases are present, EC becomes
stage IVB [122, 136, 137] (Fig. 11.20).
11 The Female Urogenital System in Geriatric Patients 299
a b
Fig. 11.20 Endometrial cancer, FIGO stage IVB. A tumor involving less than half of the myome-
trium can be appreciated on the sagittal T2-weighted image (arrow, a). Axial T2-weighted imaging
(b) shows a peritoneal metastasis in the right iliac fossa (white arrow, b and c), which has high
signal intensity on DWI (c). The staging was histologically confirmed after hysterectomy
MRI can also be useful in the evaluation of myometrial and cervical stromal
invasion [136]. On T1-weighted sequences, EC usually appears as a lesion isoin-
tense to the myometrium [137], whereas, on T2-weighted images, it appears as a
diffuse or well-delineated mass with heterogeneous intermediate signal intensity,
higher than the hyperintense endometrium and lower than the hypointense myome-
trium [122, 136].
On post-contrast T1-weighted images, the lesion shows an early enhancement
compared to the surrounding endometrium and slow enhancement compared to the
myometrium, appearing hypointense compared to the myometrium in the late phase
[122]. Post-contrast T1-weighted sequences can play a key role in the evaluation of
the infiltration of the cervical stroma and the myometrium. In case of EC confined
to the endometrium, a continuous enhancement of the subendometrial zone can be
seen, whereas a disruption of this zone can be present in case of myometrial inva-
sion [136].
EC shows restricted diffusion on DWI due to increased cellularity. Indeed, DWI
increases the capability of detecting EC, in particular smaller ones, and can be use-
ful in the assessment of the infiltration of the myometrium and the cervical stroma;
DWI also improves the identification of metastases in the vagina, cervix, adnexa,
and peritoneum [122, 136, 137].
300 M. A. Cova et al.
DWI is sensitive but not specific for the identification of malignant lymph nodes
because reactive lymph nodes also show high signal intensity on this sequence.
Therefore, a size threshold of 10 mm in short axis diameter for para-aortic nodes
and 8 mm for pelvic ones is used to define neoplastic infiltration. Round shape,
spiculated margins, heterogeneous signal intensity, or necrosis are other morpho-
logic patterns suggesting tumor involvement [122, 136, 137].
After therapy, diffusion-weighted and post-contrast T1-weighted sequences can
also be used to differentiate the inflammation after radiotherapy from recur-
rence [122].
Sometimes the differential diagnosis between EC and uterine polyps or endome-
trial hyperplasia can be difficult: DWI may be a useful tool in these cases because
the ADC value of normal endometrium and polyps is significantly higher than
EC. Nevertheless, also hyperplastic endometrium may show low ADC values like
EC. Blood products may also show low ADC values; a careful evaluation of
T1-weighted images is important to confirm this finding. In addition, the evaluation
of T2-weighted images can be useful for the differential diagnosis between EC and
a leiomyoma mimicking an endometrial thickening because leiomyomas appear
hypointense on this sequence [112].
Cervical cancer (CC) is the fourth most frequent neoplasm in women, with an aver-
age age of 53 years at diagnosis [138]. Patients usually present with vaginal bleed-
ing and discharge [139, 140]. Various risk factors are associated with CC, including
young age at first sexual intercourse, multiple sexual partners, sexually transmitted
viral infections (HPV, HSV2), and multiparity [140–142]. CC usually arises from
the transformation zone, between the ectocervix and the endocervix [143]. The
most common cancer histotypes are adenocarcinoma and squamous cell carcinoma
(up to 89% of cases) [140]. Dysplasia, which can be divided into mild, moderate,
and severe, precedes the development of neoplasia, which is defined as preinvasive
(cervical intraepithelial neoplasia, CIN) if the basement membrane is not involved,
and as invasive in case of penetration of the basement membrane and involvement
of stroma of the cervix.
Staging is obtained according to the FIGO system [132]. While TVUS is an
important tool for the initial evaluation of EC, evidence supporting its use in CC is
weak because the parametrial involvement cannot be surely assessed on
11 The Female Urogenital System in Geriatric Patients 301
a b
c d
e f
Fig. 11.21 Cervical cancer, FIGO stage IV. Gray-scale TVUS (a) showing a hypoechoic cervical
lesion (between calipers). Contrast-enhanced axial CT scan (b) showing the cervical cancer
involving the posterior wall of the bladder (black arrow). Note the dilation of the right ureter, better
shown on curvilinear sagittal reconstructions (white arrow, c), secondary to the infiltration of the
right ureteral meatus. Axial (d) and sagittal (e) T2-weighted images showing the cervical cancer
involving the bladder; the organ involvement appears as a focal disruption of the hypointense mus-
cular layer of the bladder (arrow, e). The post-contrast axial T1-weighted sequence (f) is useful in
the evaluation of tumor infiltration because the lesion usually shows stronger enhancement than
the muscular layer; note also the involvement of the anterior wall of the rectum (arrow, f)
11 The Female Urogenital System in Geriatric Patients 303
The squamous cell vaginal carcinoma occurs more commonly in the upper third
of the vagina, on the posterior wall, and can appear as a diffuse mass with ill-defined
and irregular margins, as a well-defined lobulated mass, or as a circumferential
thickening [152, 156]. The tumor is best assessed on T2-weighted images, on which
it shows an intermediate signal intensity, which is distinguished from the low inten-
sity of the vaginal wall. On T1-weighted images, it is isointense to muscles and can
be identified only when large enough to alter the contour of the vagina [152].
Vaginal cancer spreads by direct extension to the surrounding pelvic organs,
including paravaginal tissue, parametria, urethra, bladder, and rectum [148]. If the
T2-hypointensity of the outer layer of the vaginal wall is preserved, the tumor is
limited to the mucosa and is a stage I; when the wall hypointensity is disrupted,
the tumor has extended to the paravaginal tissues and is stage II. The extension
through the vaginal wall is best assessed on the oblique axial plane. A stage III
tumor involves the pelvic sidewalls, which are best assessed on axial and coronal
planes, and it is seen as a higher T2-signal intensity within the muscles due to
edema or direct tumor invasion. In stage IVA, the cancer involves the mucosa of
the rectum or bladder, and direct infiltration with loss of the normal hypointensity
of the bladder and rectal walls as well as loss of the vesicovaginal fat plane or
rectovaginal septum is appreciated. Bullous edema may be difficult to differenti-
ate from tumor infiltration and may result in overstaging. A stage IVB tumor
spreads to distant organs, and lungs, liver, and bones are commonly involved sites
[151, 152, 155] (Fig. 11.22).
The lymphatic spread of the tumor is complex and is often present, even in ear-
lier stages (6–14% in stage I, 26–32% in stage II) [156, 157]. In general, upper vagi-
nal tumors drain to pelvic lymph nodes, including obturator, internal and external
iliac lymph nodes; involvement of para-aortic nodes is rare. The lower vagina drains
to inguinal and femoral lymph nodes. The middle third of the vagina can follow
either route. Posterior wall lymphatics drain to the inferior gluteal, sacral, and rectal
nodes [148, 152].
After treatment, recurrences are most commonly seen within the first 2 years.
Tumors of the upper third tend to recur locally, while lower tumors are more
often associated with pelvic sidewalls invasion or distant recurrences [151, 152].
MRI is useful in differentiating residual tumor from post-treatment changes
although it may be difficult, especially in the few months after treatment, when
T2-hyperintense edema is also present. In time, the scar tissue appears T2
hypointense, while the tumor has T2-intermediate to high signal intensity and
enhances avidly [151].
Complications after treatment generally occur within 5 years, but they may be as
late as 20 years and are due to radiation-induced bladder, vaginal, and rectal toxic-
ity. They include cystitis, proctitis, bowel stricture and perforation, bone osteone-
crosis, and stress fractures [151, 158]. Common complications are rectovaginal and
vesicovaginal fistulas, best demonstrated on axial or sagittal high-resolution
T2-weighted or short tau inversion recovery (STIR) sequences, where they appear
as a tract with fluid hyperintensity and air hypointensity [151, 152].
11 The Female Urogenital System in Geriatric Patients 305
a b
c d
Fig. 11.22 Vaginal cancer. Axial T2-weighted sequence (a), axial fat-suppressed sequence (b),
and fat-suppressed contrast-enhanced T1-weighted sequences on the axial (c) and sagittal (d)
plane, showing a prevalently exophytic mass (white arrow, a) with spiculated margins, involving
the postero-lateral fornix and the postero-lateral region of the cervix on the left. The mass infil-
trates the adjacent adipose tissue and the left uterosacral ligament, and it infiltrates the rectal serosa
at 2 o’ clock (black arrow, a). Note the right presacral small lymph node with irregular margins
(arrow, c). According to FIGO staging, this is a stage IV tumor
bowel, occur [152]. In general, the MR features of vaginal metastases mimic the
primary tumor [152].
11.5 Vulva
revised in 2009 and can be applied to most vulvar cancer except melanoma [159].
MRI is preferred for local staging. The exam is performed after bladder emptying,
and ultrasound gel may be used to distend the vagina to better assess vaginal wall
infiltration and possibly to identify smaller vulvar lesions. The protocol should
include axial T1-weighted FSE images with a large FOV to evaluate lymphade-
nopathies and bone marrow abnormalities; axial and coronal high-resolution
T2-weighted FSE sequences to evaluate the primary tumor; and sagittal dynamic
fat-saturated contrast-enhanced T1-weighted images with a small FOV for the
extent of tumor involvement. High-resolution axial T2-weighted FSE sequences
with a small FOV and 3-mm thick sections can be obtained through the perineum.
The tumor may be better appreciated on T2-weighted images with fat suppression
than non-fat saturated images, and contrast-enhanced sequences can help identify
small tumors and assess invasion of the urethra, anus, and vagina. Diffusion-
weighted sequences can also be useful to assess the primary tumor and lymphade-
nopathies [163].
On MRI, the tumor has low signal intensity on T1-weighted images and
intermediate-to-high signal intensity on T2-weighted images [20]. In two-thirds
of cases, the labia are involved, with the clitoris and Bartholin glands [20]
(Fig. 11.23). Small stage I cancers may be missed and stage I or II tumors with an
“en-plaque” aspect may also be difficult to identity on MRI [164]. The urethra,
anorectum, vagina, perineal muscles, and the bladder should be carefully assessed
for signs of infiltration, which will appear as a tissue of intermediate signal inten-
sity, in continuity with the primary tumor, within the hypointense muscular walls
of the urethra, vagina, and anorectum. T2-weighted images in the sagittal and
coronal planes could be helpful to assess the whole tumor if deep infiltration is
suspected [164].
The tumor spreads mainly by local invasion, followed by lymphatic spread and
hematogenous spread to distant organs (lung, liver, bone) [161]. Inguinal and femo-
ral lymph nodes are the first to be involved, followed by pelvic nodes [165]. Lymph
node involvement can be uni- or bilateral, depending on tumor size and its closeness
to the middle line [159].
The most important prognostic factors in vulvar cancer are tumor size, depth of
infiltration, and particularly the presence of lymph node metastases. Recurrences
can be local or distant and are generally seen within 2 years after initial treatment
[20] (Fig. 11.24).
308 M. A. Cova et al.
a b
c
d
e f
Fig. 11.23 Vulvar cancer. At the vaginal introitus, on the right side, there is a vulvar lesion (arrow,
a), with intermediate T2 signal intensity (a), restricted diffusion (b, c), and enhancement after
contrast administration (d). At the PET/CT (e–g), on the unenhanced CT, a paravaginal hypodense
tissue in the perineum can be recognized (arrow, e), and it shows an intense uptake of the
[18F]-fluorodeoxyglucose tracer (f), well-shown on the fusion imaging (g)
11 The Female Urogenital System in Geriatric Patients 309
a b
c d
e f
g
310 M. A. Cova et al.
Fig. 11.24 Locoregional recurrence of vulvar cancer. Patient with previous vulvectomy per-
formed 5 years before and a urinary catheter in place. At the vaginal introitus, there is a solid lesion
(black arrow, a), heterogeneously hypointense on T2-weighted sequences (a, b), that involves the
full thickness of the vaginal wall, extending cranially up to the middle vaginal third (b), and sur-
rounding the urethra in its inferior two-thirds (white arrow, a). The lesion shows restricted diffu-
sion (c, d) and a heterogeneous enhancement for the presence of central necrotic areas (e). At the
PET/CT scan, the lesion can be seen as a solid mass on the unenhanced CT images (arrow, f), with
an intense uptake of the [18F]-fluorodeoxyglucose tracer (g)
Functional disorders of the pelvic floor refer to a group of medical conditions affect-
ing the ligaments, fasciae, and muscles, supporting the pelvic organs [166]. These
conditions are relatively common and predominantly affect older women [167].
The appearance of the pelvic organs can be evaluated on MRI at rest and during
active contraction, the so-called Valsalva maneuver. A state-of-the-art MRI protocol
includes axial, coronal, and sagittal rapid half-Fourier T2-weighted imaging
sequences at rest; successively, six to eight rapid T2-weighted images are also
acquired during the Valsalva maneuver [168].
The pelvic floor can be divided into three anatomic compartments: the anterior,
the middle, and the posterior. In the anterior compartment there are the bladder and
the urethra, in the middle one the uterus, the cervix, and the vagina, in the posterior
one the rectum, the anus, and the anal sphincter. This paragraph will be focused on
the pathology of the middle compartment, while the pathology of the anterior and
the posterior ones will not be included in this chapter.
In the pelvic middle compartment, disorders include uterine or cervical prolapse.
Pelvic organ prolapse is the abnormal descent or herniation of the pelvic organs
through their respective hiatus due to failure of support structures and perineal hia-
tal weakening. Uterine prolapse is the uterine herniation beyond the vagina, caused
by a failure of the ligamentous and fascial supports (the pubocervical ligaments,
parametrium and paracolpium, uterosacral ligaments, rectovaginal fascia, and peri-
neal body). The laxity of the uterosacral ligaments contributes to an anterior move-
ment of the cervix with progressive uterine retroversion, causing the prolapse [168,
169]. Cervical prolapse can be seen as a bulging mass outside the external genita-
lia [168].
On MR defecography, it can be diagnosed when the cervix is located 1 cm below
the pubococcygeal line (PCL), a straight line connecting the inferior border of the
pubic symphysis to the last coccygeal joint [167]. The distance between the PCL
and the most anterior and inferior aspect of the cervix is used as reference for grad-
ing: a prolapse is considered small when the distance is less than 3 cm, moderate if
3 to 6 cm, and severe if over 6 cm [166]. In case of previous hysterectomy, the vagi-
nal apex should be at least 1 cm above the PCL line, using the most posterior and
superior aspect of the vaginal vault [166]. Vaginal vault prolapse is generally associ-
ated with other pelvic prolapses, most commonly an enterocele [170].
11 The Female Urogenital System in Geriatric Patients 311
References
1. Permuth-Wey J, Sellers TA. Epidemiology of ovarian cancer. In: Verma M, editor. Cancer.
Epidemiology: modifiable factors. Totowa, NJ: Humana Press; 2009. p. 413–37.
2. Tinelli A, Malvasi A, Rahimi S, Negro R, Vergara D, Martignago R, Pellegrino M, Cavallotti
C. Age-related pelvic floor modifications and prolapse risk factors in postmenopausal
women. Menopause. 2010;17:204–12.
3. Langer JE, Oliver ER, Lev-Toaff AS, Coleman BG. Imaging of the female pelvis through the
life cycle. Radiographics. 2012;32:1575–97.
4. Hall DA, McCarthy KA, Kopans DB. Sonographic visualization of the normal postmeno-
pausal ovary. J Ultrasound Med. 1986;5:9–11.
5. Pavlik EJ, DePriest PD, Gallion HH, Ueland FR, Reedy MB, Kryscio RJ, van Nagell
JR. Ovarian volume related to age. Gynecol Oncol. 2000;77:410–2.
6. Cohen HL, Tice HM, Mandel FS. Ovarian volumes measured by US: bigger than we think.
Radiology. 1990;177:189–92.
7. Aviram R, Gassner G, Markovitch O, Cohen I, Fishman A, Tepper R. Volumes of normal
ovaries, ovaries with benign lesions, and ovaries with cancer in menopausal women: is there
an optimal cut-off value to predict malignancy? J Clin Ultrasound. 2008;36:1–5.
8. Revzin MV, Moshiri M, Katz DS, Pellerito JS, Mankowski Gettle L, Menias CO. Imaging
evaluation of fallopian tubes and related disease: a primer for radiologists. Radiographics.
2020;40:1473–501.
9. Yitta S, Hecht EM, Mausner EV, Bennett GL. Normal or abnormal? Demystifying uterine
and cervical contrast enhancement at multidetector CT. Radiographics. 2011;31:647–61.
10. ESUR Female Pelvis Imaging Working Group. ESUR quick guide to female pelvis imaging,
1.0. Vienna: European Society of Urogenital Radiology; 2019.
11. Outwater EK, Mitchell DG. Normal ovaries and functional cysts: MR appearance. Radiology.
1996;198:397–402.
12. Bartoli JM, Moulin G, Delannoy L, Chagnaud C, Kasbarian M. The normal uterus on mag-
netic resonance imaging and variations associated with the hormonal state. Surg Radiol Anat.
1991;13:213–20.
13. Hricak H, Alpers C, Crooks LE, Sheldon PE. Magnetic resonance imaging of the female
pelvis: initial experience. AJR Am J Roentgenol. 1983;141:1119–28.
14. Takeuchi M, Matsuzaki K, Nishitani H. Manifestations of the female reproductive organs on
MR images: changes induced by various physiologic states. Radiographics. 2010;30:1147.
15. Brown HK, Stoll BS, Nicosia SV, Fiorica JV, Hambley PS, Clarke LP, Silbiger ML. Uterine
junctional zone: correlation between histologic findings and MR imaging. Radiology.
1991;179:409–13.
16. Meylaerts LJ, Wijnen L, Grieten M, Palmers Y, Ombelet W, Vandersteen M. Junctional zone
thickness in young nulliparous women according to menstrual cycle and hormonal contracep-
tion use. Reprod Biomed Online. 2017;34:212–20.
17. Kılıçkesmez Ö, Fırat Z, Oygen A, Bozkurt DK, Güzelbey T, Gürses B, Taşdelen N. Diffusion
tensor imaging of the uterine zones related to the menstrual cycle and menopausal status at 3
tesla MRI. Balkan Med J. 2016;33:607–13.
18. deSouza NM, Hawley IC, Schwieso JE, Gilderdale DJ, Soutter WP. The uterine cervix on
in vitro and in vivo MR images: a study of zonal anatomy and vascularity using an enveloping
cervical coil. AJR Am J Roentgenol. 1994;163:607–12.
19. Togashi K, Nakai A, Sugimura K. Anatomy and physiology of the female pelvis: MR imag-
ing revisited. J Magn Reson Imaging. 2001;13:842–9.
20. Tsili AC. Vagina and vulva. In: Forstner R, Cunha TM, Hamm B, editors. MRI CT female
pelvis. Cham: Springer International; 2019. p. 343–68.
21. Landes CJ, Blair JC. Normal growth and puberty. In: Imaging of gynecological disorders in
infants and children. Berlin: Springer; 2011. p. 81–113.
312 M. A. Cova et al.
22. Walker DK, Salibian RA, Salibian AD, Belen KM, Palmer SL. Overlooked diseases of the
vagina: a directed anatomic-pathologic approach for imaging assessment. Radiographics.
2011;31:1583–98.
23. Grant LA, Sala E, Griffin N. Congenital and acquired conditions of the vulva and vagina on
magnetic resonance imaging: a pictorial review. Semin Ultrasound CT MR. 2010;31:347–62.
24. Ellenson LH, Pirog EC. The female genital tract. In: Robbins and Cotran pathologic basis of
disease. Amsterdam: Elsevier; 2014. p. 991–1042.
25. Bulun SE. Endometriosis. N Engl J Med. 2009;360:268–79.
26. Secosan C, Balulescu L, Brasoveanu S, Balint O, Pirtea P, Dorin G, Pirtea L. Endometriosis in
menopause-renewed attention on a controversial disease. Diagnostics (Basel). 2020;10:E134.
27. Punnonen R, Klemi PJ, Nikkanen V. Postmenopausal endometriosis. Eur J Obstet Gynecol
Reprod Biol. 1980;11:195–200.
28. Bendon CL, Becker CM. Potential mechanisms of postmenopausal endometriosis. Maturitas.
2012;72:214–9.
29. Gemmell LC, Webster KE, Kirtley S, Vincent K, Zondervan KT, Becker CM. The manage-
ment of menopause in women with a history of endometriosis: a systematic review. Hum
Reprod Update. 2017;23:481–500.
30. Cope AG, VanBuren WM, Sheedy SP. Endometriosis in the postmenopausal female: clinical
presentation, imaging features, and management. Abdom Radiol. 2020;45:1790–9.
31. Streuli I, Gaitzsch H, Wenger J-M, Petignat P. Endometriosis after menopause: physiopathol-
ogy and management of an uncommon condition. Climacteric. 2017;20:138–43.
32. Li J, Liu R, Tang S, et al. Impact of endometriosis on risk of ovarian, endometrial and cervical
cancers: a meta-analysis. Arch Gynecol Obstet. 2019;299:35–46.
33. Pearce CL, Templeman C, Rossing MA, et al. Association between endometriosis and risk
of histological subtypes of ovarian cancer: a pooled analysis of case-control studies. Lancet
Oncol. 2012;13:385–94.
34. Stern RC, Dash R, Bentley RC, Snyder MJ, Haney AF, Robboy SJ. Malignancy in endome-
triosis: frequency and comparison of ovarian and extraovarian types. Int J Gynecol Pathol.
2001;20:133–9.
35. Melin A, Sparén P, Persson I, Bergqvist A. Endometriosis and the risk of cancer with special
emphasis on ovarian cancer. Hum Reprod. 2006;21:1237–42.
36. Gougoutas CA, Siegelman ES, Hunt J, Outwater EK. Pelvic endometriosis: various manifes-
tations and MR imaging findings. AJR Am J Roentgenol. 2000;175:353–8.
37. Siegelman ES, Oliver ER. MR imaging of endometriosis: ten imaging pearls. Radiographics.
2012;32:1675–91.
38. Cumiskey J, Whyte P, Kelehan P, Gibbons D. A detailed morphologic and immunohistochem-
ical comparison of pre- and postmenopausal endometriosis. J Clin Pathol. 2008;61:455–9.
39. Woodward PJ, Sohaey R, Mezzetti TP. Endometriosis: radiologic-pathologic correlation.
Radiographics. 2001;21:193–216; questionnaire 288-294.
40. Bazot M, Bharwani N, Huchon C, et al. European society of urogenital radiology (ESUR)
guidelines: MR imaging of pelvic endometriosis. Eur Radiol. 2017;27:2765–75.
41. Schreiter V, Kinkel K. Endometriosis. In: Forstner R, Cunha TM, Hamm B, editors. MRI CT
female pelvis. Cham: Springer International; 2019. p. 325–41.
42. Jung SI. Ultrasonography of ovarian masses using a pattern recognition approach.
Ultrasonography. 2015;34:173–82.
43. Patel MD, Feldstein VA, Chen DC, Lipson SD, Filly RA. Endometriomas: diagnostic perfor-
mance of US. Radiology. 1999;210:739–45.
44. Jain KA. Prospective evaluation of adnexal masses with endovaginal gray-scale and duplex
and color Doppler US: correlation with pathologic findings. Radiology. 1994;191:63–7.
45. McDermott S, Oei TN, Iyer VR, Lee SI. MR imaging of malignancies arising in endometrio-
mas and extraovarian endometriosis. Radiographics. 2012;32:845–63.
46. Khashper A, Addley HC, Abourokbah N, Nougaret S, Sala E, Reinhold C. T2-hypointense
adnexal lesions: an imaging algorithm. Radiographics. 2012;32:1047–64.
47. Vargas HA, Barrett T, Sala E. MRI of ovarian masses. J Magn Reson Imaging. 2013;37:265–81.
11 The Female Urogenital System in Geriatric Patients 313
48. Outwater EK, Siegelman ES, Hunt JL. Ovarian teratomas: tumor types and imaging charac-
teristics. Radiographics. 2001;21:475–90.
49. Foti PV, Farina R, Palmucci S, et al. Endometriosis: clinical features, MR imaging findings
and pathologic correlation. Insights Imaging. 2018;9:149–72.
50. Dhanda S, Thakur M, Kerkar R, Jagmohan P. Diffusion-weighted imaging of gynecologic
tumors: diagnostic pearls and potential pitfalls. Radiographics. 2014;34:1393–416.
51. Tanaka YO, Okada S, Yagi T, Satoh T, Oki A, Tsunoda H, Yoshikawa H. MRI of endometri-
otic cysts in association with ovarian carcinoma. AJR Am J Roentgenol. 2010;194:355–61.
52. Timmerman D, Planchamp F, Bourne T, et al. ESGO/ISUOG/IOTA/ESGE consen-
sus statement on preoperative diagnosis of ovarian tumors. Ultrasound Obstet Gynecol.
2021;58:148–68.
53. Jacobs I, Oram D, Fairbanks J, Turner J, Frost C, Grudzinskas JG. A risk of malignancy
index incorporating CA 125, ultrasound and menopausal status for the accurate preoperative
diagnosis of ovarian cancer. Br J Obstet Gynaecol. 1990;97:922–9.
54. Moore RG, Brown AK, Miller MC, et al. The use of multiple novel tumor biomark-
ers for the detection of ovarian carcinoma in patients with a pelvic mass. Gynecol Oncol.
2008;108:402–8.
55. Amor F, Vaccaro H, Alcázar JL, León M, Craig JM, Martinez J. Gynecologic imaging report-
ing and data system: a new proposal for classifying adnexal masses on the basis of sono-
graphic findings. J Ultrasound Med. 2009;28:285–91.
56. Andreotti RF, Timmerman D, Benacerraf BR, et al. Ovarian-adnexal reporting lexicon for
ultrasound: a white paper of the ACR ovarian-adnexal reporting and data system committee.
J Am Coll Radiol. 2018;15:1415–29.
57. Ameye L, Timmerman D, Valentin L, et al. Clinically oriented three-step strategy for assess-
ment of adnexal pathology. Ultrasound Obstet Gynecol. 2012;40:582–91.
58. Timmerman D, Valentin L, Bourne TH, Collins WP, Verrelst H, Vergote I, International
Ovarian Tumor Analysis (IOTA) Group. Terms, definitions and measurements to describe the
sonographic features of adnexal tumors: a consensus opinion from the International Ovarian
Tumor Analysis (IOTA) Group. Ultrasound Obstet Gynecol. 2000;16:500–5.
59. Van Calster B, Van Hoorde K, Valentin L, et al. Evaluating the risk of ovarian cancer before
surgery using the ADNEX model to differentiate between benign, borderline, early and
advanced stage invasive, and secondary metastatic tumours: prospective multicentre diagnos-
tic study. BMJ. 2014;349:g5920.
60. Thomassin-Naggara I, Poncelet E, Jalaguier-Coudray A, et al. Ovarian-adnexal reporting
data system magnetic resonance imaging (O-RADS MRI) score for risk stratification of sono-
graphically indeterminate adnexal masses. JAMA Netw Open. 2020;3:e1919896.
61. Timmerman D, Testa AC, Bourne T, et al. Simple ultrasound-based rules for the diagnosis of
ovarian cancer. Ultrasound Obstet Gynecol. 2008;31:681–90.
62. Timmerman D, Ameye L, Fischerova D, et al. Simple ultrasound rules to distinguish between
benign and malignant adnexal masses before surgery: prospective validation by IOTA group.
BMJ. 2010;341:c6839.
63. Forstner R, Thomassin-Naggara I, Cunha TM, Kinkel K, Masselli G, Kubik-Huch R, Spencer
JA, Rockall A. ESUR recommendations for MR imaging of the sonographically indetermi-
nate adnexal mass: an update. Eur Radiol. 2017;27:2248–57.
64. Kubik-Huch RA, Weston M, Nougaret S, Leonhardt H, Thomassin-Naggara I, Horta M,
Cunha TM, Maciel C, Rockall A, Forstner R. European Society of Urogenital Radiology
(ESUR) guidelines: MR imaging of leiomyomas. Eur Radiol. 2018;28:3125–37.
65. Forstner R. CT and MRI in ovarian carcinoma. In: Forstner R, Cunha TM, Hamm B, editors.
MRI CT female pelvis. Cham: Springer International; 2019. p. 287–323.
66. Hennessy BT, Coleman RL, Markman M. Ovarian Cancer Lancet. 2009;374:1371–82.
67. Jung SE, Lee JM, Rha SE, Byun JY, Jung JI, Hahn ST. CT and MR imaging of ovarian tumors
with emphasis on differential diagnosis. Radiographics. 2002;22:1305–25.
68. Seidman JD, Ronnett BM, Shih I-M, Cho KR, Kurman RJ. Epithelial tumors of the ovary. In:
Kurman RJ, Hedrick Ellenson L, Ronnett BM, editors. Blaustein’s pathology of the female
genital tract. Cham: Springer International; 2019. p. 841–966.
314 M. A. Cova et al.
69. Kurman RJ, International Agency for Research on Cancer, World Health Organization, edi-
tors. WHO classification of tumours of female reproductive organs. 4th ed. Lyon: International
Agency for Research on Cancer; 2014.
70. Jeong YY, Outwater EK, Kang HK. Imaging evaluation of ovarian masses. Radiographics.
2000;20:1445–70.
71. Foti PV, Attinà G, Spadola S, et al. MR imaging of ovarian masses: classification and dif-
ferential diagnosis. Insights Imaging. 2016;7:21–41.
72. Outwater EK, Huang AB, Dunton CJ, Talerman A, Capuzzi DM. Papillary projections in
ovarian neoplasms: appearance on MRI. J Magn Reson Imaging. 1997;7:689–95.
73. Thomassin-Naggara I, Daraï E, Cuenod CA, Rouzier R, Callard P, Bazot M. Dynamic
contrast-enhanced magnetic resonance imaging: a useful tool for characterizing ovarian epi-
thelial tumors. J Magn Reson Imaging. 2008;28:111–20.
74. Wasnik AP, Menias CO, Platt JF, Lalchandani UR, Bedi DG, Elsayes KM. Multimodality
imaging of ovarian cystic lesions: review with an imaging based algorithmic approach. World
J Radiol. 2013;5:113–25.
75. Schlattau A, Cunha TM, Forstner R. Adnexal masses: benign ovarian lesions and character-
ization. In: Forstner R, Cunha TM, Hamm B, editors. MRI CT female pelvis. Cham: Springer
International; 2019. p. 241–71.
76. Laurent P-E, Thomassin-Piana J, Jalaguier-Coudray A. Mucin-producing tumors of the
ovary: MR imaging appearance. Diagn Interv Imaging. 2015;96:1125–32.
77. Shen DH, Ng TY, Khoo US, Xue WC, Cheung AN. Pseudomyxoma peritonei—a heterog-
enous disease. Int J Gynaecol Obstet. 1998;62:173–82.
78. Tang YZ, Liyanage S, Narayanan P, Sahdev A, Sohaib A, Singh N, Rockall A. The MRI fea-
tures of histologically proven ovarian cystadenofibromas-an assessment of the morphological
and enhancement patterns. Eur Radiol. 2013;23:48–56.
79. Tanaka YO, Yoshizako T, Nishida M, Yamaguchi M, Sugimura K, Itai Y. Ovarian car-
cinoma in patients with endometriosis: MR imaging findings. AJR Am J Roentgenol.
2000;175:1423–30.
80. Shaaban AM. Ovarian endometrioid carcinoma. Diagn Imaging Gynecol. 2014.
81. Maniar KP, Vang R. Germ cell tumors of the ovary. In: Kurman RJ, Hedrick Ellenson L,
Ronnett BM, editors. Blaustein’s pathology of the female genital tract. Cham: Springer
International; 2019. p. 1047–124.
82. Boussios S, Attygalle A, Hazell S, Moschetta M, McLachlan J, Okines A, Banerjee
S. Malignant ovarian germ cell tumors in postmenopausal patients: the Royal Marsden
Experience and literature review. Anticancer Res. 2015;35:6713–22.
83. Smith HO, Berwick M, Verschraegen CF, Wiggins C, Lansing L, Muller CY, Qualls
CR. Incidence and survival rates for female malignant germ cell tumors. Obstet Gynecol.
2006;107:1075–85.
84. Ozgur T, Atik E, Silfeler DB, Toprak S. Mature cystic teratomas in our series with review of
the literature and retrospective analysis. Arch Gynecol Obstet. 2012;285:1099–101.
85. Saba L, Guerriero S, Sulcis R, Virgilio B, Melis G, Mallarini G. Mature and immature ovarian
teratomas: CT, US and MR imaging characteristics. Eur J Radiol. 2009;72:454–63.
86. Tamai K, Koyama T, Saga T, Kido A, Kataoka M, Umeoka S, Fujii S, Togashi K. MR features
of physiologic and benign conditions of the ovary. Eur Radiol. 2006;16:2700–11.
87. Wei F, Jiang Z, Yan C. Analysis of 20 mature ovarian cystic teratoma cases in postmeno-
pausal women. Chin Med J (Engl). 2001;114:137–8.
88. Park SB, Kim JK, Kim K-R, Cho K-S. Imaging findings of complications and unusual mani-
festations of ovarian teratomas. Radiographics. 2008;28:969–83.
89. Outwater EK, Wagner BJ, Mannion C, McLarney JK, Kim B. Sex cord-stromal and steroid
cell tumors of the ovary. Radiographics. 1998;18:1523–46.
90. Jung SE, Rha SE, Lee JM, Park SY, Oh SN, Cho KS, Lee EJ, Byun JY, Hahn ST. CT and MRI
findings of sex cord-stromal tumor of the ovary. AJR Am J Roentgenol. 2005;185:207–15.
91. Schumer ST, Cannistra SA. Granulosa cell tumor of the ovary. J Clin Oncol Off J Am Soc
Clin Oncol. 2003;21:1180–9.
11 The Female Urogenital System in Geriatric Patients 315
92. Staats PN, Young RH. Sex cord-stromal, steroid cell, and other ovarian tumors with endo-
crine, paraendocrine, and paraneoplastic manifestations. In: Kurman RJ, Hedrick Ellenson
L, Ronnett BM, editors. Blaustein’s pathology of the female genital tract. Cham: Springer
International; 2019. p. 967–1045.
93. Troiano RN, Lazzarini KM, Scoutt LM, Lange RC, Flynn SD, McCarthy S. Fibroma and
fibrothecoma of the ovary: MR imaging findings. Radiology. 1997;204:795–8.
94. Chang HC, Bhatt S, Dogra VS. Pearls and pitfalls in diagnosis of ovarian torsion.
Radiographics. 2008;28:1355–68.
95. Cohen A, Solomon N, Almog B, Cohen Y, Tsafrir Z, Rimon E, Levin I. Adnexal torsion
in postmenopausal women: clinical presentation and risk of ovarian malignancy. J Minim
Invasive Gynecol. 2017;24:94–7.
96. Baines PA, Allen GM. Pelvic pain and menstrual related illnesses. Emerg Med Clin North
Am. 2001;19:763–80.
97. Rha SE, Byun JY, Jung SE, Jung JI, Choi BG, Kim BS, Kim H, Lee JM. CT and MR imaging
features of adnexal torsion. Radiographics. 2002;22:283–94.
98. Dawood MT, Naik M, Bharwani N, Sudderuddin SA, Rockall AG, Stewart VR. Adnexal tor-
sion: review of radiologic appearances. Radiographics. 2021;41:609–24.
99. Mashiach R, Melamed N, Gilad N, Ben-Shitrit G, Meizner I. Sonographic diagnosis of ovar-
ian torsion: accuracy and predictive factors. J Ultrasound Med. 2011;30:1205–10.
100. Vandermeer FQ, Wong-You-Cheong JJ. Imaging of acute pelvic pain. Clin Obstet Gynecol.
2009;52:2–20.
101. Moribata Y, Kido A, Yamaoka T, Mikami Y, Himoto Y, Kataoka M, Fujimoto K, Konishi I,
Togashi K. MR imaging findings of ovarian torsion correlate with pathological hemorrhagic
infarction. J Obstet Gynaecol Res. 2015;41:1433–9.
102. Jackson SL, Soper DE. Pelvic inflammatory disease in the postmenopausal woman. Infect
Dis Obstet Gynecol. 1999;7:248–52.
103. Wilbur AC, Aizenstein RI, Napp TE. CT findings in tuboovarian abscess. AJR Am J
Roentgenol. 1992;158:575–9.
104. Bennett GL, Slywotzky CM, Giovanniello G. Gynecologic causes of acute pelvic pain: spec-
trum of CT findings. Radiographics. 2002;22:785–801.
105. Kim SH, Kim SH, Yang DM, Kim KA. Unusual causes of tubo-ovarian abscess: CT and MR
imaging findings. Radiographics. 2004;24:1575–89.
106. Dohke M, Watanabe Y, Okumura A, Amoh Y, Hayashi T, Yoshizako T, Yasui M, Nakashita
S, Nakanishi J, Dodo Y. Comprehensive MR imaging of acute gynecologic diseases.
Radiographics. 2000;20:1551–66.
107. Rezvani M, Shaaban AM. Fallopian tube disease in the nonpregnant patient. Radiographics.
2011;31:527–48.
108. Fogel SR, Slasky BS. Sonography of Nabothian cysts. AJR Am J Roentgenol.
1982;138:927–30.
109. Kier R. Nonovarian gynecologic cysts: MR imaging findings. AJR Am J Roentgenol.
1992;158:1265–9.
110. Sudderuddin S, Helbren E, Telesca M, Williamson R, Rockall A. MRI appearances of benign
uterine disease. Clin Radiol. 2014;69:1095–104.
111. Van den Bosch T, de Bruijn AM, de Leeuw RA, Dueholm M, Exacoustos C, Valentin L,
Bourne T, Timmerman D, Huirne J. Sonographic classification and reporting system for diag-
nosing adenomyosis. Ultrasound Obstet Gynecol. 2019;53:576–82.
112. Cova MA, Marrocchio C, Bozzato AM. Female genital system. MRI Abdomen Tech. Imaging
Find; 2021.
113. Kröncke TJ. Benign uterine lesions. In: Hamm B, Forstner R, editors. MRI CT female pelvis.
Berlin, Heidelberg: Springer; 2007. p. 61–100.
114. Agostinho L, Cruz R, Osório F, Alves J, Setúbal A, Guerra A. MRI for adenomyosis: a picto-
rial review. Insights Imaging. 2017;8:549–56.
115. Takeuchi M, Matsuzaki K. Adenomyosis: usual and unusual imaging manifestations, pitfalls,
and problem-solving MR imaging techniques. Radiographics. 2011;31:99–115.
316 M. A. Cova et al.
116. Reinhold C, Tafazoli F, Wang L. Imaging features of adenomyosis. Hum Reprod Update.
1998;4:337–49.
117. Kinkel K, Ascher SM, Reinhold C. Benign disease of the uterus. In: Diseases of the abdomen
and pelvis 2018-2021: diagnostic imaging—IDKD book. Cham: Springer; 2018.
118. Reinhold C, Tafazoli F, Mehio A, Wang L, Atri M, Siegelman ES, Rohoman L. Uterine
adenomyosis: endovaginal US and MR imaging features with histopathologic correlation.
Radiographics. 1999;19:S147–60.
119. Kröncke TJ. Benign uterine lesions. In: Forstner R, Cunha TM, Hamm B, editors. MRI CT
female pelvis. Cham: Springer International; 2017. p. 77–116.
120. Peterson M, Dabbs DJ, Weidner N. Uterus. In: Modern surgical pathology. Philadelphia: WB
Saunders; 2009. p. 1295–340.
121. Robertson M, Scott P, Ellwood DA, Low S. Endocervical polyp in pregnancy: gray scale and
color Doppler images and essential considerations in pregnancy. Ultrasound Obstet Gynecol.
2005;26:583–4.
122. Otero-García MM, Mesa-Álvarez A, Nikolic O, Blanco-Lobato P, Basta-Nikolic M, de
Llano-Ortega RM, Paredes-Velázquez L, Nikolic N, Szewczyk-Bieda M. Role of MRI in
staging and follow-up of endometrial and cervical cancer: pitfalls and mimickers. Insights
Imaging. 2019;10:19.
123. Deshmukh SP, Gonsalves CF, Guglielmo FF, Mitchell DG. Role of MR imaging of uterine
leiomyomas before and after embolization. Radiographics. 2012;32:E251–81.
124. Wilde S, Scott-Barrett S. Radiological appearances of uterine fibroids. Indian J Radiol
Imaging. 2009;19:222–31.
125. DeMulder D, Ascher SM. Uterine Leiomyosarcoma: can MRI differentiate Leiomyosarcoma
from benign leiomyoma before treatment? AJR Am J Roentgenol. 2018;211:1405–15.
126. Nougaret S, Sbarra M, Robbins J. Imaging Spectrum of benign uterine disease and treatment
options. Radiol Clin North Am. 2020;58:239–56.
127. Bharambe BM, Deshpande KA, Surase SG, Ajmera AP. Malignant transformation of leio-
myoma of uterus to leiomyosarcoma with metastasis to ovary. J Obstet Gynaecol India.
2014;64:68–9.
128. Leibsohn S, d’Ablaing G, Mishell DR, Schlaerth JB. Leiomyosarcoma in a series of hysterec-
tomies performed for presumed uterine leiomyomas. Am J Obstet Gynecol. 1990;162:968–74;
discussion 974–976.
129. Santos P, Cunha TM. Uterine sarcomas: clinical presentation and MRI features. Diagn Interv
Radiol Ank Turk. 2015;21:4–9.
130. Morice P, Leary A, Creutzberg C, Abu-Rustum N, Darai E. Endometrial cancer. Lancet.
2016;387:1094–108.
131. Hedrick Ellenson L, Ronnett BM, Soslow RA, Lastra RR, Kurman RJ. Endometrial carci-
noma. In: Kurman RJ, Hedrick Ellenson L, Ronnett BM, editors. Blaustein’s pathology of the
female genital tract. Cham: Springer International; 2019. p. 473–533.
132. Amant F, Mirza MR, Koskas M, Creutzberg CL. Cancer of the corpus uteri. Int J Gynaecol
Obstet. 2018;143(Suppl 2):37–50.
133. Horta M, Cunha TM. Endometrial cancer. In: Forstner R, Cunha TM, Hamm B, editors. MRI
CT female pelvis. Cham: Springer International; 2017. p. 179–208.
134. Aune D, Sen A, Vatten LJ. Hypertension and the risk of endometrial cancer: a systematic
review and meta-analysis of case-control and cohort studies. Sci Rep. 2017;7:44808.
135. McCartney CR, Marshall JC. Polycystic ovary syndrome. N Engl J Med. 2016;375:1398–9.
136. Nougaret S, Horta M, Sala E, et al. Endometrial cancer MRI staging: updated guidelines of
the European Society of Urogenital Radiology. Eur Radiol. 2019;29:792–805.
137. Freeman SJ, Aly AM, Kataoka MY, Addley HC, Reinhold C, Sala E. The revised FIGO
staging system for uterine malignancies: implications for MR imaging. Radiographics.
2012;32:1805–27.
138. Arbyn M, Weiderpass E, Bruni L, de Sanjosé S, Saraiya M, Ferlay J, Bray F. Estimates
of incidence and mortality of cervical cancer in 2018: a worldwide analysis. Lancet Glob
Health. 2020;8:e191–203.
11 The Female Urogenital System in Geriatric Patients 317
139. Collettini F, Hamm B. Cervical cancer. In: Forstner R, Cunha TM, Hamm B, editors. MRI CT
female pelvis. Cham: Springer International; 2019. p. 117–77.
140. Pirog EC, Wright TC, Ronnett BM, Kurman RJ. Carcinoma and other tumors of the cer-
vix. In: Kurman RJ, Hedrick Ellenson L, Ronnett BM, editors. Blaustein’s pathology of the
female genital tract. Cham: Springer International; 2019. p. 315–74.
141. Hinkula M, Pukkala E, Kyyrönen P, Laukkanen P, Koskela P, Paavonen J, Lehtinen M,
Kauppila A. A population-based study on the risk of cervical cancer and cervical intraepi-
thelial neoplasia among grand multiparous women in Finland. Br J Cancer. 2004;90:1025–9.
142. Li S, Wen X. Seropositivity to herpes simplex virus type 2, but not type 1 is associated with
cervical cancer: NHANES (1999-2014). BMC Cancer. 2017;17:726.
143. Burghardt E, Ostör AG. Site and origin of squamous cervical cancer: a histomorphologic
study. Obstet Gynecol. 1983;62:117–27.
144. Balleyguier C, Sala E, Da Cunha T, et al. Staging of uterine cervical cancer with MRI: guide-
lines of the European Society of Urogenital Radiology. Eur Radiol. 2011;21:1102–10.
145. Haldorsen IS, Lura N, Blaakær J, Fischerova D, Werner HMJ. What is the role of imaging at
primary diagnostic work-up in uterine cervical cancer? Curr Oncol Rep. 2019;21:77.
146. Bhatla N, Berek JS, Cuello Fredes M, et al. Revised FIGO staging for carcinoma of the cervix
uteri. Int J Gynaecol Obstet. 2019;145:129–35.
147. Edge SB, Compton CC. The American joint committee on cancer: the 7th edition of the
AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 2010;17:1471–4.
148. Adams TS, Rogers LJ, Cuello MA. Cancer of the vagina: 2021 update. Int J Gynaecol Obstet.
2021;155(Suppl 1):19–27.
149. Hacker NF, Eifel PJ, van der Velden J. Cancer of the vagina. Int J Gynaecol Obstet.
2012;119(Suppl 2):S97–9.
150. Adhikari P, Vietje P, Mount S. Premalignant and malignant lesions of the vagina. Diagn
Histopathol. 2017;23:28–34.
151. Gardner CS, Sunil J, Klopp AH, Devine CE, Sagebiel T, Viswanathan C, Bhosale
PR. Primary vaginal cancer: role of MRI in diagnosis, staging and treatment. Br J Radiol.
2015;88:20150033.
152. Parikh JH, Barton DPJ, Ind TEJ, Sohaib SA. MR imaging features of vaginal malignancies.
Radiographics. 2008;28:49–63; quiz 322.
153. Dixit S, Singhal S, Baboo HA. Squamous cell carcinoma of the vagina: a review of 70 cases.
Gynecol Oncol. 1993;48:80–7.
154. Amin MB, American Joint Committee on Cancer, American Cancer Society. In: Amin MB,
Edge SB, Gress DM, Meyer LR, editors. AJCC cancer staging manual. 8th ed. Chicago IL:
American Joint Committee on Cancer, Springer; 2017.
155. FIGO Committee on Gynecologic Oncology. Current FIGO staging for cancer of the
vagina, fallopian tube, ovary, and gestational trophoblastic neoplasia. Int J Gynaecol Obstet.
2009;105:3–4.
156. Al-Kurdi M, Monaghan JM. Thirty-two years experience in management of primary tumours
of the vagina. Br J Obstet Gynaecol. 1981;88:1145–50.
157. Davis KP, Stanhope CR, Garton GR, Atkinson EJ, O’Brien PC. Invasive vaginal carcinoma:
analysis of early-stage disease. Gynecol Oncol. 1991;42:131–6.
158. Chyle V, Zagars GK, Wheeler JA, Wharton JT, Delclos L. Definitive radiotherapy for car-
cinoma of the vagina: outcome and prognostic factors. Int J Radiat Oncol Biol Phys.
1996;35:891–905.
159. Rogers LJ, Cuello MA. Cancer of the vulva. Int J Gynaecol Obstet. 2018;143(Suppl 2):4–13.
160. Tan A, Bieber AK, Stein JA, Pomeranz MK. Diagnosis and management of vulvar cancer: a
review. J Am Acad Dermatol. 2019;81:1387–96.
161. Alkatout I, Schubert M, Garbrecht N, Weigel MT, Jonat W, Mundhenke C, Günther V. Vulvar
cancer: epidemiology, clinical presentation, and management options. Int J Womens Health.
2015;7:305–13.
162. Serrado MA, Horta M, Cunha TM. State of the art in vulvar cancer imaging. Radiol Bras.
2019;52:316–24.
318 M. A. Cova et al.
With the increase in life expectancy, degenerative pathologies of the spine are
becoming more and more common during clinical practice, with a high medical and
socioeconomic impact.
Degenerative changes in the spine are usually associated with back pain and/or
symptoms of neural structure compression, caused by anatomical and biomechani-
cal alterations.
Imaging the degenerative spine might be quite challenging. In fact, the presence
of symptoms and pain does not always correspond to abnormal findings at imaging,
as well as the presence of degenerative imaging changes does not necessarily cor-
relate with the presence and the severity of pain. In addition, sometimes it is not
easy to discriminate between imaging findings of normal aging and degenerative
pathology.
The spine is a multiarticular structure, which enables motions in different direc-
tions and absorbs multidirectional loads. Specifically, two adjacent vertebrae, the
intervertebral disc, spinal ligaments, and facet joints between them constitute a
functional spinal unit [1].
The main functions of the spine are to provide structural support, enable trunk
movement, and protect the neural elements [2].
Approximately 70% of applied axial compression is transmitted by the vertebral
body and the intervertebral disc, with the remaining 30% of the load being distrib-
uted through the facet joints (Fig. 12.1) [3].
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 319
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_12
320 F. Serpi et al.
12.2.1.2 Imaging
At imaging, normal nucleus pulposus has a hyperintense signal on T2-weighed
images (WI), which directly correlates with proteoglycan and subsequently water
concentration. Disc dehydration corresponds to progressive signal loss on T2-WI
and loss of disc height [12].
Pfirrmann et al. developed a grading system for lumbar disc degeneration accord-
ing to magnetic resonance imaging (MRI) T2-WI, discal structure, distinction
between nucleus pulposus and annulus fibrosus and disc height (Table 12.1) [13].
Other imaging signs that can be associated with disc degenerations are: the vac-
uum phenomenon (Fig. 12.2), accumulation of nitrogen within the disc, which can
be visible both on X-ray and computed tomography (CT) as the presence of gas
within the disc and represented as a signal void on both T1 and T2-WI at MRI [14,
15]; intradiscal fluid accumulation, with hyperintense signal at T2-WI, which can
be associated with endplates degeneration, mimicking early spondylodiscitis [15];
intradiscal calcification (Fig. 12.2), which frequently involves the annulus fibrosus
and is located in the lower part of the thoracic spine [16].
322 F. Serpi et al.
a b
Fig. 12.2 Signs of disc degeneration. The vacuum phenomenon is due to the accumulation of
nitrogen within the disc (arrows), represented as air density within the disc in CT (a). Intradiscal
calcification typically involve the annulus fibrosus, as in this picture (arrow) at the level of the
posterior fibers of the intervertebral disc L5-S1 (b)
Fig. 12.3 Circumferential fissure of the posterior annular fibers at L4-L5. It consists of the separa-
tion or delamination of annular transverse fibers parallel to the peripheral contour of the disc (arrows)
Fig. 12.5 Bulging L4-L5. A bulging refers to the presence of disc tissue extending beyond the
edges of the ring apophyses, involving more than 25% of the circumference of the disc on axial
plane and typically extending a relatively short distance
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 325
Fig. 12.6 A bulging is the presence of disc tissue extending beyond the edges of the ring apophy-
ses more than 25% of the circumference of the disc and typically extends a relatively short dis-
tance. Disc herniation refers to the extension of disc material less than 25% of the circumference
of the disc
a b
c d
Fig. 12.7 Protrusion L5-S1 (arrow), the distance between the edges of the disc herniation is less
than the distance between the edges of the base (a, b). Extrusion L5-S1 (arrow), the distance
between the edges of the disc material is greater than the distance at the base, extending in the right
subarticular space with possible conflict with the nerve root of S1 (c, d)
AL GRAWANY
326 F. Serpi et al.
a b
c d
Fig. 12.8 Contained left subarticular hernia at L5-S1. L5-S1 (arrows), without transligament
involvement (a, b). Uncontained right subarticular hernia at L5-S1 (arrows), with transligament
caudal migration (c, d)
distance at the base [10]. Extruded hernia on the sagittal plane sometimes causes
the posterior longitudinal ligament to tent, which often causes neurological
symptoms and pain [3].
• Containment: contained or uncontained (Fig. 12.8). A hernia is contained if the
displaced portion is covered by outer annulus fibers and/or the posterior longitu-
dinal ligament, or uncontained in the absence of posterior covering. Referring
specifically to the posterior longitudinal ligament, some authors have distin-
guished displaced disc material as subligamentous, extraligamentous, transliga-
mentous, or perforated. The term subligamentous is favored as an equivalent to
contained [10]. Due to limitation of CT and MRI, it is not always possible to
discriminate between contained and uncontained herniation. Nevertheless, if the
posterior margin of the herniated disc is smooth on axial plane, it is likely con-
tained, whereas if the margins are irregular, it is likely uncontained. CT discog-
raphy does not always allow to distinguish whether the herniated components of
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 327
a disc are contained, but only whether there is a communication between the disc
space and the vertebral canal [10].
• Continuity: Extruded discs in which all continuity with the disc of origin is lost
may be further characterized as “sequestrated.” A sequestrated hernia can be
either contained (subligamentous) or not. More generally, disc material displaced
away from the site of extrusion, in either sagittal or axial plane, may be character-
ized as “migrated.” Migration refers to the position of the displaced disc material,
rather than to its continuity with the disc of origin; therefore, it is not synonymous
with sequestration. A migrated disc can be sequestrated or not (Fig. 12.9) [10].
• Volume and Composition: Due to anatomical interindividual differences, there is
no universal classification to assess spinal canal involvement. A simple scheme
is to assess spinal canal compromission on axial plane. Less than one-third is
considered “mild,” from one- to two-third “moderate,” and more than two-third
“severe” [10]. Acute disc herniation has usually high water content with hyper-
intense signal on T2-WI and tends to dehydrated and shrink over time, which
leads to progressively volume decrease and loss of signal in T2-WI.
a b
Fig. 12.9 L4-L5 hernia (arrow), caudally migrated without sequestrum (a). L4-L5 hernia, crani-
ally migrated with sequestrum (arrow). The extruded material has lost the continuity with the disc
of origin (b)
328 F. Serpi et al.
FORAMINAL
SUBARTICULAR
CENTRAL
SUPRAPEDICULAR
PEDICULAR
INFRAPEDICULAR
DISCAL
patients with symptoms and pain can be divided into acute (up to 4 weeks), sub-
acute (between 4 and 12 weeks), and chronic (more than 12 weeks) [20].
• Vascular complications: They develop secondary to acute or chronic com-
pression of the vertebral artery or medullary segmental arteries feeding the
spinal cord (large cervical radiculomedullary at C5–C7; dominant radiculo-
medullary artery at T4–T5; the artery of Adamkiewicz located at T10 and the
additional radiculomedullary artery of Desproges-Gotteron arises at L4–L5),
which may cause a severe neurological deficit and also may require interven-
tion [3].
• Focal complications: They can occur because of chronic persistent inflamma-
tion, such as epidural scarring, which may limit nerve roots passage through
foramina and may cause nerve root tethering. This process is virtually impossi-
ble to identify at imaging [3]. Intradural herniation (very rare) and epidural vein
varicosis are other possible focal complications [3].
Fig. 12.12 Modic classification of endplates degenerative changes. Modic I: bone marrow edema
and vascularized fibrous tissue; Modic II: yellow marrow metaplasia; Modic III: sclerotic reaction
quickly, with diffuse infiltrate of pathogens and edema of the marrow, without a
well-defined border and a claw sign [3, 23, 24].
Facet joints are true synovial joints, presented at every intervertebral level, except at
C1-C2. They contribute to spinal movement and, as any other synovial articulation,
they can be subjected to degenerative disease. Although facet joint osteoarthritis
may occur independently and could be a source of pain on its own, it typically rep-
resents a secondary process that is associated with disc degeneration and loss of disc
height [3]. The consequently altered motion and increased stress on the facet joint
result in arthrosis, osteophytes, synovial cysts, and craniocaudal subluxation
(Fig. 12.13) [3]. Moreover, with aging, paraspinal muscles mass decreases, which
contributes to facet joint osteoarthrosis by allowing poorly controlled segmental
motion [25]. Joint osteoarthrosis can be classified based on osteophytes formation
and joint space narrowing [3, 26].
Degenerative synovial changes can also produce synovial cysts (Fig. 12.14),
with the majority located at the lumbar (L4-L5) level. They are usually hyperintense
on T2-WI but can also be hyperintense at T1-WI if they contain hemorrhagic or
proteinaceous components [3]. Clinically speaking, hypertrophic facet joint osteo-
arthritis can determine stenosis of the canal and of the preforaminal or foraminal
space, leading to symptoms related also to neural elements compression. Moreover,
facet joint osteoarthritis plays an important role in spinal instability and anterior
332 F. Serpi et al.
Ligamentum flavum is also called yellow ligament, due to the high content of yel-
low elastin. It is composed of two adjacent laminae, extending from the second
cervical vertebra to the first sacral vertebra, forming the posterior boundary of the
spinal canal [3]. Degenerative disc alterations and herniation, together with facet
joint osteoarthritis, determine abnormal movements and instability, which is a
potential trigger for ligamentum flavum thickening (Fig. 12.16). The term “hyper-
trophy” should be discouraged because the degenerative process is not character-
ized by an enlargement of cellular elements, but by a degeneration of elastic fibers
and an accumulation of collagen due to chronic inflammation; this process deter-
mines corrugation of the ligament and predisposes to calcification (Fig. 12.17) [28].
However, this can contribute to reduction of the diameter of the spinal canal and
represents one of the elements that can participate in the spinal canal stenosis [3].
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 333
a b
c d
Fig. 12.14 Left facet joint fluid accumulation at L4-L5, arrows (a, b), with initial formation of a
synovial cyst. In the axial plane, facet fluid is also accumulated on the right side (b) but in less
quantity. Left facet joint synovial cyst at L4-L5 (c, d), extending in the spinal canal (arrows)
Fig. 12.15 L4-L5 anterior degenerative spondylolisthesis (star) with facet joints sublux-
ation (arrow)
Fig. 12.16 Ligamentum flavum thickening (arrows). This contributes to the reduction of spinal
canal size
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 335
Fig. 12.18 Degenerative changes of the interspinous processes: interspinous bursitis. L3-L4
small fluid collection both on STIR sequence of the left and T2 weighted sequence on the right
(arrows)
AL GRAWANY
336 F. Serpi et al.
Due to degenerative changes of the disc, facet joints, and ligamentous apparatus, the
ability to maintain the anatomical alignment of the functional spinal unit, at static
position and/or during movements, decreases. This results in functional instability
and degenerative spondylolisthesis. It typically occurs at lumbar or cervical levels
and is virtually absent in the thoracic spine, thanks to costovertebral joints which act
as a further stabilizer.
Instability can be defined as an abnormal response to applied loads characterized
kinematically by abnormal movement beyond normal constraints [30].
Spondylolisthesis refers to forward slippage of a vertebra on the subjacent one in
the sagittal plane. Backward vertebral slippage, a type of spondylolisthesis, has
been called retrolisthesis [14].
The process of degenerative instability is divided into three phases: early dys-
function, instability, and stabilization [31]. The first phase (early dysfunction) is
characterized by initial and reversible anatomical modifications induced by altered
axial load. In the second phase (instability), anatomical changes progress with disc
height reduction, capsule-ligament laxity, and facet joint osteoarthrosis. Finally, in
the third phase (stabilization), new biomechanical constraints occur induced by ana-
tomical changes, such as osteophytes and intervertebral space reduction, which lead
to a new spinal mechanical stabilization, however at the cost of movement reduction
[32]. In the third stage sometimes spondylolisthesis has already occurred. In fact,
the radiologic observation of degenerative spondylolisthesis does not necessarily
imply intervertebral instability at the time of imaging because a new stabilization
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 337
may have already occurred [14]. Degenerative instability consists of pure motion
dysfunctional syndrome with no or minimal anatomical changes, undetectable on
imaging (microinstability), or overt instability, which can be radiologically detect-
able [33]. As persistent uni- or multisegmental instability progresses, it leads to
rotational and translational vertebral subluxation, resulting in degenerative spondy-
lolisthesis, which may be stable or unstable [3].
Conventional MRI and CT performed in the prone position provides limited
information on the functional status of the affected segment as spondylolisthesis
with instability may “self-reduce” without a normal axial load [3]. Therefore, diag-
nosis is based both on direct and indirect signs of instability, such as joint facets
fluid, facet synovial cysts, interspinous fluid, facet joints hypertrophy, and intradis-
cal vacuum phenomenon (Fig. 12.20) [3].
Functional flexion/extension radiographs are considered the gold standard for
diagnosing the presence of degenerative instability in the setting of spondylolisthe-
sis [34]. For lumbar spine spondylolisthesis, the Meyerding classification is a com-
mon method for grading anterior vertebral spondylolisthesis, based on the ratio of
the overhanging part of the superior vertebral body to the anteroposterior length of
the adjacent inferior vertebral body (Figs. 12.21 and 12.22).
For the lumbar spine, on flexion-extension radiographs, values of 10° for sagittal
rotation and 4 mm for sagittal translation are typically used to infer instability [14].
For the cervical spine, a slippage of 3 mm on functional radiographs is considered a
reliable cut-off [3].
Degenerative spondylolisthesis is an important factor that can contribute to spi-
nal canal stenosis, which is defined as a decrease in the area that can affect the spinal
canal, the lateral recesses, or neural foramina, compressing neural and vascular
structure, thus resulting in various degrees of clinical disabilities [3, 16].
Degenerative spinal canal stenosis is considered a multifactorial condition, rather
than an answer to a single insult. In fact, there are four major factors that can con-
tribute to spinal canal stenosis that should be always checked carefully: disc hernia-
tion, hypertrophic facet joint osteoarthrosis, ligamentum flavum hypertrophy, and
spondylolisthesis (Fig. 12.23) [3].
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 339
Fig. 12.23 Major factors that contribute to spinal canal stenosis, which is usually the result of a
multifactorial condition
and occupy the majority of the dural sac area. Grade B, moderate stenosis, includes
cases where the rootlets occupy the entire dural sac, but can still be individualized.
Grade C (Fig. 12.24), severe stenosis, refers to cases where no rootlets can be rec-
ognized, with the dural sac demonstrating a homogeneous gray signal with no vis-
ible CSF signal, but epidural fat present posteriorly. Grade D (Fig. 12.25), extreme
stenosis, refers to no rootlets being recognizable and no epidural fat posteriorly [36].
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 341
Conventional radiography usually represents the first level examination for the
spine, allowing a good assessment of spinal alignment, vertebral bodies morpholo-
gies, calcifications, and the vacuum phenomenon. However, it has a marginal role
due to technical limitations, mostly due to air and other structures overlap and the
impossibility to study bone marrow, neural structures, and soft tissue. The standard
projections are the anteroposterior and latero-lateral view. In addition, the oblique
view is used for the assessment of foramens and, eventually, spondylolysis (scotty
dog sign) [32]. Moreover, functional flexion/extension radiographs are considered
the gold standard for diagnosing the presence of degenerative instability in the set-
ting of spondylolisthesis [34].
MRI is the first-choice technique for the study of discs and ligaments changes.
MRI of the spine is generally performed with the patient in supine position. Upright
MRI is also feasible and favored by some groups due to the plausible explanation
that axial loads on the intervertebral disc are reduced in supine position. However,
upright MRI is limited by poor resolution, very limited availability, increased
motion artifacts, and high false-positive findings [37, 38].
References
1. Dupré DA, Cook DJ, Brad Bellotte J, Oh MY, Whiting D, Cheng BC. Disc nucleus forti-
fication for lumbar degenerative disc disease: a biomechanical study. J Neurosurg Spine.
2016;24:708–14.
2. Oxland TR. Fundamental biomechanics of the spine--what we have learned in the past 25
years and future directions. J Biomech. 2016;49:817–32.
3. Kushchayev SV, Glushko T, Jarraya M, Schuleri KH, Preul MC, Brooks ML, Teytelboym
OM. ABCs of the degenerative spine. Insights Imag. 2018;9:253–74.
12 Osteoarthritis in Axial Skeleton in Geriatric Patients 343
4. Bogduk N. Degenerative joint disease of the spine. Radiol Clin N Am. 2012;50:613–28.
5. Inoue N, Espinoza Orías AA. Biomechanics of intervertebral disk degeneration. Orthop Clin
North Am. 2011;42:487–99, vii.
6. Niosi CA, Oxland TR. Degenerative mechanics of the lumbar spine. Spine J. 2004;4:202S–8S.
7. Horst M, Brinckmann P. 1980 Volvo award in biomechanics. Measurement of the distribution
of axial stress on the end-plate of the vertebral body. Spine. 1981;6:217–32.
8. Virk SS, Niedermeier S, Yu E, Khan SN. Adjacent segment disease. Orthopedics.
2014;37:547–55.
9. Rousseau M-A, Lazennec J-Y. Degenerative disease supra- and infra-jacent to fused lumbar
and lumbo-sacral levels. Orthop Traumatol Surg Res. 2016;102:S1–8.
10. Fardon DF, Williams AL, Dohring EJ, Murtagh FR, Gabriel Rothman SL, Sze GK. Lumbar
disc nomenclature: version 2.0: recommendations of the combined task forces of the North
American Spine Society, the American Society of Spine Radiology and the American Society
of Neuroradiology. Spine J. 2014;14:2525–45.
11. Ferguson SJ, Steffen T. Biomechanics of the aging spine. Eur Spine J. 2003;12(Suppl
2):S97–S103.
12. Modic MT, Masaryk TJ, Ross JS, Carter JR. Imaging of degenerative disk disease. Radiology.
1988;168:177–86.
13. Pfirrmann CW, Metzdorf A, Zanetti M, Hodler J, Boos N. Magnetic resonance classification of
lumbar intervertebral disc degeneration. Spine. 2001;26:1873–8.
14. Leone A, Guglielmi G, Cassar-Pullicino VN, Bonomo L. Lumbar intervertebral instability: a
review. Radiology. 2007;245:62–77.
15. D’Anastasi M, Birkenmaier C, Schmidt GP, Wegener B, Reiser MF, Baur-Melnyk
A. Correlation between vacuum phenomenon on CT and fluid on MRI in degenerative disks.
AJR Am J Roentgenol. 2011;197:1182–9.
16. Chanchairujira K, Chung CB, Kim JY, Papakonstantinou O, Lee MH, Clopton P, Resnick
D. Intervertebral disk calcification of the spine in an elderly population: radiographic prev-
alence, location, and distribution and correlation with spinal degeneration. Radiology.
2004;230:499–503.
17. Munter FM, Wasserman BA, Wu H-M, Yousem DM. Serial MR imaging of annular tears in
lumbar intervertebral disks. AJNR Am J Neuroradiol. 2002;23:1105–9.
18. Kobayashi S, Yoshizawa H, Yamada S. Pathology of lumbar nerve root compression. Part
1: Intraradicular inflammatory changes induced by mechanical compression. J Orthop Res.
2004;22:170–9.
19. Farshad-Amacker NA, Farshad M, Winklehner A, Andreisek G. MR imaging of degenerative
disc disease. Eur J Radiol. 2015;84:1768–76.
20. Qaseem A, Wilt TJ, McLean RM, Forciea MA, Clinical Guidelines Committee of the
American College of Physicians. Noninvasive treatments for acute, subacute, and chronic low
back pain: a clinical practice guideline from the American College of Physicians. Ann Intern
Med. 2017;166:514–30.
21. Kerttula L, Luoma K, Vehmas T, Grönblad M, Kääpä E. Modic type I change may predict rapid
progressive, deforming disc degeneration: a prospective 1-year follow-up study. Eur Spine
J. 2012;21:1135–42.
22. Modic MT, Ross JS. Lumbar degenerative disk disease. Radiology. 2007;245:43–61.
23. Patel KB, Poplawski MM, Pawha PS, Naidich TP, Tanenbaum LN. Diffusion-weighted MRI
“claw sign” improves differentiation of infectious from degenerative modic type 1 signal
changes of the spine. AJNR Am J Neuroradiol. 2014;35:1647–52.
24. Eguchi Y, Ohtori S, Yamashita M, et al. Diffusion magnetic resonance imaging to differ-
entiate degenerative from infectious endplate abnormalities in the lumbar spine. Spine.
2011;36:E198–202.
25. Gellhorn AC, Katz JN, Suri P. Osteoarthritis of the spine: the facet joints. Nat Rev Rheumatol.
2013;9:216–24.
26. Pathria M, Sartoris DJ, Resnick D. Osteoarthritis of the facet joints: accuracy of oblique radio-
graphic assessment. Radiology. 1987;164:227–30.
344 F. Serpi et al.
27. Guglielmi G, Schiavon F, Cammarota T. Radiologia geriatrica. Milan: Springer Milan; 2006.
https://doi.org/10.1007/88-470-0486-1.
28. Pizzini FB, Poletti M, Beltramello A, et al. Degenerative spine disease: Italian position paper
on acquisition, interpretation and reporting of magnetic resonance imaging. Insights Imag.
2021;12:14.
29. Clarençon F, Law-Ye B, Bienvenot P, Cormier É, Chiras J. The degenerative spine. Magn
Reson Imaging Clin N Am. 2016;24:495–513.
30. Frymoyer JW, Selby DK. Segmental instability. Rationale for treatment. Spine. 1985;10:280–6.
31. Izzo R, Popolizio T, D’Aprile P, Muto M. Spinal pain. Eur J Radiol. 2015;84:746–56.
32. Leone A, Martino F. Imaging del rachide. Milan: Springer Milan; 2008. https://doi.
org/10.1007/978-88-470-0836-6.
33. Manfrè L. Spinal instability. Milan: Springer Milan; 2015. https://doi.
org/10.1007/978-3-319-12901-3.
34. Even JL, Chen AF, Lee JY. Imaging characteristics of “dynamic” versus “static” spondy-
lolisthesis: analysis using magnetic resonance imaging and flexion/extension films. Spine
J. 2014;14:1965–9.
35. Lee GY, Guen YL, Lee JW, Joon WL, Choi HS, Hee SC, Oh K-J, Kyoung-Jin O, Kang HS,
Heung SK. A new grading system of lumbar central canal stenosis on MRI: an easy and reli-
able method. Skelet Radiol. 2011;40:1033–9.
36. Schizas C, Theumann N, Burn A, Tansey R, Wardlaw D, Smith FW, Kulik G. Qualitative grad-
ing of severity of lumbar spinal stenosis based on the morphology of the dural sac on magnetic
resonance images. Spine. 2010;35:1919–24.
37. Khalil JG, Nassr A, Maus TP. Physiologic imaging of the spine. Radiol Clin N Am.
2012;50:599–611.
38. Tarantino U, Fanucci E, Iundusi R, Celi M, Altobelli S, Gasbarra E, Simonetti G, Manenti
G. Lumbar spine MRI in upright position for diagnosing acute and chronic low back pain:
statistical analysis of morphological changes. J Orthop Traumatol. 2013;14:15–22.
Osteoarthritis in Appendicular Skeleton
in Geriatric Patients 13
Antonio Barile, Riccardo Monti, Federico Bruno,
Julia Daffinà, Francesco Arrigoni, and Carlo Masciocchi
13.1 Introduction
Osteoarthritis (OA) is the most common form of arthritis and is the third leading
cause of disease burden in developed countries with significant social and health
impact. As the average age and life expectancy are increasing, this form of arthritis
is expected to increase in the incoming decades. OA commonly affects weight-
bearing joints such as the knee, which is most commonly affected, and the main
clinical features are pain and stiffness. The gravity of this disease leads to a progres-
sive decline in physical functioning. Imaging plays a vital role in initial diagnosis,
staging, and monitoring of longitudinal progression and provides indications for
conservative, minimally invasive, or surgical treatment. Although the primary focus
of imaging lies in bone alterations, osteoarthritis should be framed as a whole organ
disease, and multimodal instrumental evaluation is essential to highlight the various
joint components involved and their alterations.
Compared to other appendicular joints, the glenohumeral joint is one of the least
commonly affected by osteoarthritis. The estimated radiographic prevalence is in
the range of 16–20% in an elderly population. The main risk factor for glenohu-
meral osteoarthritis is age. Other factors that increase the likelihood risk of devel-
oping shoulder osteoarthritis include female gender, obesity, Caucasians, previous
trauma, rotator cuff tears, glenohumeral instability, and crystalline arthropathy. In
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 345
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_13
AL GRAWANY
346 A. Barile et al.
To assess the presence and degree of arthritis in the glenohumeral joint the first step
is conventional radiography. Standard projections include an anteroposterior view,
a Grashey view (AP oblique internal rotation), and a further axillary view. These
views allow grant the assessment of the presence, type, and degree of arthritis and
rule out other conditions, including fractures, dislocations, and bone injuries [3].
To determine the extent of osteoarthritis of the glenohumeral joint various radio-
graphic classifications were established. The most widely adopted is the Samilson-
Prieto classification. This classification acknowledges grade 0 is normal, grade 1 is
mild with osteophytes smaller than 3 mm on the humeral head, grade 2 is moderate
with osteophytes between 3 and 7 mm on the humeral head or glenoid rim, and
grade 3 is severe with osteophytes over 7 mm, with or without contextual joint
incongruity. The state of the rotator cuff can be inferred from the radiographic eval-
uation of Grashey’s view. This view is accessed from a lateral oblique projection at
30°, tangential to the glenohumeral joint, to obtain an image parallel to the glenoid
face in order to reveal any degenerative modifications [4].
For the rotator cuff the radiographic classification used integrity is the Hamada–
Fukuda classification, a radiographic morphological description of the natural
course of massive rotator cuff tear assessing the height of the acromiohumeral
space. There are five distinctions within this classification:
For an effective preoperative planning and to assess the humeral and glenoid bone
condition, computed tomography (CT) provides greater bony detail compared to
radiographs. If there are concerns regarding the glenoid’s bone loss, the presence of
cyst, or retroversion on standard radiographs, CT should be suggested as it may
influence both the type of arthroplasty choice and the location of the glenoid com-
ponent. The CT study of the affected shoulder is necessary to estimate the glenoid
bone loss, which may require preoperative planning before eccentric reaming, aug-
mentation of bone graft, use of augmented glenoid components, or consideration of
total reverse shoulder arthroplasty. Therefore, CT evaluation should be considered
mandatory in all patients undergoing an arthroplasty procedure that requires glenoid
resurfacing (e.g., total shoulder arthroplasty and total reverse shoulder arthroplasty)
as it allows the quantification of the glenoid border and recognition of different
forms of glenoid bone loss such as cyst that can alter implant fixation and place-
ment [6].
Besides visualizing the glenoid and humeral head morphology, MRI can help detect
the underlying etiology. Thanks to MRI, the evaluation of various tissue abnormali-
ties, regarding cartilage, labrum, and glenohumeral ligaments can be assessed.
However, the capacity of detecting cartilage lesions is limited by the comparison
with to other joints. Additionally, magnetic imaging provides valuable information
for the rotator cuff evaluation, which forms an integral part of surgical planning.
Shoulder MRI is suggested in patients with rotator cuff deficiency doubts on clinical
examination. Indeed, an intact rotator cuff is required for both hemiarthroplasty and
total shoulder arthroplasty. Therefore, the integrity of the rotator cuff is a crucial
factor in determining whether the patient is a candidate for total anatomical shoul-
der arthroplasty, hemiarthroplasty, or conversely to total reverse shoulder arthro-
plasty [7, 8]. MRI shows soft tissues with an excellent detail, it can also add
information on rotator cuff tears and on the presence and degree of muscle atrophy.
348 A. Barile et al.
The Goutallier Grading Scale of Fat Infiltration of Rotator Cuff Muscles was ini-
tially described using CT to assess the degree of fat infiltration of individual rotator
cuff muscles. Goutallier’s classification consists of: grade 0 is normal muscle, grade
1 is some fat streaks, grade 2 is less than 50% fat muscle atrophy, grade 3 is 50% fat
muscle atrophy, and grade 4 is greater than 50% of fat muscle atrophy. The impor-
tance of this classification scale is its implication in the reparability of the rotator
cuff: a degree of Goutallier fat infiltration of 3 or greater (i.e., fat infiltration equal
to or greater than 50% of muscle mass) has a 50–70% tear rate [9].
Although AC osteoarthritis is less common than other locations such as the knee or
the hip, it is differently much more frequent than glenohumeral osteoarthritis.
Around 54–57% of elderly patients have an X-ray evidence of degenerative changes
in the AC joint. On the other hand, clinically relevant AC osteoarthritis is uncom-
mon, although it is more frequently related to other pathologies, such as the CR
upper impingement syndrome [10]. Primary osteoarthritis is strongly age-related, as
a matter of fact the degenerative process begins in early adulthood. Secondary
osteoarthritis, mainly following trauma such as joint sprains or distal clavicular
fractures, appears to be even more prevailing than primary osteoarthritis. The clini-
cal picture is pain in the anterior/superior aspect of the shoulder, sometimes radiat-
ing to the base of the neck/trapezius muscle. Daily movements or activities that
involve overhead or transverse movements increment pain. Local tenderness can be
caused by AC joint palpation. This range of symptoms is not specific and is also
reported in cervical spine disease and CR impingement syndromes, which, as afore-
mentioned, are predominant causes of shoulder pain. The direct intra-articular
injection of anesthetics can grant a differential diagnosis. The imaging evaluation of
the AC joint begins with an X-ray. This joint can be studied with average AP views
of the shoulder. However, the best option according to literature is the Zanca view
(a cephalad inclination of 10–15° with a 50% reduction in exposure compared to
standard AP view shoulder). Imaging findings are typical of degenerative diseases:
sclerosis, osteophytes, subchondral cysts, and joint space narrowing [11]. Bone
modifications seen on X-rays are evidenced more precisely on CT scan. At the same
time, MRI is more useful for evaluating changes in capsuloligamentous structures,
bone edema, and abnormalities in surrounding soft tissues (e.g., effusion of bursal
or tendon pathology) [12]. The AC joint can only be partially evaluated with
US. Still, it should be a part of routine shoulder examination, as AC joint osteoar-
thritis can sometimes mimic rotator cuff tendinopathy and may cause anterosupe-
rior impingement. AC osteophytes are found in 50% of patients with rotator cuff
tears but also in 14% of patients without rotator cuff tears. By placing the high-
frequency linear probe on a coronal plane at the level of the joint, the evaluation of
the two articular ends of the acromion and clavicle is possible. The superior AC
ligament is clearly seen as a banded arch echo structure that overstays the bones;
below it, the joint space can vary in size and echogenicity with movements. In case
13 Osteoarthritis in Appendicular Skeleton in Geriatric Patients 349
Despite the high prevalence, hand OA generally receives less attention compared to
OA of the weight-bearing joints. It typically affects the distal interphalangeal (DIP)
joints and the thumb base and, less frequently, the proximal interphalangeal (PIP)
joints. Patients with hand OA can experience considerable pain, stiffness, and dis-
ability with a high impact on health-related quality of life. Outcome measures in OA
usually include evaluation of pain and disability and structural changes in the joint
can be studied with outcome [14].
Currently the cheapest, most feasible, and available imaging modality for morpho-
logical assessment of the structural features of the OA hand is conventional
350 A. Barile et al.
Fig. 13.2 AP and oblique radiographic view showing initial osteoarthritis changes at the level of
the DIP with joint space narrowing and sclerosis (arrow). More advanced OA changes of the tra-
peziometacarpal joint (circle)
radiography (CR). At present, there is no established gold standard for the definition
of radiographic hand OA. Studies also differ in classification systems most com-
monly used and in the radiographic definitions of radiographic.
CR provides a two-dimensional picture of bone modifications, such as osteo-
phytes, erosions, cysts, and sclerosis and joint space narrowing (JSN) as an indirect
measure of cartilage loss (Fig. 13.2). Osteophytes can be divided into “true” intra-
articular osteophytes and traction spurs. “True” intra-articular osteophytes are found
at joint margins and can be easily seen on CR with a traditional posteroanterior
view. Traction spurs are differently located at the extensor tendon insertion or on the
central shaft and are most easily seen on CR with an oblique or lateral view. Whether
these enthesophytic changes are related to OA is not entirely clear, previous studies
have suggested that they are mainly related to age and local biomechanical factors
and not to systemic enthesopathy [15].
Since cartilage is indirectly evaluated by the inter-osseous distance, the radio-
graphic measurement of JSN is currently recommended as an imaging endpoint for
clinical trials of disease-modifying OA drugs. The radiological assessment may be
affected by the hand positioning (e.g., flexion deformity) and is further complicated
by erosive development in the fingers joints, which can lead to increased joint space
width (JSW) (pseudo-enlargement) despite the worsening of the disease.
Radiographic erosions in hands with OA are seen as bone damage in the central part
13 Osteoarthritis in Appendicular Skeleton in Geriatric Patients 351
of the joints with a typical gull-wing configuration. These erosions typically occur
in the DIP and PIP joints, but they have been described in the joints of the base of
the thumb as well. Longitudinal studies have shown that JSN precedes erosive
development, suggesting that local biomechanical factors are important for erosive
development. These findings may suggest that erosive hand OA represents severe
hand OA rather than a different disease entity. Whereas cysts are identified by the
loss of trabecular structure, sclerosis gives an increased density in the CR. Both
features can be related to bone remodeling [16].
At present, there is no consensus on the preferred grading scale. The first pro-
posed radiographic scoring system was the Kellgren and Lawrence (K&L) scale
which is the most widely used so far. The K&L scale classifies OA over a range
from 0 to 4 points (where grade of at least 2 is OA) based on different factors. These
include: the presence/severity of osteophytes, JSN, sclerosis, pseudocystic areas,
and altered shape of the bony ends. In spite of different grading descriptions for
various joint groups and difference between publications, there is general confusion
in the way of interpreting the various grades. Furthermore the K&L scale is criti-
cized for the emphasis given to osteophytes; however, sclerotic joints cannot be
classified as OA unless osteophytes are present. Therefore, several studies used
modified K&L scales to overcome these limitations. The evaluation of individual
characteristics instead of using a global score can optimize the joint assessment,
hence the OARSI (Osteoarthritis Research Society International) atlas is more fre-
quently used. With this atlas as a reference, the presence and severity of individual
characteristics (osteophyte, JSN, malalignment, erosion, subchondral sclerosis,
subchondral cysts) are assessed on semi-quantitative scales at the level of DIP, PIP,
first CMC, thumb and trapezionavicular joint. However, scoring individual features
can take longer [17].
Standard radiographs to characterize the basal thumb joint include PA, lateral
and oblique views of the hand or wrist. Arthritis of the basal joint of the thumb is
most commonly described using the Eaton-Littler classification which was first pro-
posed in 1973 and modified in 1987 by Eaton and Glickel. In this classification,
stage I is given by normal joint contours with mild joint widening (secondary to
synovitis, ligamentous laxity, or effusion), while stage II shows mild joint space
narrowing (<2 mm), mild sclerosis, subchondral cysts, and/or periarticular debris.
Stage III follows with noticeable joint space narrowing, prominent sclerosis, sub-
chondral cysts, and periarticular debris. Finally, stage IV concerns the scapho-
trapezius joint, plus the narrowing’s worsening, increased sclerosis, and the presence
of subchondral cysts. In the clinical examination, CMC subluxation, metacarpal
adduction, and MCP hyperextension are seen. However, the Eaton-Littler classifica-
tion has its flaws, including only moderate compatibility with clinical presentations,
morphological findings and therapeutic recommendations, and sub-optimal inter-
and intra-observer variability. Although some authors underline the convenience of
transverse imaging (e.g., MRI, ultrasound, CT) in basal thumb joint arthritis diag-
nosing, there is currently no recommended role for advanced imaging [18].
352 A. Barile et al.
In recent years, ultrasonography has been acknowledged as a useful tool for finger
joints’ inflammation evaluation in patients with rheumatoid arthritis. Recently, the
prevalence, validity, and reliability of US characteristics have also been studied in
patients with hand OA. By scanning the joint in both longitudinal and transverse
projection we can obtain conditions regarding the dorsal appearance with the joint
in full flexion, while volar aspects are studied with the joints in a neutral position.
US allows visualization of a broad spectrum of OA features of the hand, including
osteophytes, marginal erosions, and synovitis (Fig. 13.3). It may also be considered
a feasible and prompt tool for visualizing inflammation in patients with hand
OA. Conversely, one of the US disadvantages is the inability of the beam to pene-
trate the cortex. Because of joint anatomy, the visualization of the cartilage and
bone damage is mainly limited to its peripheral parts. Overlying osteophytes, which
interfere with the acoustic window, further complicate the assessment. In severely
damaged joints, it may be difficult to determine where an erosion begins and an
osteophyte ends. Most US studies of patients with hand OA reported a high preva-
lence of grayscale synovitis, while potency Doppler activity was less frequent. In
erosive OA, often called “inflammatory” OA, a greater power Doppler activity,
synovial hypertrophy, and joint effusion compared to patients with non-erosive
radiographic OA joints can be found. Synovitis appears to be more prevalent in
joints with active erosions, while the prevalence is lower in joints that have been
remodeled [19, 20].
With the use of MRI, OA is now recognized as a disease that affects the entire joint.
Currently, only limited research is available on the prevalence, reliability, and valid-
ity of pathology defined by MRI in hand OA. Common features of hand osteoarthri-
tis MRI can provide a multiplane image of all joint components, including structural
features such as osteophytes, cartilage, erosions/cysts, misalignment, and inflam-
matory features such as synovitis and tenosynovitis (Fig. 13.4). MRI is the only
technique capable of showing bone marrow’s injury, which is an important feature
of structural progression and nonetheless, a source of pain. The prevalence of MRI
pathology in patients with hand OA has been studied in several cohorts, founding a
high prevalence of synovitis based on gadolinium enhancement. Synovitis was also
widespread in joints without radiographic OA, and this is in line with previous
observations in knee OA. However, minimal gadolinium enhancement can also
occur in the population without OA, and therefore synovitis cannot be seen unless
there is an accompanying thickness of the synovium. In the joints of the little fin-
gers, it is also important to be aware of partial volume artifacts that can mimic
BMLs [21].
Haugen et al. recently proposed an extensive preliminary MRI scoring system
with an accompanying atlas for hand OA, validated with good intra- and inter-reader
reliability. Their system includes osteophytes evaluation, JSN, erosions, cysts, mis-
alignment, synovitis, flexor tenosynovitis, BML, and collateral ligament pathology
such as absence/discontinuity at insertion sites. The scoring was developed for the
DIP and PIP joints, and future studies need to confirm whether it can be further
applied to the metacarpophalangeal (MCP) and base of the thumb joints [22].
a b
Fig. 13.4 Coronal T1 (a) and STIR (b) slices of the hand showing advanced trapeziometacarpal
joint osteoarthritis changes with joint space narrowing, joint capsule thickening, and reactive bone
marrow edema
354 A. Barile et al.
Knee OA is the most common joint disease in the elderly and, overall, is very com-
mon. It is estimated to affect ~12.5% of patients >45 years. The medial femorotibial
joint district is more commonly affected and is usually more severe than the lat-
eral one.
The hallmarks of knee OA are like the aforementioned for other joints, This includes
joint space narrowing which is usually asymmetric, typically regarding the medial
tibiofemoral and/or the patellofemoral region. JSN <3 mm on weight-bearing knee
radiographs is considered a finding of absolute joint space narrowing with a normal
joint space >5 mm (Fig. 13.5). Compared to non-weight-bearing radiographs,
weight-bearing radiographs evidence a bigger joint space narrowing, hence affect-
ing the radiographic severity.
Plain radiographs are the imaging flagships including follow-up, although there
is a poor correlation between radiographic findings and clinical symptoms. The ini-
tial study of a patient with knee OA suspect should include a Rosenberg view, a PA
radiograph with weight-bearing, and 45° flexion, which is more sensitive in detect-
ing joint space narrowing [23].
a b
Fig. 13.5 Frontal (a) and lateral (b) plain film view in a patient with knee osteoarthritis showing
marked medial joint space narrowing, subchondral bone sclerosis, and osteophytes
13 Osteoarthritis in Appendicular Skeleton in Geriatric Patients 355
Kellgren and Lawrence first described a grading system in 1957 which was later
adopted as the standard measure for assessing radiographic OA by the World Health
Organization in 1961. The original description was graded as follows:
AL GRAWANY
356 A. Barile et al.
a b
Fig. 13.6 Coronal T1 (a) and STIR (b) knee MR images depicting high-grade lateral femoral
condyle and tibial plateau chondropathy
and lateral femoral condyle), and several joint features (e.g., cartilage signal and
morphology, synovitis, subchondral bone) are analyzed and scored according to the
severity of the involvement (Fig. 13.6). Numerous studies validated the reproduc-
ibility of these scoring systems; the MOAKS is currently the most used one for the
knee, bringing together the advantages of these scoring systems. The clinical assess-
ment of semi-quantitative analysis was demonstrated by the presence of some spe-
cific alterations (such as Hoffa synovitis, joint effusion, medial meniscus lesions)
associated with an increased risk of OA radiographic progression. Other studies,
using these transversal and longitudinal comparisons methods of disease evolution,
highlighted how the presence of cartilage damage and the presence of subchondral
edema correlate with an increased risk of prosthetic surgery necessity.
Quantitative MRI assessment provides a more sensitive and specific evaluation
of cartilage’s degeneration degree and it is superior to semi-quantitative techniques
in evaluating structural changes. Three-dimensional (3D), high-resolution sequences
are required to image the bone–cartilage interface and the cartilage surface with
adequate contrast. After image acquisition, the post-processing analysis involves
automatic or manual segmentation of the articular cartilage (that is, the separation
of the cartilage from the underlying bone and adjacent tissues). This data sets and
image reconstructions allow the evaluation of several quantitative features (e.g.,
cartilage thickness, area, volume) as continuous variables. Studies on quantitative
cartilage evaluation showed good inter-operator reproducibility at different degrees
of cartilage degeneration and excellent correlation with the surgical and histological
findings. Quantitative methods have a good correlation with semi-quantitative
results, even if more sensitive and specific in predicting cartilage loss (especially in
small widespread defects using regional analysis); therefore, some authors suggest
a combined use of these techniques. Modifications in the cartilage’s volume and
13 Osteoarthritis in Appendicular Skeleton in Geriatric Patients 357
proteoglycans, and water as well. Ultrashort time echo (UTE) T1 and T2 mapping
sequences can be used to analyze low intrinsic relaxation time tissues such as
menisci, tendons, deep layers of cartilage, deep cartilage areas where non-UTE
imaging is not sensitive enough.
Sodium imaging (23Na): This compositional imaging technique is based on the
detection of sodium, the positive cation linked to the negatively charged glycosami-
noglycan (GAG) of the cartilage’s matrix. More specifically, the sodium concentra-
tion within the cartilage matrix is directly correlated to the concentration of GAG
and hence to proteoglycans. The main strength of sodium (23Na) MRI is in fact the
high specificity to proteoglycan. As in relaxometry and diffusion imaging, exoge-
nous contrast medium administration is not required to obtain sufficient tissue con-
trast. However, in vivo sodium imaging of cartilage limits includes low intrinsic
SNR, caused by the low 23Na MRI signal compared to the one from protons.
Delayed gadolinium enhancement MRI of cartilage (dGEMRIC): Contrast
medium (gadolinium), injected intravenously is necessary for this imaging method.
The scan is performed 60–90 min after injection, to allow diffusion of the contrast
medium into the cartilage matrix. Gadolinium is negatively charged and is rejected
by positively charged GAGs in cartilage, while in case of cartilage matrix degrada-
tion, the amount of contrast in cartilage tissue will be increased in an inversely
related manner. The dGEMRIC technique showed high sensitivity and specificity;
the routine clinical use is limited by the need for high doses of gadolinium.
Chemical exchange saturation transfer imaging of GAG (gagCEST): This
sequence is based on the constant labile protons transfer between solutes (in the
case of cartilage, GAGs) and water. The difference between water–water transfer
and water–GAG transfer is measured as the magnetic transfer ratio. The signal
obtained from the energy transferred after radiofrequency proton saturation is pro-
portional to the concentration of GAG in the tissue. Unfortunately, strong magnetic
fields (7 T scanners) are required to obtain sufficient signal, thus widespread use,
even in the research field, is currently limited.
Compositional MRI sequences were widely explored in literature for the assess-
ment of cartilage, menisci, and tendons in degenerative osteoarthropathies of periph-
eral joint, mostly the results concerning the use of T2 mapping on knee articular
cartilage. The most important results were obtained by longitudinal studies on disease
progression, demonstrating the association and the predictive value of compositional
cartilage changes with potential risk factors such as age, sex, BMI, sport, injuries,
surgery. Imaging with advanced MRI sequences is becoming increasingly important
in cartilage’s degeneration studies. Because of the recent widespread development of
disease-modifying drugs and regenerative therapies (e.g., platelet-rich plasma, hyal-
uronic acid, chondrocyte implantation), MRI is also crucial in assessing new therapies
for OA prevention or for approaches to avoid progression. As the efficacy is closely
connected with early treatment, their use requires suitable biomarkers to provide an
early diagnosis and detect signs of progression during treatment. Advanced MRI find-
ings can represent, in this scenario, a powerful tool to understand how to better treat
and manage OA and this will possibly allow the creation of a “target-based therapy”
for every single component of the cartilage matrix [26].
13 Osteoarthritis in Appendicular Skeleton in Geriatric Patients 359
The hip is the third most common joint affected by osteoarthritis after the knee and
the hand. Women are more commonly affected than men. The reported prevalence
varies in different studies and is also subject to geographic distribution. The risk of
symptomatic hip osteoarthritis in people reaching the age of 85 is estimated up to
25% in some regions. Attributes, characteristics, or exposures that increase the like-
lihood of developing hip osteoarthritis are advanced age, obesity, genetics, repeti-
tive stress and mechanical overload, acetabular dysplasia, femoroacetabular
impingement, epiphysis capital femoral slip, Perthes disease, and trauma.
Patients usually experience slowly progressive hip pain or hip-related groin pain
that radiates into the thigh, gluteus, or knee. Pain can be worse at night, during rest,
or after strenuous activity, reducing motion and limiting the walking distance. It can
be associated with morning stiffness or after rest. Other symptoms include joint
locking, grinding and instability, fatigue, and pain-related psychological distress.
Occasionally, a striking discrepancy is observed between radiological findings and
clinical symptoms, in fact, patients with pronounced radiological changes have only
mild symptoms, while patients with minor radiographic findings complain of acute
pain. Therefore, OA diagnosis and, above all, the therapeutic indication, should be
made only after both radiological and clinical evaluation [27].
Plain hip radiographs are inexpensive, widely available, and readily obtainable, and
they allow a prompt OA assessment.
For hip osteoarthritis definition, an anteroposterior radiograph of the hip and a
lateral cross or lateral view of the frog leg are crucial. As for other joints, reliable
radiological indicators are joint space narrowing, subchondral sclerosis, subchon-
dral cysts, and the formation of osteophytes. Narrowing of the hip joint space
≤2 mm or <2.5 mm or the combination of joint space narrowing and the presence
of osteophytes, especially in the absence of elevated inflammatory markers (e.g.,
ESR < 20 mm/h), can be used as an indicator of osteoarthritis [28]. In addition,
loose bodies (<10), joint deformities, and subluxations can be observed. In advanced
stages of OA, the head of the femur is deformed assuming a cylindrical or mushroom-
shaped form. The classic radiological sign of osteoarthritis is the joint space narrow-
ing, particularly seen on anteroposterior radiographs taken while the patient is
standing (Fig. 13.7). When joint space and cartilage narrowing occurs, the femoral
head changes its position relatively to the socket. Femoral head migration is primar-
ily cranial (combined with anterolateral or anteromedial motion) but occasionally
axial or medial. This description of the migration is based on what can be observed
in the anteroposterior X-ray image. Radiographic signs of medial-caudal migration
of the femoral head are the joint space narrowing in the medial joint with subchon-
dral sclerosis and the osteophytes formation in case of laterocranial joint space
enlargement. The orthopedic surgeon gives joint replacement indication without
360 A. Barile et al.
a b
Fig. 13.8 CT (a) and STIR MRI (b) of the hip joint showing osteoarthritis changes with subchon-
dral geodes, bone marrow edema, and joint effusion
MRI is most commonly indicated for the evaluation for surgery where there is a
large discrepancy between clinical symptoms and osteoarthritis degree of severity
in X-ray images. The orthopedic surgeon examines the MRI to judge the labral and
cartilage damage, the presence of effusion/synovitis, and of subchondral and paral-
abral cysts. When there is hip joint OA, the primary significance of MRI is to show
both early signs of arthritis (joint cartilage, labrum) and active signs of osteoarthri-
tis. Additionally, MRI is also capable of showing any associated muscle atrophy.
Further indications are to evidence active osteoarthritis (bone marrow edema, syno-
vitis, effusion), as well as assessing the cartilage prior to hip arthroscopy (or the use
of endoprostheses). On selected patients MRI is suggested for preoperative
362 A. Barile et al.
References
1. Kobayashi T, Takagishi K, Shitara H, Ichinose T, Shimoyama D, Yamamoto A, et al. Prevalence
of and risk factors for shoulder osteoarthritis in Japanese middle-aged and elderly populations.
J Shoulder Elb Surg. 2014;23(5):613–9.
2. Mehl J, Imhoff AB, Beitzel K. [Osteoarthritis of the shoulder: pathogenesis, diagnostics and
conservative treatment options]. Orthopade. 2018;47(5):368–76.
3. Tauber M, Martetschlager F. [Shoulder Osteoarthritis-pathogenesis, classification, diagnostics
and treatment]. Orthopade. 2019;48(9):795–808.
4. Thomas M, Bidwai A, Rangan A, Rees JL, Brownson P, Tennent D, et al. Glenohumeral osteo-
arthritis. Should Elb. 2016;8(3):203–14.
5. Dekker TJ, Steele JR, Vinson EV, Garrigues GE. Current peri-operative imaging concepts sur-
rounding shoulder arthroplasty. Skelet Radiol. 2019;48(10):1485–97.
6. Bercik MJ, Kruse K II, Yalizis M, Gauci MO, Chaoui J, Walch G. A modification to the Walch
classification of the glenoid in primary glenohumeral osteoarthritis using three-dimensional
imaging. J Shoulder Elb Surg. 2016;25(10):1601–6.
7. Choate WS, Shanley E, Washburn R, Tolan SJ, Salim TI, Tadlock J, et al. The incidence and
effect of fatty atrophy, positive tangent sign, and rotator cuff tears on outcomes after total
shoulder arthroplasty. J Shoulder Elb Surg. 2017;26(12):2110–6.
8. Barreto RPG, Braman JP, Ludewig PM, Ribeiro LP, Camargo PR. Bilateral magnetic res-
onance imaging findings in individuals with unilateral shoulder pain. J Shoulder Elb Surg.
2019;28(9):1699–706.
9. Davis DL, Gilotra MN, Calderon R, Roberts A, Hasan SA. Reliability of supraspinatus intra-
muscular fatty infiltration estimates on T1-weighted MRI in potential candidates for rotator
cuff repair surgery: full-thickness tear versus high-grade partial-thickness tear. Skelet Radiol.
2021;50(11):2233–43.
10. McDonald S, Hopper MA. Acromioclavicular joint disease. Semin Musculoskelet Radiol.
2015;19(3):300–6.
11. Krill MK, Rosas S, Kwon K, Dakkak A, Nwachukwu BU, McCormick F. A concise evidence-
based physical examination for diagnosis of acromioclavicular joint pathology: a systematic
review. Phys Sportsmed. 2018;46(1):98–104.
12. Veen EJD, Donders CM, Westerbeek RE, Derks RPH, Landman EBM, Koorevaar
CT. Predictive findings on magnetic resonance imaging in patients with symptomatic acromio-
clavicular osteoarthritis. J Shoulder Elb Surg. 2018;27(8):e252–e8.
13. Precerutti M, Formica M, Bonardi M, Peroni C, Calciati F. Acromioclavicular osteoarthritis
and shoulder pain: a review of the role of ultrasonography. J Ultrasound. 2020;23(3):317–25.
14. Kalichman L, Hernandez-Molina G. Hand osteoarthritis: an epidemiological perspective.
Semin Arthritis Rheum. 2010;39(6):465–76.
15. Leung GJ, Rainsford KD, Kean WF. Osteoarthritis of the hand I: aetiology and pathogenesis,
risk factors, investigation and diagnosis. J Pharm Pharmacol. 2014;66(3):339–46.
16. Haugen IK, Boyesen P. Imaging modalities in hand osteoarthritis--and perspectives of con-
ventional radiography, magnetic resonance imaging, and ultrasonography. Arthritis Res Ther.
2011;13(6):248.
17. Marshall M, van der Windt D, Nicholls E, Myers H, Hay E, Dziedzic K. Radiographic hand
osteoarthritis: patterns and associations with hand pain and function in a community-dwelling
sample. Osteoarthr Cartil. 2009;17(11):1440–7.
18. Weiss AC, Goodman AD. Thumb basal joint arthritis. J Am Acad Orthop Surg.
2018;26(16):562–71.
AL GRAWANY
366 A. Barile et al.
19. Sivakumaran P, Hussain S, Ciurtin C. Comparison between several ultrasound hand joint
scores and conventional radiography in diagnosing hand osteoarthritis. Ultrasound Med Biol.
2018;44(3):544–50.
20. Fjellstad CM, Mathiessen A, Slatkowsky-Christensen B, Kvien TK, Hammer HB, Haugen
IK. Associations between ultrasound-detected synovitis, pain, and function in interpha-
langeal and thumb base osteoarthritis: data from the nor-hand cohort. Arthritis Care Res.
2020;72(11):1530–5.
21. Marshall M, Watt FE, Vincent TL, Dziedzic K. Hand osteoarthritis: clinical phenotypes,
molecular mechanisms and disease management. Nat Rev Rheumatol. 2018;14(11):641–56.
22. Saltzherr MS, Muradin GSR, Haugen IK, Selles RW, van Neck JW, Coert JH, et al. Cartilage
evaluation in finger joints in healthy controls and early hand osteoarthritis patients using high-
resolution MRI. Osteoarthr Cartil. 2019;27(8):1148–51.
23. Teichtahl AJ, Wluka AE, Davies-Tuck ML, Cicuttini FM. Imaging of knee osteoarthritis. Best
Pract Res Clin Rheumatol. 2008;22(6):1061–74.
24. Jacobson JA, Girish G, Jiang Y, Sabb BJ. Radiographic evaluation of arthritis: degenerative
joint disease and variations. Radiology. 2008;248(3):737–47.
25. Kornaat PR, Bloem JL, Ceulemans RY, Riyazi N, Rosendaal FR, Nelissen RG, et al.
Osteoarthritis of the knee: association between clinical features and MR imaging findings.
Radiology. 2006;239(3):811–7.
26. Bruno F, Arrigoni F, Palumbo P, Natella R, Maggialetti N, Reginelli A, et al. New advances
in MRI diagnosis of degenerative osteoarthropathy of the peripheral joints. Radiol Med.
2019;124(11):1121–7.
27. Aresti N, Kassam J, Nicholas N, Achan P. Hip osteoarthritis. BMJ. 2016;354:i3405.
28. Nilsdotter A, Bremander A. Measures of hip function and symptoms: Harris Hip Score
(HHS), Hip Disability and Osteoarthritis Outcome Score (HOOS), Oxford Hip Score (OHS),
Lequesne Index of Severity for Osteoarthritis of the Hip (LISOH), and American Academy of
Orthopedic Surgeons (AAOS) Hip and Knee Questionnaire. Arthritis Care Res. 2011;63(Suppl
11):S200–7.
29. Weber MA, Merle C, Rehnitz C, Gotterbarm T. Modern radiological imaging of osteoarthritis
of the hip joint with consideration of predisposing conditions. RöFo. 2016;188(7):635–51.
30. Gold GE, Cicuttini F, Crema MD, Eckstein F, Guermazi A, Kijowski R, et al. OARSI clini-
cal trials recommendations: hip imaging in clinical trials in osteoarthritis. Osteoarthr Cartil.
2015;23(5):716–31.
31. Jaremko JL, Lambert RG, Zubler V, Weber U, Loeuille D, Roemer FW, et al. Methodologies for
semiquantitative evaluation of hip osteoarthritis by magnetic resonance imaging: approaches
based on the whole organ and focused on active lesions. J Rheumatol. 2014;41(2):359–69.
32. Huang BK, Tan W, Scherer KF, Rennie W, Chung CB, Bancroft LW. Standard and advanced
imaging of hip osteoarthritis. What the radiologist should know. Semin Musculoskelet Radiol.
2019;23(3):289–303.
33. Barg A, Pagenstert GI, Hugle T, Gloyer M, Wiewiorski M, Henninger HB, et al. Ankle osteoar-
thritis: etiology, diagnostics, and classification. Foot Ankle Clin. 2013;18(3):411–26.
34. Kraus VB, Kilfoil TM, Hash TW II, McDaniel G, Renner JB, Carrino JA, et al. Atlas
of radiographic features of osteoarthritis of the ankle and hindfoot. Osteoarthr Cartil.
2015;23(12):2059–85.
35. Wilkinson VH, Rowbotham EL, Grainger AJ. Imaging in foot and ankle arthritis. Semin
Musculoskelet Radiol. 2016;20(2):167–74.
36. Acanfora C, Bruno F, Palumbo P, Arrigoni F, Natella R, Mazzei MA, et al. Diagnostic and inter-
ventional radiology fundamentals of synovial pathology. Acta Biomed. 2020;91(8-S):107–15.
37. Barile A, La Marra A, Arrigoni F, Mariani S, Zugaro L, Splendiani A, et al. Anaesthetics,
steroids and platelet-rich plasma (PRP) in ultrasound-guided musculoskeletal procedures. Br J
Radiol. 2016;89(1065):20150355.
Metabolic Bone Disease in Geriatric
Patients 14
Maria Pilar Aparisi Gómez, Francisco Aparisi,
Giuseppe Guglielmi, and Alberto Bazzocchi
14.1 Introduction
As humans age, the lean components of the organism such as total body water,
organ mass, mineral bone, and skeletal muscle decrease, while total body fat
increases, with an associated redistribution: it becomes more abundant in the
abdominal region than in peripheral locations [1].
The process of aging involves endocrine and metabolic alterations, but besides,
an increasingly sedentary lifestyle generates a positive imbalance between intake
and use of energy [2, 3]. Aging is characterized by a low-grade chronic inflamma-
tory status known as “inflammaging” [4].
Due to the role fat has as an endocrine organ, the increase in body fat and the redis-
tribution of the fat in geriatric population have been demonstrated to be associated
with risk factors for non-insulin-dependent diabetes and cardiovascular disease [5].
M. P. Aparisi Gómez
Department of Radiology, Auckland City Hospital, Auckland, New Zealand
Department of Radiology, IMSKE, Valencia, Spain
e-mail: [email protected]
F. Aparisi
Department of Radiology, Hospital Nueve de Octubre, Valencia, Spain
G. Guglielmi
Clinical and Experimental Medicine, University of Foggia, Foggia, Foggia, Italy
e-mail: [email protected]
A. Bazzocchi (*)
Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
e-mail: [email protected]
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 367
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_14
368 M. P. Aparisi Gómez et al.
The loss of mineral contents in bone and structural changes are a major risk for
morbidity, need for institutionalization, and mortality. Sarcopenia, with a decrease
in muscle mass and function, has been associated with impaired immunity and func-
tional status [6, 7].
The three tissues (fat, muscle, and bone) have a very close relationship, and
therefore analysis and evaluation should be approached in a combined way.
Osteoporosis is described as a systemic bone disease characterized by low bone
mass and microarchitectural deterioration of bone tissue, with a consequent increase
in bone fragility and susceptibility to fracture [8].
Osteoporosis as an entity may result either from defective skeletal development
leading to a start point of low bone mass and quality, or from an imbalance in cou-
pling, with an increase in the resorption of bone, exceeding formation.
The most prevalent cause for osteoporosis and therefore statistically the most
common cause for fragility fractures is postmenopausal osteoporosis, which is
inherently linked to aging in women; however, any situation where there is osteopo-
rosis, either primary or secondary to several conditions, in both genders, increases
the risk of occurrence of a fragility fracture. Postmenopausal osteoporosis develops
after a decrease in estrogen levels following menopause.
Senile osteoporosis is another primary cause for osteoporosis and affects both
genders. This is age related, occurring in individuals older than 75 years. Bone for-
mation is impaired through a mechanism of decreased renal production of 1,25
dihydroxyvitamin D with a subsequent drop in calcium absorption from the diet that
results in secondary hyperparathyroidism [9].
The most prevalent type of fragility/insufficiency fracture is the vertebral com-
pression fracture, but the effect of osteoporosis on the skeleton is systemic, and
there is increased risk of almost all types of fractures. Other frequent locations for
insufficiency fractures are the pelvic girdle and the proximal femur. Locations such
as the femoral diaphysis, tibia, fibula, and calcaneus and metatarsal bones are less
frequent and can represent a diagnostic challenge [10].
In the bone, the aging process is characterized by a progressive accumulation of
adipose cells within the bone marrow (BM). The role of marrow adipose tissue
(MAT) as a component of the BM microenvironment has been thoroughly investi-
gated in the last few years. A growing amount of evidence shows that there is an
inverse association between MAT content and both bone mineral density (BMD)
and bone integrity [11].
In this chapter, we aim to summarize the current knowledge on changes in bone
metabolism occurring in the elderly and review the possibilities of assessment that
each one of the imaging techniques offers for the adequate assessment of bone min-
eral density in the geriatric population.
The contents of this chapter need to be taken into consideration together with the
chapter on body composition in geriatric patients. The effects of aging on fat and
muscle are extensively reviewed in a dedicated chapter, but it is important to
acknowledge the close relationship and interrelation existing between all
components.
14 Metabolic Bone Disease in Geriatric Patients 369
14.2.1 Menopause
Estrogen has a very important role in normal physiologic remodeling, and its defi-
ciency after menopause results in remodeling imbalance with an increase in bone
turnover. The imbalance leads to a progressive loss of trabecular bone first (most
metabolically active) and then progressively cortical bone [9].
Menopause is defined by the World Health Organization (WHO) as the “perma-
nent cessation of menstruation resulting from the loss of ovarian follicular activity,”
initiated by the decline in estrogen and progesterone production and by increasing
follicle stimulating hormone (FSH) and luteinizing hormone (LH) levels. Twelve
months of consecutive amenorrhea are necessary to establish the onset of menopause.
The perimenopausal stage is defined as the time elapsed from the commencement of
the first clinical signs (cycle irregularity, known as “menopause transition”) and a year
after the last period. Elevation of early follicular phase FSH is a clinical marker of
reduced ovarian reserve and decreased response of the ovary to ovulation induction.
The endocrinology of menopause is complex and results from changes in the
pituitary-ovarian axis with age in regularly cycling women. The decline in follicle
numbers results in a decline in ovarian hormone production, which alters the pitu-
itary feedback.
The rise in serum FSH is accompanied by minimal changes in circulating levels
of LH. Concentrations of androgens appear to be lower in the postmenopausal sta-
tus, it is thought that this happens as a function of increasing age during the repro-
ductive years as opposed to a consequence of menopause as such. By 12–24 months
after menopause, the levels of estradiol in serum are normally <80 pm/L, compared
with mean values of approximately 550 pm/L in premenopausal stage. The levels of
FSH are 10–15 times higher than the levels that would be usual in earlier follicular
phases in young women. LH levels are approximately 3 times higher. Plasma testos-
terone levels decrease from ~1 to 0.6 nmol/L. There is an approximately 40%
decrease in the ovarian androstenedione production, dehydroepiandrosterone sul-
fate (DHEAS) production declines in a linear fashion with age, not related to post-
menopausal status as such [12].
The decrease in estradiol levels has different systemic effects and can contribute
to the development of different disorders: osteoporosis, cardiovascular risk, changes
in mood, and mental health.
14.2.2 Calcitriol
Senile osteoporosis is another primary cause for osteoporosis and affects both gen-
ders. This is age related, occurring in individuals older than 75 years. Bone forma-
tion is impaired through an alteration in the metabolism of vitamin D. In this context
the cortical as well as the trabecular bone is affected [9].
370 M. P. Aparisi Gómez et al.
In the past, it was believed that obesity had a protective role in bone and in muscle,
providing mechanical load and therefore stimulus for their maintenance. Fat is as
well a source of estrogens, which are beneficial to maintain bone health, reducing
bone resorption, contributing to muscle repair and regeneration and reducing adipo-
genesis [23–25].
14 Metabolic Bone Disease in Geriatric Patients 371
However, some concepts have shifted. Fat has been seen to act as an endocrine
organ, releasing hormones such as leptin and releasing cytokines, which are proin-
flammatory agents [26]. In particular, visceral fat (VAT) has a negative impact on
bone and muscle [27]. VAT secretes proinflammatory cytokines such as tumor
necrosis factor-alpha (TNF α), interleukin 1 and 6 (IL-1 and IL-6), and even
C-reactive protein in high inflammatory status [28]. These factors promote and sus-
tain low-grade chronic inflammation, which in turn causes derangement of all three
tissues simultaneously and causes more fat deposition, perpetuating the problem
[27, 28].
Weight gain in older adults leads to greater visceral fat accumulation and long
term impairments in bone and muscle as a consequence.
A study performed in 500 healthy women demonstrated that body fat higher than
33% was negatively correlated to femoral neck bone mineral density (BMD), and if
this increased to 38%, it was negatively correlated to BMD in the lumbar spine and
total-body BMD [29].
Another study identified a cut off of 38.3% body fat as the inflection point where
the slope of the relation between visceral fat and percent body fat increases signifi-
cantly [30]. This disputes the concept that obesity is protective for bone health,
especially for women, but the relationship between obesity and bone is ultimately
of complex nature [31], and more research is needed to determine the threshold in
which body fat becomes harmful for bones and muscle [29], as we will review when
addressing the topic of frailty.
The amount of bone marrow adipose tissue (MAT) increases with aging, in obe-
sity and osteoporosis. MAT appears to reduce osteoblast and osteoclast activity,
slowing down bone turnover (which may be beneficial in some cases like meno-
pause) and decreasing the rate of bone accrual [32]. A negative correlation between
MAT and BMD has been proven [33]. However, it is still confusing whether the
relationship between MAT and osteoporosis is causative or correlative [32].
It is clear that MAT has a role in bone health, through its paracrine and endocrine
interaction with the other components of bone. There is evidence that the bone mar-
row stem cells (precursors of adipocytes and osteoblasts) may favor adipogenic
differentiation in the presence of excessive adiposity, and this is one of the reasons
osteoporosis has been labeled as the “obesity of bone” [34] and increased adipogen-
esis in the marrow “osteosteatosis” [35].
The MAT–bone interaction is a fertile area of research. The development of
imaging, and more especially MR based techniques has unlocked numerous
pathways to assess and quantify MAT and thus set the ground to carry out stud-
ies to further elucidate the implications of MAT in physiologic and pathologic
conditions [36].
A prospective study in a population of Korean women demonstrated that post-
menopausal women with higher VAT levels lost significantly more lean mass over a
period of 27 months than women with lower VAT levels. The decrease in VAT did
not result in a parallel change in BMI, which suggested that fat was replacing the
lost muscle tissue and possibly infiltrating it [27, 37].
372 M. P. Aparisi Gómez et al.
Aging in skeletal muscle involves fat deposit, in the form of intra- and extra-
myocellular adipocytes, known as myosteatosis. This is seen in older women, even
if they are not obese [38], but can also be seen in younger individuals [39].
Myosteatosis has been considered the “obesity of muscle” [35].
The role of proinflammatory cytokines such as TNF α and IL-6 in muscle wast-
ing and their elevated serum concentration in sarcopenia and sarcopenic obesity has
been established [40]. Additionally, muscle mass is the main determinant of resting
metabolic rate, and loss of muscle would in turn also promote weight gain and fat
accumulation.
In this way, it is easy to see how muscle and bone loss and accumulation of VAT
with aging, aggravated by overall excess of adiposity are part of a cycle where
increased inflammation from visceral fat favors sarcopenia and osteopenia, pro-
motes obesity and as a consequence, greater fat accumulation.
Although fat infiltration of bone and muscle is a part of normal aging, its eleva-
tion in an obesogenic environment exacerbates loss of bone and muscle. The
increase in MAT and myosteatosis, combined with the age related loss of bone and
muscle mass contributes even more to loss of bone and muscle and therefore strength
and overall functionality [38, 41].
Loss of functionality and mobility increases the risk for falls and fractures.
Besides, processes like myosteatosis lead to the development of disorders like dia-
betes, in which the risk of falls is increased, secondary to impaired vision or neu-
ropathy [11, 38].
All this explains the increased risk of frailty in older adults, in cases of osteosar-
copenic obesity and of osteopenic obesity and sarcopenic obesity [42, 43].
Older women suffering from any of these conditions were inferior in several
functional performance measures to only obese counterparts and those suffering
from osteosarcopenic obesity showed significantly poorer performance in hand grip
strength, balance, and walking speed, compared to each one of the other groups [43].
Chung et al. found in a study on older adults that sarcopenic obesity put them at
greater risk of osteoporosis, and the physical decline from sarcopenia appeared to
promote greater loss of bone [44]. The physical decline from any of the conditions
may easily aggravate other declines, ultimately leading to osteosarcopenic obesity.
Overall changes in body composition, leading to impairments in bone, muscle,
and fat tissues have to be taken into consideration when evaluating the health of the
elderly.
low vitamin K status, often present in the elderly, there will be an excess of under-
carboxylated osteocalcin which will not bind to hydroxyapatite and therefore less
bone stabilization, resulting in bone loss. Osteocalcin also stimulates the secretion
of adiponectin from fat cells. Some studies demonstrated that adiponectin had a
negative impact on bone mass by decreasing osteoblast proliferation [46, 47] but
other studies showed that through the same pathway, adiponectin inhibited osteo-
clastogenesis [48, 49].
Undercarboxylated osteocalcin in serum was shown to stimulate pancreatic beta
cell proliferation and insulin secretion and thus positively contribute to modulate
energy metabolism [50].
Mounting evidence on systemic hormonal actions of osteocalcin have gained it
the consideration of an osteokine [26].
Hip, vertebral, and wrist fractures are the most frequent fractures associated with
osteoporosis. The effect of osteoporosis is systemic, though, so there is increased
risk for almost all types of fractures.
The combined lifetime risk for a hip, forearm, and vertebral fracture is
approximately 40%, which is equivalent to the risk of developing cardiovascular
disease [51].
Fragility fractures due to osteoporosis are one of the most substantial challenges
to public health. The World Health Organization considers osteoporosis to be sec-
ond to cardiovascular risk as a critical health problem.
Approximately 1 in 3 women and 1 in 5 men over the age of 50 will have a fragil-
ity fracture in their remaining lifetime, as per data in Caucasian populations, from
the International Osteoporosis Foundation [52].
In 2000 there were an estimated nine million new osteoporosis fractures, of
which 1.6 million were at the hip, 1.7 million were at the forearm, and 1.4 million
were clinical vertebral fractures. Europe and the Americas accounted for 51% of all
these fractures, while most of the remainder occurred in the Western Pacific region
and Southeast Asia.
In 2006 it was estimated that osteoporosis caused more than 8.9 million fractures
annually worldwide, resulting in an osteoporosis fracture every 3 s [53].
In patients with a hip fracture, it is estimated that up to 20% will die within the
following year due to associated morbidity, with a mortality at 5 years 20% greater
than expected, and approximately 20% will require permanent care [54] (Fig. 14.1).
Patients with vertebral fractures have less severe complications, but these are
more frequent, and approximately only 30% of them come to clinical attention [55].
In these cases there is substantial disability from pain and generally increased tho-
racic kyphosis.
Vertebral fractures have a prevalence of about 35–50% among women over
50 years of age (postmenopausal status) [56], frequently occur in absence of a major
374 M. P. Aparisi Gómez et al.
Fig. 14.1 Subcapital femoral neck fracture of the right hip. These fractures normally result from
a fall from height in the context of osteopenia or osteoporosis. In patients with a hip fracture, it is
estimated that up to 20% will die within the following year due to associated morbidity, with a
mortality at 5 years 20% greater than expected, and approximately 20% will require permanent
care. The main radiographic features of osteoporosis consist of increased bone radiolucency, corti-
cal thinning, and changes in the trabecular pattern, all visible on this radiograph
trauma and may be asymptomatic [57], and increase the risk of a new incidental VF
and other fragility fractures. Individuals with a pre-existing VF have a four- to five-
fold increase in risk of sustaining a new VF, the risk increases with the number of
prevalent fractures at baseline and is BMD-independent [56]. Conventional radiog-
raphy and DXA represent the techniques of choice for VFs detection [57].
Wrist fractures increase almost twofold the risk of subsequent hip or vertebral
fractures, but also the risk of a new forearm fracture is increased by 3.3 times, and
other skeletal fractures by 2.4 times [58].
Humeral fractures, which are the third most common type of fracture in people
over 65 years, have been associated with a higher risk of hip fractures more than 5
times in the following year [59].
Fractures at the foot or ribs were seen to double the risk of hip, vertebral, fore-
arm, and other types [58].
The existence of an insufficiency fracture is an indication for treatment of
osteoporosis.
14.5.2 Frailty
indicators: muscle strength loss, slowness, fatigue, low physical activity, and body
weight loss [62].
Frail older adults are at increased risk of falls, hip fracture, disability, and mortal-
ity [63]. Evidence that body weight is positively associated with bone health in
older adults is increasing [25]; there is also evidence that lean and fat masses, con-
stituting 95% of body weight, might have a different relationship with bone
mass [64].
The potential consequences of body-composition change in frail older persons
should be put both in the context of bone health and with regard to risks of osteopo-
rotic fractures. Frail people have lower muscle mass and higher fat mass than non-
frail people, but osteoporosis is highly prevalent in the elderly population.
Derangements in inflammatory, endocrine, coagulation, and metabolic systems
should be individually assessed in frail adults and not allow for generalization in
comparison with non-frail aging populations [65, 66]. Zaslavsky et al. demonstrated
that adiposity in the context of frailty has a different impact on survival than the one
that can be observed in non-frail population [67]. A recent study from the same
group showed that appendicular, trunk, and total body fat, as well as lean mass
indexes, are significant determinants of total hip BMD in frail women. Higher lean
and fat mass indexes are associated with lower risks of hip fractures, and whole-
body fat is the only index to retain indirect association, independently of total hip
BMD. Change over time in body-composition indexes was not a significant deter-
minant of bone health in older women with frailty [68]. These data confirm previous
studies showing an association between whole-body and abdominal fat mass mea-
sures and lower risks of hip fracture. The association was independent of BMD,
indicating that central adiposity might be informative in predicting fractures over
and beyond BMD [69]. In summary, central adiposity may have some benefits for
bone health in the context of frailty, and this should be put in the balance with the
risks of cardiovascular disease.
In studies that included men and women, higher lean mass was not significantly
correlated with hip fractures in models adjusted for total hip BMD [38, 68] which
seems contradictory, given that the association of low BMD and low lean mass on
increasing fracture risk has also been proven [70]. The positive impact of lean mass
on hip fracture risk might thus be channeled through anabolic processes on the bone.
Using the pool of patients from the Women’s Health Initiative, with sample size
of over 120,000 postmenopausal women, Harris et al. concluded that women with
low BMD (T-score <−1), with and without sarcopenia, had a higher risk of fracture
than women with isolated sarcopenia and those considered normal. These results
suggest that sarcopenia does not carry additional risk for fracture in women [71].
The study was limited to fractures around the hip; although women with the combi-
nation of low BMD and sarcopenia had a higher risk of fracture, whereas sarcopenia
alone was not an independent risk factor for fracture in women, the fact that adding
sarcopenia to low BMD resulted in a greater risk suggests that there is communica-
tion between muscle and bone at this site, associated with frailty [72]. The interac-
tion may be mediated by mechanical stimuli, genetic factors, hormonal influences,
and body composition. Total bone mineral content is associated more closely with
376 M. P. Aparisi Gómez et al.
lean tissue than with fat tissue mass, and regional BMD is predicted by changes in
fat tissue mass [73].
A number of mechanisms for the fat–bone relationship in older adults have been
proposed. These include the effect of soft tissue mass on skeletal loading, the asso-
ciation of fat mass with the pancreatic beta cell, and adipocyte secretion of hor-
mones involved in bone metabolism [25]. Additionally, weight reduction may lead
to accelerated rates of bone loss in postmenopausal women. In a large study that
also used data across the Women’s Health Initiative, postmenopausal women who
lost more than 5% of their baseline weight within 3 years of follow-up had 65%
higher rate of hip fractures as compared with women with stable weight (<5%
change). This confirms that body weight is positively associated with bone
health [74].
Frail women are at increased risk of recurrent falls compared with non-frail
women [75], and most hip fractures are secondary to falls [76]. A lower muscle
mass may lead to accidents or falls [77], with secondary fractures, but falls could
also be a confounding factor, indicative of poor general health [78]. Conversely,
higher fat mass might be protective during falls by fat cushioning [69].
14.6.1 Radiography
Fig. 14.2 Multiple vertebral insufficiency fractures in the thoracolumbar transition, with different
morphology and severity. Based on Genant’s method vertebral deformities are graded according to
shape and severity. Cortical thinning results from the reabsorption of the periosteal, intracortical,
and endosteal layers. When this happens in the vertebral bodies concomitantly to the increase in
bone radiolucency, the vertebrae acquire a “picture frame” appearance, also known as “ghost
vertebra”
have a great inter-observer variation and are dependent on the quality of radiographs
and superimposition of soft tissues.
Radiographic absorptiometry (RA) (method comparing densities) and metacar-
pal radiogrammetry (evaluation of cortical changes—ratios using radiographs of
metacarpals) were developed from radiography for quantitative purposes.
Metacarpal radiogrammetry has evolved into digital X-ray radiogrammetry (DXR),
in which automated measurements obtained from three metacarpal bones (instead
of one in conventional radiogrammetry) provide more accuracy and precision,
through the calculation of a “bone volume per projected area” (VPA) from which
BMD is derived via a geometrical operation [84]. A significant correlation exists
between DXR-derived BMD (DXR-BMD) at the three mid-metacarpals and DXA-
derived BMD at the spine, total hip, and distal radius [85].
Radiographs are also important for the diagnosis of vertebral fractures. The exis-
tence of an insufficiency fracture is an indication for treatment of osteoporosis.
Several radiograph-based methods have been developed to identify and score
VFs. Quantitative morphometry (QM), the visual semiquantitative (SQ) method,
and an “algorithm-based qualitative” (ABQ) method [86] are now available, the
most commonly used being the visual SQ method proposed by Genant et al. [87].
This method has been extensively validated and according to the ISCD official posi-
tions it represents the technique of choice to characterize VFs [57, 88]. Based on
378 M. P. Aparisi Gómez et al.
Genant’s method vertebral deformities are graded according to shape and severity.
Readers are asked to estimate the percentage of height and/or area reduction semi-
quantitatively—without a direct measurement (Fig. 14.2). The deformity is classi-
fied based on the location (anterior—wedge, middle—biconcave, or posterior and
anterior loss—crush) and on severity of height loss (normal: 0, mild 20–25%: 1,
moderate 25–40%: 2, and severe >40%: 3, plus grade 0.5 for uncertain or question-
able vertebrae). A spinal fracture index can be calculated by adding the individual
vertebral body scores. This allows a quantification of the extent of deformation [57].
An important remark about radiographs is that they represent an opportunistic
method to diagnose vertebral fractures. Fractures can be incidentally found in a
number of methods and examinations performed for other clinical purposes (e.g.
chest or abdominal radiographs) [89–91].
These fractures are currently underreported by radiologists, probably because
the main focus is set on evaluating different pathology [92]. Lastly, vertebral frac-
tures can occur as pathological fractures in the context of malignancy. In some cases
radiographs will be able to give enough information to provide unsuspected
diagnoses.
Artificial Intelligence applied to imaging is an evolving field and will be funda-
mental for this task [93].
Dual energy X-ray absorptiometry (DXA) represents the most widely used tech-
nique for the assessment of BMD, thanks to its availability, the very low radiation
dose, and its low cost. It represents the standard for diagnosis and monitoring of
osteoporosis and conditions involving low bone mass. It is normally the first clinical
imaging tool used to diagnose osteoporosis.
DXA is based on the use of two X-ray beams of different energy. The ratio
between the degree of attenuation of the lower energy and the higher energy beam
is the “R value” and is specific for each tissue. From the R value, using complex
algorithms, it is possible to obtain the amount of BMC in pixels containing bone.
BMD is then calculated as the ratio BMC/area (in g/cm2) [79]. The DXA measure-
ment of BMD is an areal measurement [areal-BMD (a-BMD)], as opposed to a true
“density” (per volume).
BMD is expressed in terms of standard deviation (SD) comparing individual
BMD measurement to a reference range obtained from a population of healthy
young adults (T-score) and from an age-matched population of the same gender and
ethnic group (Z-score). In postmenopausal women and in men older than 50, osteo-
porosis is defined by a T-score ≤−2.5 SD at the lumbar spine (from L1 to L4),
femoral neck, or total hip [68]; BMD ≥ −1 SD is considered normal, while BMD in
the range between −1 and −2.5 SD is in the range of osteopenia (Fig. 14.3). BMD
measured by DXA accounts only for 60–70% of variation in bone strength (other
factors such as bone architecture are also contributory) and the majority of
14 Metabolic Bone Disease in Geriatric Patients 379
Fig. 14.3 DXA, lumbar spine. Woman, 73 years old, 67 kg. T-score is within the normal range
(above −1 SD)
Fig. 14.4 DXA of the forearm. Typically, when a central site, lumbar spine and/or proximal fem-
ora, cannot be reliably assessed in patients investigated for osteoporosis, and in specific clinical
scenarios, the forearm is scanned
Other non-BMD measures from DXA scans focused on hip geometry measures,
including hip structural analysis, hip axis length (HAL), and neck-shaft angle, can
be performed, with limited value in clinical practice. HAL derived from DXA is
associated with hip fracture risk in postmenopausal women [101].
DXA and conventional radiography are the techniques of choice for the detection
of vertebral fractures [57] (Fig. 14.9). This is because of the possibility to perform
panoramic views, the lower radiation exposure, the timing (the patient can be
scanned for both BMD and VFs in the same session), the integrated morphometric
tool advantages of DXA, and the spatial resolution and better qualitative assessment
advantage of radiography. Quantitative morphometry as a scoring method remains
a useful tool in specific settings [102].
Lateral spine imaging with standard radiography or densitometric VFA is indi-
cated when T-score is <−1.0 and of one or more of the following is present: Women
age ≥70 years or men ≥ age 80 years; historical height loss >4 cm (>1.5 in.); self-
reported but undocumented prior vertebral fracture; glucocorticoid therapy equiva-
lent to ≥5 mg of prednisone or equivalent per day for ≥3 months [ISCD 2019
guidelines].
Fig. 14.5 A 71 years old female patient presented with multiple vertebral fractures (including
more than two levels at L1-L4) (a), previous fractures and surgical fixation at both proximal
femurs, and post-traumatic changes at the non-dominant forearm. The site for BMD analysis was
the dominant wrist, which confirmed a status of osteoporosis, with a T-score = −3.5 SD (b)
Fig. 14.6 TBS analysis (explanation of software). Same patient as in Fig. 14.3. TBS shows a
normal bone structure
The main parameters assessed by QUS include speed of sound (SoS), a param-
eter closely related to bone mineralization, and broadband ultrasound attenuation
(BUA), closely related to the structural characteristics of trabecular bone [105].
There is a strong level of correlation between trabecular transmission parameters
(SoS and BUA) in the heel and BMD derived by DXA at lumbar spine and femoral
neck. Validated heel QUS devices have been demonstrated to predict fragility frac-
tures in postmenopausal women (hip, vertebral, and global fracture risk) and in men
over 65 (hip and all non-VFs), independently of central DXA BMD [104].
QUS is usually performed in the distal metaphysis of the phalanx, calcaneus,
radius, and tibia. It is important to emphasize that the only validated measurement
site in the context of osteoporosis diagnosis and management is the heel. In the
study of osteoporosis, DXA remains the method of choice in clinical practice for
therapeutic decisions, but if a DXA scan cannot be performed, pharmacologic treat-
ment can be initiated on the basis of a sufficient high fracture probability, assessed
by heel QUS (using device specific thresholds) in conjunction with clinical risk
factors (according to ISCD position) [104].
14 Metabolic Bone Disease in Geriatric Patients 383
Fig. 14.7 Woman, 71 years old, 42 kg. DXA shows very low aBMD values (T-score in the range
of osteoporosis according to WHO criteria) (a), TBS has values within the normal limits (b)
384 M. P. Aparisi Gómez et al.
Fig. 14.8 Woman, 73 years old, 92 kg. DXA shows a T-score in the normal according to WHO
criteria (a). TBS deonstrates low values - abnormal bone structure (b)
14 Metabolic Bone Disease in Geriatric Patients 385
Fig. 14.10 DXA scan of the lumbar spine and of the non-dominant proximal femur. With aging,
osteoarthritic (OA) changes increase and particularly affect lumbar spine assessment of
BMD. Calcified ateromatosis, scoliosis, sequelae of vertebral fractures, and other conditions also
significantly impact the assessment. In the patient scanned and presented in this figure (Woman,
77 years old, 60 kg), lumbar spine shows inaccurate values due to OA changes and scoliosis (a),
while the proximal femur (b) can reliably depict a status of osteopenia (T-score −2.3 SD at the
femoral neck, not far from osteoporosis)
including diagnostic cut-off points that may be used for assigning a spine QCT
diagnostic category equivalent to the WHO guidelines (The ACR introduced guide-
lines for evaluating QCT studies [https://www.acr.org//media/ACR/Files/Practice-
Parameters/qct.pdf]) (Fig. 14.11).
14 Metabolic Bone Disease in Geriatric Patients 387
The region of interest for QCT is the lumbar spine. Other measurement sites
commonly include the proximal femur, forearm, and tibia [109]. In clinical applica-
tion, spine and proximal femur are analyzed using standard whole-body CT scan-
ners equipped with dedicated software for the analysis. Multiple studies have
evaluated the clinical utility of QCT in fracture prediction and in longitudinal stud-
ies: the ability of spinal trabecular BMD obtained by QCT to predict spinal fractures
in postmenopausal women is comparable to or better than that of lumbar spine
BMD obtained by DXA; sufficient evidence to support this statement in men is still
necessary [110]. Total femur trabecular BMD obtained by QCT has the capability
to predict hip fractures as well as hip BMD obtained by DXA in both menopausal
woman and older men [111].
QCT has also been extensively tested in monitoring age-, disease-, and treatment-
related BMD changes [110, 111]. However, DXA should be favored for therapeutic
decisions and in clinical practice to limit radiation exposure, unless QCT can pro-
vide superior information [111].
Also CT offers the possibility of opportunistic screening. BMD can be poten-
tially calculated from a pre-existing acquisition in patients at increased risk of frac-
ture, without need for any additional DXA scan. However, the absence of in-scan
calibration phantom and the lack of a standardized acquisition protocol are common
problems. According to the ISCD official positions (last version 2019), the identifi-
cation of patients with high fracture risk (according to low BMD or strength mea-
sures derived by CT at the spine or proximal femur) is possible with conventional
CT scan only if machine-specific cut-off values and scanner stability have been
established [110].
Multidetector spiral CT (MDCT) of the thorax and abdomen is one of the most
useful tools for opportunistic diagnosis of vertebral fractures in postmenopausal
women. On the midline-sagittal CT plane, the central area of the vertebral body end
plate (the site where vertebral fractures appear) can be very accurately analyzed.
Axial images are not very sensitive, but coronal and sagittal reconstructions are now
widely used and these may easily demonstrate fractures. The initial localizer views
of CT (scout) are suitable for detection of incidental vertebral fractures [112]. Intra-
and inter-observer agreement based on a semiquantitative method is fair to good.
Mild degrees of fracture and fractures located in levels T4 to T9 represent the main
sources of error [113].
388 M. P. Aparisi Gómez et al.
Several studies have stated that incidental osteoporotic fractures are underre-
ported and that sagittal CT reformations provide additional information and should
be a part of standard CT analyses, to improve detection rate [92]. Clinical studies
have demonstrated that MDCT structure measurements at the proximal femur and
spine improve differentiation between osteoporotic patients with proximal femur
fractures or spine fractures and healthy control patients [114]. Besides, it has been
proven useful in monitoring treatment with teriparatide [115]. Data obtained with
MDCT are useful for final element analysis (FEA), which has been used to study
bone strength and monitor changes after the administration of treatment. Using con-
version factors, reliable measurements can be calculated from the spine and hip
from routine abdominal and pelvic MDCT scans [116].
Dedicated pQCT scanners (peripheral scanners) are used to evaluate v-BMD and
bone microarchitecture at the distal radius and tibia. The use of high-resolution
peripheral scanners (HR-pQCT) is currently limited to research. Its use is yielding
important information on bone deterioration in secondary osteoporosis and related
bone diseases [117]. HR-pQCT can assess BMD, microstructural, and mechanical
parameters of both cortical and trabecular bone separately at the distal radius and
tibia with a very low effective dose, in a very localized field. HR-pQCT is limited to
the appendicular skeleton, but a good correlation has been proven between density
(a-BMD and v-BMD), geometry through cross-sectional area (CSA) and stiffness
parameters measured peripherally and those derived by QCT at lumbar spine and
proximal femur, the sites where the vast majority of osteoporotic fractures occur.
Using HR-pQCT at the distal radius and tibia, a relatively recent study docu-
mented that postmenopausal women with primary hyperparathyroidism (PHPT)
demonstrated thinner cortices, reduced trabecular BMD and cortical BMD in com-
parison with healthy controls; in the PHPT group, analysis of the microarchitecture
(individual trabecular segmentation) documented a large heterogeneity in the distri-
bution of the trabeculae and a depletion of plate-like trabeculae, with a lower tra-
becular plate-to-rod ratio [118].
In postmenopausal women with type 2 DM and history of fragility fractures
HR-pQCT has shown that both cortical bone porosity and pore volume are increased
at the distal radius and distal tibia [119]. The increased cortical porosity and the
impaired trabecular microarchitecture among type 2 DM patients could explain, at
least in part, the high incidence of fragility fractures, even though these patients
display a normal or even elevated BMD in DXA examination [120].
On MRI, the cortical bone is dark (void of signal) because of the small number of
mobile protons and the very short T2 relaxation time. The trabecular bone is also
void of signal, but the surrounding bone marrow has high signal, with intensity
proportional to the amount of fatty content [106].
High-resolution MRI depicts trabecular bone density and structure in vitro and
in vivo with a high spatial resolution, however at resolutions similar to individual
14 Metabolic Bone Disease in Geriatric Patients 389
trabeculae dimension, partial volume effects may arise. MRI is time-consuming and
technically challenging. Since QCT and HR-pQCT can directly depict the trabecu-
lar network, they still appear the most suitable techniques for the investigation of
trabecular bone, but studies comparing high-resolution MRI with QCT and
HR-pQCT have documented that MRI performs equally well with trabecular bone
measurements [121].
The main clinical use of MRI is for the diagnosis of insufficiency/fragility frac-
tures, characterized by the presence of bone marrow edema, due to trabecular dis-
ruption. MRI allows us to confidently rule out malignancy as the cause for fracture
[122]. Optimal sequences are T1-weighted and water sensitive ones, such as fat
suppressed T2-weighted or short tau inversion recovery (STIR) [10].
Localizer images on MRI are a set of three-plane (axial, coronal, and sagittal),
low-resolution, and large field-of-view images that serve to plan the exact position
and angulation of slices of the projected MRI sequences. Localizers, despite their
limited quality, are able to demonstrate incidental vertebral fractures that can be fur-
ther confirmed by subsequently acquired T2-weighted sagittal images [90, 92]. MRI
features also allow discriminate between benign and malignant vertebral fractures
and for the correct identification of recent and old vertebral compression fractures.
14.7 Conclusion
References
1. Baumgartner RN, Stauber PM, McHugh D, Koehler KM, Garry PJ. Cross-sectional age dif-
ferences in body composition in persons 60+ years of age. J Gerontol A Biol Sci Med Sci.
1995;50:M307–16.
2. Hughes VA, Frontera WR, Roubenoff R, Evans WJ, Singh MAF. Longitudinal changes in
body composition in older men and women: role of body weight change and physical activity.
Am J Clin Nutr. 2002;76:473–81.
3. Ley CJ, Lees B, Stevenson JC. Sex- and menopause-associated changes in body-fat distribu-
tion. Am J Clin Nutr. 1992;55:950–4.
4. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–7.
5. Chumlea WC, Baumgartner RN, Garry PJ, Rhyne RL, Nicholson C, Wayne S. Fat distribu-
tion and blood lipids in a sample of healthy elderly people. Int J Obes Relat Metab Disord.
1992;16:125–33.
6. Roche AF. Sarcopenia: a critical review of its measurements and health-related significance
in the middle-aged and elderly. Am J Hum Biol. 1994;6:33–42.
7. Schaap LA, Koster A, Visser M. Adiposity, muscle mass, and muscle strength in relation to
functional decline in older persons. Epidemiol Rev. 2013;35:51–65.
8. Bouxsein ML, Karasik D. Bone geometry and skeletal fragility. Curr Osteoporos Rep.
2006;4:49–56.
9. Adams JE. Imaging of the musculoskeletal system. Pope T. L.; Bloem H. L.; Beltran J.;
Morrison W. B.; Wilson D. J.; Ed. Philadelphia, PA: Saunders Elsevier; 2008. p. 1489–508.
10. Aparisi Gómez MP. Non spinal fragility fractures. Semin Musculoskelet Radiol.
2016;20:330–44.
11. Hamrick MW, McGee-Lawrence ME, Frechette DM. Fatty infiltration of skeletal muscle:
mechanisms and comparisons with bone marrow adiposity. Front Endocrinol. 2016;7:69.
https://doi.org/10.3389/fendo.2016.00069.
12. Butler L, Santoro N. The reproductive endocrinology of the menopausal transition. Steroids.
2011;76:627–35.
13. Gallagher JC. Vitamin D and aging. Endocrinol Metab Clin N Am. 2013;42:319–32.
14. Pereira-Santos M, Costa PRF, Assis AMO, Santos CAST, Santos DB. Obesity and vitamin
D deficiency: a systematic review and meta-analysis: obesity and vitamin D. Obes Rev.
2015;16:341–9.
15. Cheng S, Massaro JM, Fox CS, et al. Adiposity, cardiometabolic risk, and vitamin D status:
the Framingham Heart Study. Diabetes. 2010;59:242–8.
16. Wood RJ. Vitamin D and adipogenesis: new molecular insights. Nutr Rev. 2008;66:40–6.
17. Zemel MB, Richards J, Milstead A, Campbell P. Effects of calcium and dairy on body com-
position and weight loss in African-American adults. Obes Res. 2005;13:1218–25.
18. Garnero P, Munoz F, Sornay-Rendu E, Delmas PD. Associations of vitamin D status with
bone mineral density, bone turnover, bone loss and fracture risk in healthy postmenopausal
women. The OFELY study. Bone. 2007;40:716–22.
19. Snellman G, Melhus H, Gedeborg R, Byberg L, Berglund L, Wernroth L, Michaëlsson
K. Determining vitamin D status: a comparison between commercially available assays.
PLoS One. 2010;5:e11555.
20. Lee S-G, Lee Y-H, Kim KJ, Lee W, Kwon OH, Kim J-H. Additive association of vitamin
D insufficiency and sarcopenia with low femoral bone mineral density in noninstitution-
alized elderly population: the Korea National Health and Nutrition Examination Surveys
2009–2010. Osteoporos Int. 2013;24:2789–99.
21. Tieland M, Brouwer-Brolsma EM, Nienaber-Rousseau C, van Loon LJC, De Groot
LCPGM. Low vitamin D status is associated with reduced muscle mass and impaired physi-
cal performance in frail elderly people. Eur J Clin Nutr. 2013;67:1050–5.
14 Metabolic Bone Disease in Geriatric Patients 391
22. Visser M, Deeg DJH, Lips P. Low vitamin D and high parathyroid hormone levels as determi-
nants of loss of muscle strength and muscle mass (sarcopenia): the longitudinal aging study
Amsterdam. J Clin Endocrinol Metab. 2003;88:5766–72.
23. Bélanger C, Luu-The V, Dupont P, Tchernof A. Adipose tissue intracrinology: potential
importance of local androgen/estrogen metabolism in the regulation of adiposity. Horm
Metab Res. 2002;34:737–45.
24. Kameda T, Mano H, Yuasa T, et al. Estrogen inhibits bone resorption by directly inducing
apoptosis of the bone-resorbing osteoclasts. J Exp Med. 1997;186:489–95.
25. Reid IR. Fat and bone. Arch Biochem Biophys. 2010;503:20–7.
26. Ilich JZ, Kelly OJ, Inglis JE, Panton LB, Duque G, Ormsbee MJ. Interrelationship among
muscle, fat, and bone: connecting the dots on cellular, hormonal, and whole body levels.
Ageing Res Rev. 2014;15:51–60.
27. Zhang P, Peterson M, Su GL, Wang SC. Visceral adiposity is negatively associated with bone
density and muscle attenuation. Am J Clin Nutr. 2015;101:337–43.
28. Ilich JZ, Kelly OJ, Kim Y, Spicer MT. Low-grade chronic inflammation perpetuated by mod-
ern diet as a promoter of obesity and osteoporosis. Arch Ind Hyg Toxicol. 2014;65:139–48.
29. Liu P-Y, Ilich JZ, Brummel-Smith K, Ghosh S. New insight into fat, muscle and bone rela-
tionship in women: determining the threshold at which body fat assumes a negative relation-
ship with bone mineral density. Int J Prev Med. 2014;5:1452–63.
30. Bosch TA, Steinberger J, Sinaiko AR, Moran A, Jacobs DR, Kelly AS, Dengel
DR. Identification of sex-specific thresholds for accumulation of visceral adipose tissue in
adults: threshold accumulation of VAT in adults. Obesity. 2015;23:375–82.
31. Iwaniec UT, Turner RT. Influence of body weight on bone mass, architecture and turnover. J
Endocrinol. 2016;230:R115–30.
32. Scheller EL, Rosen CJ. What’s the matter with MAT? Marrow adipose tissue, metabolism,
and skeletal health. Ann N Y Acad Sci. 2014;1311:14–30.
33. Bredella MA, Fazeli PK, Daley SM, Miller KK, Rosen CJ, Klibanski A, Torriani M. Marrow
fat composition in anorexia nervosa. Bone. 2014;66:199–204.
34. Rosen CJ, Bouxsein ML. Mechanisms of Disease: is osteoporosis the obesity of bone? Nat
Rev Rheumatol. 2006;2:35–43.
35. Jafari Nasabian P, Inglis JE, Reilly W, Kelly OJ, Ilich JZ. Aging human body: changes
in bone, muscle and body fat with consequent changes in nutrient intake. J Endocrinol.
2017;234:R37–51.
36. Aparisi Gómez MP, Ayuso Benavent C, Simoni P, Aparisi F, Guglielmi G, Bazzocchi A. Fat
and bone: the multi perspective analysis of a close relationship. Quant Imag Med Surg.
2020;10:1614–35.
37. Kim TN, Park MS, Ryu JY, et al. Impact of visceral fat on skeletal muscle mass and vice
versa in a prospective cohort study: the Korean Sarcopenic Obesity Study (KSOS). PLoS
One. 2014;9:e115407.
38. Lang T, Cauley JA, Tylavsky F, Bauer D, Cummings S, Harris TB. Computed tomographic
measurements of thigh muscle cross-sectional area and attenuation coefficient predict hip
fracture: the health, aging, and body composition study. J Bone Miner Res. 2010;25:513–9.
39. Stefanaki C, Peppa M, Boschiero D, Chrousos GP. Healthy overweight/obese youth: early
osteosarcopenic obesity features. Eur J Clin Investig. 2016;46:767–78.
40. Mavros Y, Kay S, Simpson KA, et al. Reductions in C-reactive protein in older adults with
type 2 diabetes are related to improvements in body composition following a randomized
controlled trial of resistance training. J Cachexia Sarcopenia Muscle. 2014;5:111–20.
41. Visser M, Goodpaster BH, Kritchevsky SB, Newman AB, Nevitt M, Rubin SM, Simonsick
EM, Harris TB, for the Health ABC Study. Muscle mass, muscle strength, and muscle fat
infiltration as predictors of incident mobility limitations in well-functioning older persons. J
Gerontol Ser A Biol Med Sci. 2005;60:324–33.
392 M. P. Aparisi Gómez et al.
42. Domiciano DS, Figueiredo CP, Lopes JB, Caparbo VF, Takayama L, Menezes PR, Bonfa
E, Pereira RMR. Discriminating sarcopenia in community-dwelling older women with high
frequency of overweight/obesity: the São Paulo Ageing & Health Study (SPAH). Osteoporos
Int. 2013;24:595–603.
43. Ilich JZ, Inglis JE, Kelly OJ, McGee DL. Osteosarcopenic obesity is associated with reduced
handgrip strength, walking abilities, and balance in postmenopausal women. Osteoporos Int.
2015;26:2587–95.
44. Chung JH, Hwang HJ, Shin H-Y, Han CH. Association between sarcopenic obesity and bone
mineral density in middle-aged and elderly Korean. Ann Nutr Metab. 2016;68:77–84.
45. Lee AJ, Hodges S, Eastell R. Measurement of osteocalcin. Ann Clin Biochem. 2000;37:
432–46.
46. Luo X-H, Guo L-J, Xie H, Yuan L-Q, Wu X-P, Zhou H-D, Liao E-Y. Adiponectin stimulates
RANKL and inhibits OPG expression in human osteoblasts through the MAPK signaling
pathway. J Bone Miner Res. 2006;21:1648–56.
47. Lewerin C, Johansson H, Lerner UH, Karlsson MK, Lorentzon M, Barrett-Connor E, Smith
U, Ohlsson C, Mellström D. High serum adiponectin is associated with low blood haemoglo-
bin in elderly men: the Swedish MrOS study. J Intern Med. 2015;278:68–76.
48. Oshima K, Nampei A, Matsuda M, Iwaki M, Fukuhara A, Hashimoto J, Yoshikawa H,
Shimomura I. Adiponectin increases bone mass by suppressing osteoclast and activating
osteoblast. Biochem Biophys Res Commun. 2005;331:520–6.
49. Williams GA, Wang Y, Callon KE, et al. In vitro and in vivo effects of adiponectin on bone.
Endocrinology. 2009;150:3603–10.
50. Karsenty G, Ferron M. The contribution of bone to whole-organism physiology. Nature.
2012;481:314–20.
51. Kanis JA. Diagnosis of osteoporosis and assessment of fracture risk. Lancet.
2002;359:1929–36.
52. Link TM. Osteoporosis imaging: state of the art and advanced imaging. Radiology.
2012;263:3–17.
53. Johnell O, Kanis JA. An estimate of the worldwide prevalence and disability associated with
osteoporotic fractures. Osteoporos Int. 2006;17:1726–33.
54. Office of the Surgeon General (US). Bone health and osteoporosis: a report of the surgeon
general. Rockville, MD: Office of the Surgeon General (US); 2004.
55. Sambrook P, Cooper C. Osteoporosis. Lancet. 2006;367:2010–8.
56. Cauley JA, Hochberg MC, Lui L-Y, Palermo L, Ensrud KE, Hillier TA, Nevitt MC, Cummings
SR. Long-term risk of incident vertebral fractures. JAMA. 2007;298:2761.
57. Bazzocchi A, Guglielmi G. Vertebral fracture identification. Semin Musculoskelet Radiol.
2016;20:317–29.
58. Klotzbuecher CM, Ross PD, Landsman PB, Abbott TA, Berger M. Patients with prior frac-
tures have an increased risk of future fractures: a summary of the literature and statistical
synthesis. J Bone Miner Res. 2000;15:721–39.
59. Clinton J, Franta A, Polissar NL, Neradilek B, Mounce D, Fink HA, Schousboe JT, Matsen
FA. Proximal humeral fracture as a risk factor for subsequent hip fractures. J Bone Joint Surg
Am. 2009;91:503–11.
60. Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. Frailty in elderly people. Lancet.
2013;381:752–62.
61. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J
Gerontol A Biol Sci Med Sci. 2001;56:M146–56.
62. Makizako H, Shimada H, Doi T, Tsutsumimoto K, Suzuki T. Impact of physical frailty
on disability in community-dwelling older adults: a prospective cohort study. BMJ Open.
2015;5:e008462.
63. Fugate Woods N, LaCroix AZ, Gray SL, Aragaki A, Cochrane BB, Brunner RL, Masaki K,
Murray A, Newman AB. Frailty: emergence and consequences in women aged 65 and older
in the women’s health initiative observational study: consequences and predictors of frailty
in WHI women. J Am Geriatr Soc. 2005;53:1321–30.
14 Metabolic Bone Disease in Geriatric Patients 393
64. Yang S, Center JR, Eisman JA, Nguyen TV. Association between fat mass, lean mass, and
bone loss: the Dubbo osteoporosis epidemiology study. Osteoporos Int. 2015;26:1381–6.
65. Walston J, McBurnie MA, Newman A, Tracy RP, Kop WJ, Hirsch CH, Gottdiener J, Fried LP,
Cardiovascular Health Study. Frailty and activation of the inflammation and coagulation sys-
tems with and without clinical comorbidities: results from the Cardiovascular Health Study.
Arch Intern Med. 2002;162:2333–41.
66. Xue Q-L. The frailty syndrome: definition and natural history. Clin Geriatr Med. 2011;27:1–15.
67. Zaslavsky O, Rillamas-Sun E, LaCroix AZ, et al. Association between anthropometric mea-
sures and long-term survival in frail older women: observations from the women’s health
initiative study. J Am Geriatr Soc. 2016;64:277–84.
68. Zaslavsky O, Li W, Going S, Datta M, Snetselaar L, Zelber-Sagi S. Association between
body composition and hip fractures in older women with physical frailty: adiposity and hip
fracture in frailty. Geriatr Gerontol Int. 2017;17:898–904.
69. Yang S, Nguyen ND, Center JR, Eisman JA, Nguyen TV. Association between abdominal
obesity and fracture risk: a prospective study. J Clin Endocrinol Metab. 2013;98:2478–83.
70. Chalhoub D, Cawthon PM, Ensrud KE, et al. Risk of non spine fractures in older adults with
sarcopenia, low bone mass, or both. J Am Geriatr Soc. 2015;63:1733–40.
71. Harris R, Chang Y, Beavers K, et al. Risk of fracture in women with sarcopenia, low bone
mass, or both. J Am Geriatr Soc. 2017;65:2673–8.
72. Bonewald LF, Kiel DP, Clemens TL, Esser K, Orwoll ES, O’Keefe RJ, Fielding RA. Forum on
bone and skeletal muscle interactions: summary of the proceedings of an ASBMR Workshop:
Forum on Bone and Skeletal Muscle Interactions. J Bone Miner Res. 2013;28:1857–65.
73. Chen Z, Lohman TG, Stini WA, Ritenbaugh C, Aickin M. Fat or lean tissue mass: which one
is the major determinant of bone mineral mass in healthy postmenopausal women? J Bone
Miner Res. 1997;12:144–51.
74. Crandall CJ, Yildiz VO, Wactawski-Wende J, Johnson KC, Chen Z, Going SB, Wright NC,
Cauley JA. Postmenopausal weight change and incidence of fracture: post hoc findings from
Women’s Health Initiative Observational Study and Clinical Trials. BMJ. 2015;350:h25.
75. Ensrud KE, Ewing SK, Taylor BC, et al. Frailty and risk of falls, fracture, and mortal-
ity in older women: the study of osteoporotic fractures. J Gerontol A Biol Sci Med Sci.
2007;62:744–51.
76. Grisso JA, Kelsey JL, Strom BL, Ghiu GY, Maislin G, O’Brien LA, Hoffman S, Kaplan
F. Risk factors for falls as a cause of hip fracture in women. N Engl J Med. 1991;324:1326–31.
77. de Rekeneire N, Visser M, Peila R, Nevitt MC, Cauley JA, Tylavsky FA, Simonsick EM,
Harris TB. Is a fall just a fall: correlates of falling in healthy older persons. The Health, Aging
and Body Composition Study. J Am Geriatr Soc. 2003;51:841–6.
78. Fuller GF. Falls in the elderly. Am Fam Physician. 2000;61:2159–68, 2173–2174.
79. Guglielmi G, Muscarella S, Bazzocchi A. Integrated imaging approach to osteoporosis: state-
of-the-art review and update. RadioGraphics. 2011;31:1343–64.
80. Anil G, Guglielmi G, Peh WCG. Radiology of osteoporosis. Radiol Clin N Am.
2010;48:497–518.
81. Singh M, Nagrath AR, Maini PS. Changes in trabecular pattern of the upper end of the femur
as an index of osteoporosis. J Bone Joint Surg Am. 1970;52:457–67.
82. Singh M, Riggs BL, Beabout JW, Jowsey J. Femoral trabecular-pattern index for evaluation
of spinal osteoporosis. Ann Intern Med. 1972;77:63–7.
83. Jhamaria NL, Lal KB, Udawat M, Banerji P, Kabra SG. The trabecular pattern of the calca-
neus as an index of osteoporosis. J Bone Joint Surg (Br). 1983;65:195–8.
84. Adams JE. Radiogrammetry and radiographic absorptiometry. Radiol Clin N Am.
2010;48:531–40.
85. Hyldstrup L, Nielsen SP. Metacarpal index by digital X-ray radiogrammetry: norma-
tive reference values and comparison with dual X-ray absorptiometry. J Clin Densitom.
2001;4:299–306.
86. Oei L, Koromani F, Rivadeneira F, Zillikens MC, Oei EHG. Quantitative imaging methods in
osteoporosis. Quant Imag Med Surg. 2016;6:680–98.
394 M. P. Aparisi Gómez et al.
87. Genant HK, Wu CY, van Kuijk C, Nevitt MC. Vertebral fracture assessment using a semi-
quantitative technique. J Bone Miner Res. 2009;8:1137–48.
88. Vokes T, Bachman D, Baim S, Binkley N, Broy S, Ferrar L, Lewiecki EM, Richmond
B, Schousboe J. Vertebral fracture assessment: the 2005 ISCD official positions. J Clin
Densitom. 2006;9:37–46.
89. Bazzocchi A, Spinnato P, Garzillo G, Ciccarese F, Albisinni U, Mignani S, Battista G, Rossi
C. Detection of incidental vertebral fractures in breast imaging: the potential role of MR
localisers. Eur Radiol. 2012;22:2617–23.
90. Bazzocchi A, Garzillo G, Fuzzi F, Diano D, Albisinni U, Salizzoni E, Battista G, Guglielmi
G. Localizer sequences of magnetic resonance imaging accurately identify osteoporotic ver-
tebral fractures. Bone. 2014;61:158–63.
91. Bazzocchi A, Spinnato P, Albisinni U, Battista G, Rossi C, Guglielmi G. A careful evalua-
tion of scout CT lateral radiograph may prevent unreported vertebral fractures. Eur J Radiol.
2012;81:2353–7.
92. Adams JE. Opportunistic identification of vertebral fractures. J Clin Densitom. 2016;19:54–62.
93. Murata K, Endo K, Aihara T, et al. Artificial intelligence for the detection of vertebral frac-
tures on plain spinal radiography. Sci Rep. 2020;10:20031.
94. Johnell O, Kanis JA, Oden A, et al. Predictive value of BMD for hip and other fractures. J
Bone Miner Res. 2005;20:1185–94.
95. Guglielmi G, Damilakis J, Solomou G, Bazzocchi A. Quality assurance of imaging tech-
niques used in the clinical management of osteoporosis. Radiol Med. 2012;117:1347–54.
96. Harvey NC, Glüer CC, Binkley N, et al. Trabecular bone score (TBS) as a new complemen-
tary approach for osteoporosis evaluation in clinical practice. Bone. 2015;78:216–24.
97. Bazzocchi A, Ponti F, Diano D, Amadori M, Albisinni U, Battista G, Guglielmi G. Trabecular
bone score in healthy ageing. Br J Radiol. 2015;88:20140865.
98. Silva BC, Broy SB, Boutroy S, Schousboe JT, Shepherd JA, Leslie WD. Fracture risk predic-
tion by non-BMD DXA measures: the 2015 ISCD official positions part 2: trabecular bone
score. J Clin Densitom. 2015;18:309–30.
99. Shevroja E, Lamy O, Kohlmeier L, Koromani F, Rivadeneira F, Hans D. Use of trabecular
bone score (TBS) as a complementary approach to dual-energy X-ray absorptiometry (DXA)
for fracture risk assessment in clinical practice. J Clin Densitom. 2017;20:334–45.
100. Silva BC, Leslie WD, Resch H, Lamy O, Lesnyak O, Binkley N, McCloskey EV, Kanis JA,
Bilezikian JP. Trabecular bone score: a noninvasive analytical method based upon the DXA
image. J Bone Miner Res. 2014;29:518–30.
101. Shepherd JA, Schousboe JT, Broy SB, Engelke K, Leslie WD. Executive summary of the
2015 ISCD Position Development Conference on Advanced Measures From DXA and QCT:
Fracture Prediction Beyond BMD. J Clin Densitom. 2015;18:274–86.
102. Guglielmi G, Diacinti D, van Kuijk C, Aparisi F, Krestan C, Adams JE, Link TM. Vertebral
morphometry: current methods and recent advances. Eur Radiol. 2008;18:1484–96.
103. Glüer C-C. Quantitative ultrasound techniques for the assessment of osteoporosis: expert
agreement on current status. J Bone Miner Res. 1997;12:1280–8.
104. Krieg M-A, Barkmann R, Gonnelli S, et al. Quantitative ultrasound in the management of
osteoporosis: the 2007 ISCD official positions. J Clin Densitom. 2008;11:163–87.
105. Guglielmi G, Scalzo G, de Terlizzi F, Peh WCG. Quantitative ultrasound in osteoporosis and
bone metabolism pathologies. Radiol Clin N Am. 2010;48:577–88.
106. Krug R, Burghardt AJ, Majumdar S, Link TM. High-resolution imaging techniques for the
assessment of osteoporosis. Radiol Clin N Am. 2010;48:601–21.
107. Bazzocchi A, Ferrari F, Diano D, Albisinni U, Battista G, Rossi C, Guglielmi G. Incidental
findings with dual-energy X-ray absorptiometry: spectrum of possible diagnoses. Calcif
Tissue Int. 2012;91:149–56.
108. Li N, Li X, Xu L, Sun W, Cheng X, Tian W. Comparison of QCT and DXA: osteoporosis
detection rates in postmenopausal women. Int J Endocrinol. 2013;2013:1–5.
14 Metabolic Bone Disease in Geriatric Patients 395
109. Engelke K, Adams JE, Armbrecht G, et al. Clinical use of quantitative computed tomogra-
phy and peripheral quantitative computed tomography in the management of osteoporosis in
adults: the 2007 ISCD official positions. J Clin Densitom. 2008;11:123–62.
110. Engelke K, Lang T, Khosla S, Qin L, Zysset P, Leslie WD, Shepherd JA, Shousboe
JT. Clinical use of quantitative computed tomography–based advanced techniques in the
management of osteoporosis in adults: the 2015 ISCD official positions—Part III. J Clin
Densitom. 2015;18:393–407.
111. Engelke K, Lang T, Khosla S, Qin L, Zysset P, Leslie WD, Shepherd JA, Schousboe
JT. Clinical use of quantitative computed tomography (QCT) of the hip in the manage-
ment of osteoporosis in adults: the 2015 ISCD official positions—Part I. J Clin Densitom.
2015;18:338–58.
112. Bazzocchi A, Fuzzi F, Garzillo G, Diano D, Rimondi E, Merlino B, Moio A, Albisinni U,
Battista G, Guglielmi G. Reliability and accuracy of scout CT in the detection of vertebral
fractures. Br J Radiol. 2013;86:20130373.
113. Kim YM, Demissie S, Eisenberg R, Samelson EJ, Kiel DP, Bouxsein ML. Intra-and inter-
reader reliability of semi-automated quantitative morphometry measurements and vertebral
fracture assessment using lateral scout views from computed tomography. Osteoporos Int.
2011;22:2677–88.
114. Gruber M, Bauer JS, Dobritz M, Beer AJ, Wolf P, Woertler K, Rummeny EJ, Baum T. Bone
mineral density measurements of the proximal femur from routine contrast-enhanced MDCT
data sets correlate with dual-energy X-ray absorptiometry. Eur Radiol. 2013;23:505–12.
115. Ito M, Oishi R, Fukunaga M, Sone T, Sugimoto T, Shiraki M, Nishizawa Y, Nakamura T. The
effects of once-weekly teriparatide on hip structure and biomechanical properties assessed by
CT. Osteoporos Int. 2014;25:1163–72.
116. Bauer JS, Link TM, Burghardt A, Henning TD, Mueller D, Majumdar S, Prevrhal S. Analysis
of trabecular bone structure with multidetector spiral computed tomography in a simulated
soft-tissue environment. Calcif Tissue Int. 2007;80:366–73.
117. Nishiyama KK, Shane E. Clinical imaging of bone microarchitecture with HR-pQCT. Curr
Osteoporos Rep. 2013;11:147–55.
118. Stein EM, Silva BC, Boutroy S, et al. Primary hyperparathyroidism is associated with abnor-
mal cortical and trabecular microstructure and reduced bone stiffness in postmenopausal
women. J Bone Miner Res. 2013;28:1029–40.
119. Patsch JM, Burghardt AJ, Yap SP, Baum T, Schwartz AV, Joseph GB, Link TM. Increased
cortical porosity in type 2 diabetic postmenopausal women with fragility fractures. J Bone
Miner Res. 2013;28:313–24.
120. Burghardt AJ, Issever AS, Schwartz AV, Davis KA, Masharani U, Majumdar S, Link
TM. High-resolution peripheral quantitative computed tomographic imaging of cortical and
trabecular bone microarchitecture in patients with type 2 diabetes mellitus. J Clin Endocrinol
Metab. 2010;95:5045–55.
121. Krug R, Carballido-Gamio J, Burghardt AJ, Kazakia G, Hyun BH, Jobke B, Banerjee S,
Huber M, Link TM, Majumdar S. Assessment of trabecular bone structure comparing mag-
netic resonance imaging at 3 Tesla with high-resolution peripheral quantitative computed
tomography ex vivo and in vivo. Osteoporos Int. 2008;19:653–61.
122. Musa Aguiar P, Zarantonello P, Aparisi Gómez MP. Differentiation between osteoporotic
and neoplastic vertebral fractures: state of the art and future perspectives. Curr Med Imag.
2021;18:187. https://doi.org/10.2174/1573405617666210412142758.
AL GRAWANY
Body Composition in Geriatric Patients
15
Maria Pilar Aparisi Gómez, Francisco Aparisi,
Giuseppe Guglielmi, and Alberto Bazzocchi
15.1 Introduction
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 397
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_15
398 M. P. Aparisi Gómez et al.
The increase in body fat and the redistribution of the fat in geriatric population
have been demonstrated to be associated with risk factors for non-insulin-dependent
diabetes and cardiovascular (CV) disease [6].
The loss of mineral contents in bone and structural changes are a major risk for
morbidity, need for institutionalization, and mortality. Sarcopenia, with a decrease
in muscle mass and function, has been associated with impaired immunity and func-
tional status [7, 8].
The three tissues (fat, muscle, and bone) have a very close relationship, and
therefore analysis and evaluation should be approached in a combined way.
Several methods are available for the assessment of body composition in geriat-
ric population.
Clinical measures such as BMI, waist circumference, waist-to-hip ratio, under-
water measurement, and bioelectrical impedance (BIA) are widely used as indica-
tors of body adiposity, but growing evidence demonstrates their inaccuracy and
inability to reflect body fat distribution [9].
Imaging tools offer tremendous advantages in research that can be transferred to
clinical practice. From dual-energy X-ray absorptiometry (DXA) and ultrasound to
computed tomography and magnetic resonance, imaging tools allow a more accu-
rate estimation and characterization of body composition.
In this chapter, we aim to summarize the current knowledge on changes in body
composition occurring in the geriatric age and review the possibilities of assessment
that each one of the imaging techniques offers for their adequate measurement in
the geriatric population.
The contents of this chapter need to be taken into consideration together with the
chapter on metabolic bone disease in geriatric patients. The effects of aging on bone
are extensively reviewed in a dedicated chapter, but it is important to acknowledge
the close relationship and interrelation existing between all components.
Aging induces a decrease in basal metabolic rate (estimated as 5–25%) [10], and
this leads to an increase in body weight and body fat, even if there is no change to
the dietary intake. In most individuals, body fat increases gradually between the
ages of 20–25 and 65 [11].
At the same time, there is a redistribution of the adipose tissue to the abdominal
region (android distribution) and visceral organs and fat infiltration into muscle and
bone [12]. Bone marrow fatty infiltration occurs with aging [13], but also in the
context of diabetes, anorexia nervosa, and starvation [14].
The evaluation of fat as a factor in aging is challenging and made worse by the
lack of consensus in the clear cut off for obesity. In the clinical context, obesity clas-
sifications are based on BMI values, but the value of this measurement for classifica-
tion is limited [15]. The quantification of percentage of body fat can be performed
with DXA or bioimpedance analysis (BIA) with much more accuracy, but there is
still no consensus on the percentage of body fat that constitutes the cut off for obesity
15 Body Composition in Geriatric Patients 399
in men or women. The cut off of 32% was suggested recently [16, 17], with the dem-
onstration that 33–38% showed adverse influence on bone mineral density [18].
Muscle and bone tissues decrease with aging. The peak of muscle mass occurs at
approximately 30 years of age, with a gradual decrease after that. By the age of 70,
a decrease in muscle mass of 20–40% can be expected, leading to sarcopenia [19].
Dynapenia constitutes a different concept [20], consisting of loss of muscle strength
and not necessarily muscle mass.
The declines in bone mineral density and muscle mass are more marked in
women [21]. Bone mineral density declines with age, starting at about 50 years. At
the same time, there is an increase in the bone turnover rate, with increased bone
resorption, leading to bone loss [22]. In women, during the 5–7 years after meno-
pause, there is a decrease in bone mass of up to 20%. After this period, in normal
conditions, the rate slows down to 0.5–1% per year (National Osteoporosis
Foundation). For men, the rate is steady at 0.5–1% and starts later in life (National
Osteoporosis Foundation).
The concept of osteosarcopenic obesity syndrome represents a triad of deteriora-
tion of bone, muscle, and adipose tissues [16, 23]. The concept was first coined for
older women, but a recent study points out that this phenotype may exist in younger
overweight populations (18–21 years) [24]. Within this syndrome, two underlining
components have been recognized, osteopenic/osteoporotic obesity [12] and sarco-
penic obesity [16], which can exist separately.
The three tissues (fat, muscle, and bone) have a very close relationship and there-
fore evaluation needs to be done in a combined way.
15.3.1 Fat
With aging, there is a global increase in body fat, up to the age of 50–60 [25].
Subsequently, there is a trend of a reduction in fat mass after the age of 80 [26]. The
loss of skeletal muscle mass contributes to an increase in body fat percentage [27].
An accelerated loss of lean mass has been associated with greater body fatness in
old age [28].
The increase in body fat has an important role in the increase of pro-inflammatory
cytokines. Adipose tissue secretes interleukin (IL)-6 and tumor necrosis factor
(TNF)-alfa [29, 30]. The relative amount of truncal fat measured with DXA corre-
lates with the levels of these markers in plasma [30].
Intraabdominal fat increases with age quantitatively, but proportionally more than
peripheral fat mass [31–33]. The increase of intraabdominal fat starts before the age
of 20 in men and women, accelerating with menopause in women. A study performed
with DXA showed the ratio of upper to lower body fat increases linearly after 20 years.
From 20 to 70, the ratio increases from 1.07 to 1.67 in men and from 0.81 to 1.21 in
women [32]. On a study using CT, the increase of intraabdominal fat at L4 increases
linearly with advancing age, even without significant changes in fat mass [31].
400 M. P. Aparisi Gómez et al.
Besides from the redistribution of body fat, another feature of the changes in
body composition with aging is the infiltration of tissues by fat. This has been
mostly determined for muscles, thanks to the use of MR. Different studies suggest
that the amount of intermuscular fat increases rapidly with age, at a yearly rate of
10% for men and 6% for women. The increase is more noticeable in people with a
global increase in body weight, but it is also seen in people who lose weight [34, 35].
obese twin [45], and subsequently, different studies have demonstrated that leptin’s
effects are largely absent in obese hyperleptinemic state, probably due to resistance
or tolerance [46].
could reflect an increase in muscle mass, for example. In this regard, other clinical
markers, with a better correlation with fat distribution, which in turn has a correla-
tion with specific traits of aging have been used.
As an example, a large waist circumference has been associated with mobility
limitation and disabilities in different studies [81, 82], with a greater association in
inactive older adults [72]. Interestingly, a longitudinal study found that modification
in waist circumference was not associated with a change in self-reported disability
and that the main predictor associated with physical decline was the reduction in
appendicular fat free mas [83].
In fact, muscle fatty infiltration on CT was associated with a higher risk of inci-
dence of mobility limitations in individuals (men and women) over 70 [84, 85].
An increased waist circumference in old adults has been seen to be a predictor of
mortality, even corrected for BMI, and this association was reported to be depen-
dent of cardiorespiratory fitness [86]. In older men, waist circumference has been
reported as a stronger predictor for mortality than BMI. In a study assessing asso-
ciations between BMI, waist circumference, and specific causes for mortality, waist
circumference was the only one to show statistically significant positive associa-
tions with death from major causes (lung cancer, chronic respiratory disease, among
others) [87].
A study using DXA to quantify central adiposity and mortality described a “J”
curve between the two [88]. In a study using CT to quantify visceral fat, the conclu-
sions were similar, with an increased risk for men over the age of 50 [89].
Finally, in the subgroup of very old adults, obesity determined by BMI appears
unrelated to mortality, so it is possible that the relationship between adiposity and
mortality may vary with age [90], but more research is needed in this field.
Sarcopenia is used to refer to the gradual loss of skeletal muscle mass and strength
that takes place with aging. It should be distinguished from cachexia, which corre-
sponds to muscle loss caused by inflammatory diseases and also from the weight
loss and wasting that happens in the context of starvation or advanced disease.
There is growing evidence that sarcopenia occurring with aging has important
consequences in old age, through its association with weakness, disability, and mor-
bidity [91].
In elderly population, the coexistence of superimposed illnesses will act as an
accelerator in the loss of muscle mass, increasing the risk of disability and death.
A unique consensus with the specific cut-off values or the most appropriate tech-
nique for the assessment of low skeletal muscle mass in old adults has not been
reached yet.
DXA has been used to explore changes in total and regional body composition,
including appendicular skeletal muscle mass of legs and arms, which is the sum of
lean mass of legs and arms.
404 M. P. Aparisi Gómez et al.
The deterioration in skeletal muscle mass in the elderly has been measured in
several studies by using CT cross-sectional area, and also whole-body MRI, as tech-
niques that provide an accurate quantification of skeletal muscle mass loss, given
they allow for precise segmentation. The yearly decline has been calculated to be
between 0.64% and 1.29% per year for old men and between 0.53% and 0.84% per
year in old women [28, 83, 91–93].
The loss of muscle mass with age and the increase in body fat put old adults at
risk of developing sarcopenic obesity.
Recent studies have used DXA to demonstrate a general decrease of lean mass at
the upper and lower limbs with age in both genders. The decrease in lean mass was
seen to an increase in fat mass. Lean mass was seen to decrease after 40 years of age
in men, particularly after 50, and after 50 years in women. Women seemed to main-
tain a more favorable lean mass in arms during aging [94].
Anthropometry was also found to not be representative of lean mass of arms in
both genders, independently of age, favoring imaging techniques for the correct
assessment of body composition of the limbs [95].
muscle wasting, establishing a link between age related fat mass redistribution and
sarcopenia [30].
Sarcopenic change also involves fatty infiltration of the skeletal muscle (myoste-
atosis), which comes as a result of estrogen deficiency [100]. The pathways involved
in the development myosteatosis are two. One of them is the accumulation of intra-
cellular lipids within the myofibers (intramyocellular lipids), and the other one is
the disproportioned differentiation of the mesenchymal stem cell population into
the “adipogenic lineage,” which is responsible for the deposit of fat in between
myofibers (inter-myofiber fat). Fatty infiltration in skeletal muscle has a negative
effect on muscle health and function and results in decreased sensitivity to insu-
lin [100].
Current evidence suggests that the role of muscle changes per se is minor [101].
Vitamin D
A decreased intake of calcium, added to poor vitamin D levels and reduced renal
function during aging may result in secondary hyperparathyroidism, which leads to
sarcopenia and reduced strength [102]. Vitamin D controls together with parathy-
roid hormone (PTH) the intestinal absorption of calcium in a negative feedback loop.
Relatively recent studies have demonstrated that vitamin D deficiency is com-
mon in aging [103]. Low levels of vitamin D are therefore associated with impaired
muscle strength, leading to disability and falls, through impairment of the negative
feedback mechanism and increase in PTH levels.
PTH may have a direct effect on skeletal muscle, reducing energy production and
utilization and influencing protein metabolism [104]. PTH also increases free intra-
cellular calcium and decreases plasma phosphate, increasing calcium concentra-
tions and leading to phosphate deficiency in muscle, which alters functionality
[102]. It is hypothesized that this could be one of the pathways through which high
concentration of PTH has been significantly associated with several parameters
implicated in accidental falls and frailty [105].
One of the metabolites of vitamin D is 25-hydroxyvitamin D or calcidiol, a prod-
uct of the conversion of cholecalciferol by hydroxylases in the liver. Low concentra-
tions of calcidiol have been demonstrated to be associated with overall mortality in
older persons. At the same time, the association of high serum concentrations of
PTH and higher overall mortality and cardiovascular mortality has only been dem-
onstrated to be significant in older men, but non-significant in women. Calcidiol and
PTH can therefore be regarded as important health markers [106].
are not always predictable [107, 108]. The loss of skeletal muscle mass and function
can be prevented by specific intervention strategies in the fifth decade of life [109].
Sarcopenia is associated with adverse outcomes through a general increase in
morbidity and mortality, rates of hospitalization, loss of physical ability, and loss of
independence to perform activities of daily life [110, 111].
Regarding mortality, it has been proven that mow muscle mass in the inferior
limbs (measured with CT or DXA) was not strongly associated with a 4.9 years mor-
tality risk in individuals aged 70–79 [124]. The In Chianti study concluded that the
calf muscle area (measured with peripheral quantitative CT) was not associated with
a 6-year mortality risk [125]. The study found no association between sarcopenic
obesity and mortality. A large study of Chinese individuals demonstrated that 5 year
mortality risk between sarcopenic and non-sarcopenic individuals was similar [121].
However, results may be contradictory, a recent 4.6 year follow-up study in a
large population (4331 subjects aged 65–93 years) demonstrated that the loss of
appendicular muscle mass (measured by DXA) was associated with an increased
mortality risk [126].
Numerous clinical methods are in use for the assessment of body composition.
Among the anthropometric methods, BMI measurement has been used as a mea-
surement of body fatness, due to its simplicity; however, it is widely known that it
does not reflect the distribution of body fat. In fact, the analysis of BMI together
with body composition parameters by DXA reveals that groups with very similar
BMI have a different amount of fat, lean, and bone masses [9, 127]. Other clinical
methods such as waist circumference measurement, waist-hip ratio, and other tech-
niques such as underwater weighing and bioimpedance analysis are also available.
The attention of clinicians has turned into imaging methods, however. This is due
to the great advantages regarding reproducibility and accuracy in research. Imaging
methods allow to divide body mass into its components based on their different
physical properties.
Imaging methods are the choice for calibration of field methods designed to mea-
sure adipose tissue and skeletal muscle in vivo and are the only methods that allow
the measurement of internal tissues and organs [128].
Based on the information that is needed, the degree of accuracy, the safety of
assessment, time required, and cost, different imaging methods can be used, such as
DXA, ultrasound, computed tomography, and magnetic resonance. Each one has its
special advantages and limitations (Tables 15.1 and 15.2).
Currently, DXA represents the reference method for the assessment of body
composition. It is fast, involves low radiation exposure, and is unexpensive. It is also
an imaging tool widely used in research [9]. Recently, it has been used in the
NU-AGE study, carried out in five different European countries, among healthy
elderly individuals, showing that body composition characteristics are different
among the elderly in Europe and that a favorable adipose related inflammatory pro-
file is associated with a favorable profile of fat and lean mass markers in body com-
position [127].
408 M. P. Aparisi Gómez et al.
Table 15.1 Summary of imaging markers for assessment of visceral fat, adipose tissue distribu-
tion, and risk of cardiometabolic diseases
Method Parameters Uses Disadvantages
DXA Visceral adipose Positively correlated with clinical Need for advanced
tissue (VAT) and laboratory parameters segmentation tools (otherwise
associated with cardiovascular approximation to android fat)
and metabolic syndrome risk
US Visceral fat Satisfactorily correlated with Standard values have not
thickness clinical and laboratory parameters been determined, and
parameters therefore the application of
Subcutaneous fat Potential use to quantify the technique is limited
thickness intracellular fat in liver
parameters
Intracellular fat
thickness
parameters
CT Linear Excellent spatial resolution Radiation
measurements Cost
(similar to US) Complexity
Cross-sectional Quantification and adipose tissue Currently used in research
Area (CSA) distribution from different body
segments can be achieved with a
high level of accuracy
Volumetric Potential opportunistic use
estimates
MRI Cross-sectional High spatial resolution and Cost
Area (CSA) accuracy Complexity
Volumetric Potential opportunistic use Currently used in research
estimates
Skeletal muscle
fat
Bone marrow fat
Intracellular fat
Table 15.2 Summary of imaging markers for assessment of muscle mass and risk of sarcopenia
Method Parameters Uses Disadvantages
DXA Appendicular Lean mass status could be defined Definition of low lean mass
lean mass using ALMI with Z-scores obtained still has to be established and
index (ALMI) from an age, ethnicity, and validated
sex-matched population
US Muscle Limited use in clinical practice Parameters vary with aging to a
thickness different extent in different
studies. Needs further
validation
Cross- Potential use Operator dependent
sectional Area
(CSA)
Echo intensity
Fascicle length
Pennation
angle
15 Body Composition in Geriatric Patients 409
DXA is a standard technique for the assessment of human BC, with a good level of
correlation between the measurements of skeletal muscle mass at lower limbs
derived by DXA and those derived by CT and MRI [129]. DXA has also been vali-
dated against post-mortem measurement of muscle, skin, and viscera [130].
DXA is based on the physical principle that X-rays of different energies undergo
different attenuation when traversing different tissues. A three compartment model
is generated by radiating the body at multiple different points (pixels) using two
different energies. The pixels that do not contain bone contain a lean mass and fat
mass ratio, and the pixels that contain bone depend on bone mineral content and a
soft tissue ratio with subsequent interpolation of fat mass and lean mass ratio, based
on pixels in vicinity to the ones with bone. In this way, fat mass (FM), non-bone
lean mass (LM), and bone mineral content (BMC) can be obtained. In pixels with-
out bone, soft tissue is further characterized as FM and non-bone LM [131]
(Fig. 15.1).
With DXA, total body, and standard regional (trunk, arms, legs, android, and
gynoid regions) body composition measures can be obtained.
As we have seen, body composition variation with age involves among others a
progressive decrease in LM and increase in FM (sarcopenia and sarcopenic obesity)
and also redistribution of FM to the abdomen. These changes can be monitored by
DXA [117]. An abdominal or android region distribution of FM has been associated
with the development of higher risk profile for cardiovascular and metabolic dis-
eases [132]. The DXA android region was designed to be as representative as pos-
sible of abdominal fat, thus to predict metabolic risk of patients.
DXA with dedicated software allows the analysis of visceral and subcutaneous
adipose tissue (VAT and SAT, respectively) in the android region (a segment of the
abdomen comprised between a lower demarcation line joining the superior limits of
the iliac crests and an upper demarcation line drawn at a level representing 20% of
the distance in between the iliac crests line and the chin). SAT can be estimated and
410 M. P. Aparisi Gómez et al.
Fig. 15.1 DXA analysis of body composition. Measurements are based on a 3-compartment
model that can be simplified into fat mass (FM—yellow), non-bone lean mass (LM—red), and
bone mineral content (BMC—white). Body masses and bone mineral density (BMD) can be
assessed on a regional or a whole-body basis
then subtracted from android total FM to obtain VAT (in grams and volume). DXA-
assessed VAT measurement has been validated against CT in a wide range of age
(18–90 years old) and BMI (18–40 kg/m2) [133] (Fig. 15.2). Most of the risk is
related to VAT compartment, while SAT plays a controversial role. The two fat
depots are distinct in their endocrine function, with different impacts on glucose
metabolism.
DXA specific measurements of LM allow for the calculation of indexes such as
lean mass index (LMI: total LM/height2), appendicular lean mass (ALM: arms
LM + legs LM), and appendicular lean mass index (ALMI: ALM/height2) [134].
ALMI is of clinical significance, because the maintenance of appendicular skele-
tal muscle mass is critical in the preservation of mobility and functional indepen-
dence in advanced age, with subsequent impact on morbidity [135]. According to the
International Society for Clinical Densitometry (ISCD) guidelines, lean mass status
could be defined using ALMI with Z-scores obtained from a young adult, race, and
sex-matched population; however, the threshold for the definition of low LM is yet to
be set and validated [136]. In recent years, age- and sex-specific data on ALM
obtained from general population have been collected in different countries and
could be useful as a reference standard to monitor the loss of muscle mass [95].
Recent several studies in different populations have focused on collecting body
composition data as reference standards, especially in healthy people, to set tools
for comparison on groups of patients affected by different conditions. In this con-
text, postmenopausal status is part of normal aging in healthy women [137, 138].
15 Body Composition in Geriatric Patients 411
Abdominal
VAT SAT
wall
Fig. 15.2 Visceral fat assessment by DXA. DXA with dedicated software allows the analysis of
visceral and subcutaneous adipose tissue (VAT and SAT, respectively) in the android region (a seg-
ment of the abdomen comprised between a lower demarcation line joining the superior limits of
the iliac crests and an upper demarcation line drawn at a level representing 20% of the distance in
between the iliac crests line and the chin) (red area in the drawing and DXA image). SAT can be
estimated and then subtracted from android total FM to obtain VAT (in grams and volume)
non-limb regions of the body, which means it includes other soft tissues in the mea-
surement). Despite this, the measurement of lean mass is frequently used as a proxy
to muscle mass.
DXA may underestimate total lean mass in the body depending on hydration
status (water retention in heart, liver, kidney failure) and overestimate appendicular
lean mass [140].
Individual muscles cannot be evaluated separately, and fatty infiltration cannot
be quantified with DXA, which poses a problem for the detection of sarcopenic
obesity. DXA does not give information of the quality of muscular tissue.
Additionally, different DXA machines can measure slightly different, with dif-
ferent results. Standardization is necessary and the source of a problem. Phantoms
are not anthropometric and cannot be used as reference standards. In vivo cross cali-
bration has been suggested as alternative, but is influenced by many factors, such as
ethnicity and health status, besides from age and gender, for example [141]. Ideally
also, equations to derive lean mass and standardization of local regions of interest
should be standardized across manufacturers, or cross-manufacturer algorithms
developed [142].
Another downside of DXA is not being portable, which can pose a problem
when assessing elderly population.
15.4.2 Ultrasound
The use of ultrasound has been traditionally based on the possibility it offers to
measure thickness of tissue layers. The interfaces between layers are easy to dem-
onstrate. Ultrasound measurement procedures are accurate, reproducible, and fast.
Ultrasound is a simple, low cost, real-time innocuous technique to evaluate body
composition.
Several parameters and indexes of adipose tissue thicknesses may be measured
by ultrasound and have been proved to correlate with clinical and laboratory
parameters.
However, standardization is needed: intraabdominal fat thickness, epicardial
fat thickness, and peri and para renal fat thickness show good accuracy and reli-
ability, with good correlation with CT and MRI-derived areas and volumes
[143]. Abdominal wall fat index, pre-peritoneal fat thickness, and mesenteric fat
thickness demonstrate variable accuracy and reliability in different studies,
when compared to CT [143] (Fig. 15.3). Other indexes, such as the subcutane-
ous fat thickness, showed good correlation with MR- and CT-derived areas and
minimal and maximal subcutaneous fat thickness good correlation with
CT-derived areas [143].
A study demonstrated that reproducibility and repeatability, especially for vis-
ceral fat, were more stable in fasting state and expiration [144].
The different parameters demonstrate a variable correlation with clinical and
laboratory parameters associated with cardiovascular and metabolic risks, and in
different clinical conditions, such as type 2 diabetes mellitus [143].
15 Body Composition in Geriatric Patients 413
Fig. 15.3 Linear measurements of adipose tissue on ultrasound and CT. (a) The minimal abdomi-
nal subcutaneous fat thickness (MinASFT) is the distance between the anterior surface of the linea
alba and the fat-skin barrier, obtained at a plane through the subxiphoid region. (b) Measurement
of the pre-peritoneal fat thickness is performed from the anterior surface of the peritoneum cover-
ing the liver to the posterior surface of the linea alba, at the plane through the subxiphoid region.
(c) The maximum abdominal subcutaneous fat thickness (MaxASFT) is the distance between the
anterior surface of the linea alba and the fat-skin barrier, measured at the level of the supraumbili-
cal region. (d) Most authors measure the intraabdominal fat thickness (IAFT) as the distance from
the posterior wall of the abdominal muscle to the anterior wall of the aorta in the supraumbili-
cal region
cross-sectional area (CSA), echo intensity, fascicle length, and pennation angle of
the lower limbs are the parameters most commonly evaluated by US; in pennate
muscles, the pennation angle (angle formed at the attachment site of the fibers into
deep and superficial aponeurosis) can be evaluated in static and dynamic conditions
and provides information about mechanical and contractile proprieties [150].
All these parameters are affected by aging to a different extent but need to be
further validated. The large majority of the available studies have been conducted
with small samples and in healthy patients. As a result, no validated site-specific
cut-off points for the ultrasound-based assessment of low muscle mass in aging
patients exist.
CT and MRI represent the gold standard to investigate body composition at organ-
tissue level. Quantification of skeletal muscle composition and adipose tissue distri-
bution from different body segments and individual muscle groups can be achieved
with a high level of accuracy using dedicated reconstruction algorithms [151].
Based on the attenuation of an X-ray beam crossing different tissues, a CT scan
can differentiate between fat and fat-free mass. Attenuation values in skeletal mus-
cle tissue may vary between 0 and 100. Low attenuation values are proportional to
the amount of fat within the muscle (normal density muscles show attenuation val-
ues in the range of 31–100 HU, while low-density muscles show attenuation values
in the range of 0–30 HU) [152].
For the assessment of fat and skeletal mass, the most frequently used parameters
are CT-derived cross-sectional area (CSA), and volumetric estimates, and they show
good correlation with cadaver studies [153]. Thickness measurements used on ultra-
sound, in the case of fat mass, can also be applied, with similar correlation with
laboratory and clinical parameters described for ultrasound.
CT has the advantage of being performed routinely for the diagnosis of different
conditions. It therefore allows an opportunistic assessment of body composition.
On an axial CT image obtained at L3, information of total, visceral, subcutane-
ous adipose fat area (and estimation of volume through algorithms) and total psoas
area and skeletal muscle index (SMI) (as an estimation) can be obtained [154]. CT
has been used to derive a predictive cardiometabolic risk, adjusted to gender and
ethnicity [155]. CT has been used to analyze the contribution of pericardial fat,
intrathoracic fat, and epicardial fat to cardiometabolic risk [156].
The specific, targeted use of CT to assess body composition is limited in clinical
practice by the high radiation dose (risk factor for development of neoplasms), high
cost, and operational complexity. On the other side, CT exams performed for other
clinical questions are an incredibly big source for body composition assessment.
Artificial intelligence (AI) techniques are increasingly being developed, which
allow the assessment of body composition from CT images, as collateral informa-
tion from examinations performed for other clinical reasons.
15 Body Composition in Geriatric Patients 415
MRI methods have been used in multiple research studies to gain insights into the
pathophysiology of metabolic diseases including obesity, metabolic syndrome, or
type 2 diabetes mellitus. MRI parameters have been correlated with metabolic con-
trol in diabetes and with diet and physical activity intervention in diabetes [157, 158].
Adipose tissue is characterized by a short T1 and long T2 relaxation time. Fat
appears as bright on T1-weighted sequences because of a high concentration of
immobile protons. Variations of this sequence may be easily applied for the quanti-
fication of SAT, VAT, bone marrow fat, and intermuscular adipose tissue (IMAT).
Currently, whole-body scans can be obtained in approximately 5 min, and these
allow for the detailed quantification of total and regional fat deposits. Whole-body
MR scanning is the most accurate and reproducible protocol to map and measure
the amount of body fat, but is obviously expensive, and significantly time consum-
ing, not manually feasible [159]. The calculation of volumes requires semiauto-
matic segmentation based on signal intensity histograms and thresholds [160].
T2-weighted imaging has been mainly used for fat quantification in the lower
extremity muscles and is not suitable for SAT and VAT determination.
As an alternative, the acquisition of the abdominal region, which allows the mea-
surement of fat depots that are usually associated with cardiometabolic risks has
been proposed [161]. Single- and multi-slice protocols have also been developed to
make analysis faster [161]. Regarding this, the L4–L5 level has been used for single-
slice imaging, but poor prediction of visceral and subcutaneous tissue variation in a
longitudinal study assessing changes with weight loss was reported [162]. A level
close to L2–L3 has been considered as preferred by many groups [163] (Fig. 15.4).
MRI can be used quantitatively. Single-voxel 1H-based MRS has been consid-
ered as non-invasive gold standard for ectopic (organ contained) fat quantification
using PRESS (point resolved spectroscopy) or STEAM (stimulated echo acquisi-
tion mode) sequences. Water and fat signals are identified by their chemical shift
a b
Fig. 15.4 MRI areal measurement of the different fat compartments. (a) T1 FSE image through
the level of L2-L3. (b) segmentation of SAT (yellow), VAT (red), and non-adipose tissue (blue)
(the gas within bowel loops is depicted as black and the bone as white)
416 M. P. Aparisi Gómez et al.
15.5 Conclusion
This chapter has summarized the current knowledge on changes in body composi-
tion occurring in the geriatric age. The process of aging is associated with a modifi-
cation of body composition, with notable consequence on physical abilities and
health. Aging is characterized by a low-grade chronic inflammatory status known as
“inflammaging.”
The increase in body fat and the redistribution of the fat in geriatric population
have been demonstrated to be associated with risk factors for non-insulin-dependent
diabetes and cardiovascular disease, and besides, the loss of mineral contents in
bone and structural changes are a major risk for morbidity, need for institutionaliza-
tion, and mortality. Sarcopenia, with a decrease in muscle mass and function, has
been associated with impaired immunity and functional status. The three tissues
(fat, muscle, and bone) have a very close relationship, and therefore analysis and
evaluation should be approached in a combined way.
Imaging tools offer tremendous advantages in research that can be transferred to
clinical practice. From dual-energy X-ray absorptiometry (DXA) and ultrasound to
computed tomography and magnetic resonance, imaging tools allow a more accu-
rate estimation and characterization of body composition.
The accurate analysis of body composition in the geriatric population offers the
possibility to mitigate some of the negative effects aging has, with the potential to
implement existing interventions, such as exercise and diet, and develop new ways
to promote healthy aging in the future.
References
1. Aparisi Gómez MP, Weidekamm C, Aparisi F, Bazzocchi A. Sports and metabolic bone dis-
ease. Semin Musculoskelet Radiol. 2020;24:277–89.
2. Baumgartner RN, Stauber PM, McHugh D, Koehler KM, Garry PJ. Cross-sectional age dif-
ferences in body composition in persons 60+ years of age. J Gerontol A Biol Sci Med Sci.
1995;50:M307–16.
3. Hughes VA, Frontera WR, Roubenoff R, Evans WJ, Singh MAF. Longitudinal changes in
body composition in older men and women: role of body weight change and physical activity.
Am J Clin Nutr. 2002;76:473–81.
4. Ley CJ, Lees B, Stevenson JC. Sex- and menopause-associated changes in body-fat distribu-
tion. Am J Clin Nutr. 1992;55:950–4.
5. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–7.
418 M. P. Aparisi Gómez et al.
6. Chumlea WC, Baumgartner RN, Garry PJ, Rhyne RL, Nicholson C, Wayne S. Fat distribu-
tion and blood lipids in a sample of healthy elderly people. Int J Obes Relat Metab Disord.
1992;16:125–33.
7. Roche AF. Sarcopenia: a critical review of its measurements and health-related significance
in the middle-aged and elderly. Am J Hum Biol. 1994;6:33–42.
8. Schaap LA, Koster A, Visser M. Adiposity, muscle mass, and muscle strength in relation to
functional decline in older persons. Epidemiol Rev. 2013;35:51–65.
9. Santoro A, Bazzocchi A, Guidarelli G, et al. A cross-sectional analysis of body composition
among healthy elderly from the European NU-AGE study: sex and country specific features.
Front Physiol. 2018;9:1693.
10. St-Onge M-P, Gallagher D. Body composition changes with aging: the cause or the result of
alterations in metabolic rate and macronutrient oxidation? Nutrition. 2010;26:152–5.
11. Hunter GR, Gower BA, Kane BL. Age related shift in visceral fat. Int J Body Compos Res.
2010;8:103–8.
12. Ilich JZ, Kelly OJ, Inglis JE, Panton LB, Duque G, Ormsbee MJ. Interrelationship among
muscle, fat, and bone: connecting the dots on cellular, hormonal, and whole body levels.
Ageing Res Rev. 2014;15:51–60.
13. Aparisi Gómez MP, Ayuso Benavent C, Simoni P, Aparisi F, Guglielmi G, Bazzocchi A. Fat
and bone: the multiperspective analysis of a close relationship. Quant Imag Med Surg.
2020;10:1614–35.
14. Bredella MA, Fazeli PK, Daley SM, Miller KK, Rosen CJ, Klibanski A, Torriani M. Marrow
fat composition in anorexia nervosa. Bone. 2014;66:199–204.
15. Coutinho T, Goel K, Corrêa de Sá D, et al. Central obesity and survival in subjects with coro-
nary artery disease. J Am Coll Cardiol. 2011;57:1877–86.
16. Ilich JZ, Kelly OJ, Inglis JE. Osteosarcopenic obesity syndrome: what is it and how can it be
identified and diagnosed? Curr Gerontol Geriatr Res. 2016;2016:1–7.
17. Wanner M, Martin BW, Autenrieth CS, et al. Associations between domains of physi-
cal activity, sitting time, and different measures of overweight and obesity. Prev Med Rep.
2016;3:177–84.
18. Liu P-Y, Ilich JZ, Brummel-Smith K, Ghosh S. New insight into fat, muscle and bone rela-
tionship in women: determining the threshold at which body fat assumes negative relation-
ship with bone mineral density. Int J Prev Med. 2014;5:1452–63.
19. Kalyani RR, Corriere M, Ferrucci L. Age-related and disease-related muscle loss: the effect
of diabetes, obesity, and other diseases. Lancet Diab & Endocrinol. 2014;2:819–29.
20. Clark BC, Manini TM. Sarcopenia ≠ dynapenia. J Gerontol Ser A Biol Med Sci.
2008;63:829–34.
21. Cruz-Jentoft AJ, Baeyens JP, Bauer JM, et al. Sarcopenia: European consensus on definition
and diagnosis: report of the European Working Group on Sarcopenia in Older People. Age
Ageing. 2010;39:412–23.
22. Guglielmi G, Muscarella S, Bazzocchi A. Integrated imaging approach to osteoporosis: state-
of-the-art review and update. RadioGraphics. 2011;31:1343–64.
23. Jafari Nasabian P, Inglis J, Kelly O, Ilich J. Osteosarcopenic obesity in women: impact, prev-
alence, and management challenges. Int J Women’s Health. 2017;9:33–42.
24. Stefanaki C, Peppa M, Boschiero D, Chrousos GP. Healthy overweight/obese youth: early
osteosarcopenic obesity features. Eur J Clin Investig. 2016;46:767–78.
25. Coin A, Sergi G, Inelmen EM, Enzi G. Pathophysiology of body composition changes in
elderly people. In: Mantovani G, Anker SD, Inui A, Morley JE, Fanelli FR, Scevola D,
Schuster MW, Yeh S-S, editors. Cachexia and wasting: a modern approach. Milan: Springer
Milan; 2006. p. 369–75.
26. Ding J, Kritchevsky SB, Newman AB, et al. Effects of birth cohort and age on body composi-
tion in a sample of community-based elderly. Am J Clin Nutr. 2007;85:405–10.
27. Zamboni M, Zoico E, Scartezzini T, Mazzali G, Tosoni P, Zivelonghi A, Gallagher D, De
Pergola G, Di Francesco V, Bosello O. Body composition changes in stable-weight elderly
subjects: the effect of sex. Aging Clin Exp Res. 2003;15:321–7.
15 Body Composition in Geriatric Patients 419
28. Koster A, Ding J, Stenholm S, et al. Does the amount of fat mass predict age-related loss of
lean mass, muscle strength, and muscle quality in older adults? J Gerontol A Biol Sci Med
Sci. 2011;66:888–95.
29. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis
factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol
Endocrinol Metab. 2001;280:E745–51.
30. Pedersen M, Bruunsgaard H, Weis N, Hendel HW, Andreassen BU, Eldrup E, Dela F,
Pedersen BK. Circulating levels of TNF-alpha and IL-6-relation to truncal fat mass and mus-
cle mass in healthy elderly individuals and in patients with type-2 diabetes. Mech Ageing
Dev. 2003;124:495–502.
31. Zamboni M, Mazzali G, Fantin F, Rossi A, Di Francesco V. Sarcopenic obesity: a new cat-
egory of obesity in the elderly. Nutr Metab Cardiovasc Dis. 2008;18:388–95.
32. Horber FF, Gruber B, Thomi F, Jensen EX, Jaeger P. Effect of sex and age on bone mass,
body composition and fuel metabolism in humans. Nutrition. 1997;13:524–34.
33. Wang Q, Hassager C, Ravn P, Wang S, Christiansen C. Total and regional body-composition
changes in early postmenopausal women: age-related or menopause-related? Am J Clin Nutr.
1994;60:843–8.
34. Rossi AP, Watson NL, Newman AB, Harris TB, Kritchevsky SB, Bauer DC, Satterfield S,
Goodpaster BH, Zamboni M. Effects of body composition and adipose tissue distribution
on respiratory function in elderly men and women: the health, aging, and body composition
study. J Gerontol Ser A Biol Med Sci. 2011;66A:801–8.
35. Delmonico MJ, Harris TB, Visser M, et al. Longitudinal study of muscle strength, quality,
and adipose tissue infiltration. Am J Clin Nutr. 2009;90:1579–85.
36. Björntorp P. Body fat distribution, insulin resistance, and metabolic diseases. Nutrition.
1997;13:795–803.
37. Mykkänen L, Laakso M, Pyörälä K. Association of obesity and distribution of obesity with
glucose tolerance and cardiovascular risk factors in the elderly. Int J Obes Relat Metab
Disord. 1992;16:695–704.
38. Tchernof A, Calles-Escandon J, Sites CK, Poehlman ET. Menopause, central body fat-
ness, and insulin resistance: effects of hormone-replacement therapy. Coron Artery Dis.
1998;9:503–11.
39. Yamanouchi K, Nakajima H, Shinozaki T, Chikada K, Kato K, Oshida Y, Osawa I, Sato J,
Sato Y, Higuchi M. Effects of daily physical activity on insulin action in the elderly. J Appl
Physiol. 1992;73:2241–5.
40. Mittendorfer B, Klein S. Effect of aging on glucose and lipid metabolism during endurance
exercise. Int J Sport Nutr Exerc Metab. 2001;11(Suppl):S86–91.
41. Colberg SR, Simoneau JA, Thaete FL, Kelley DE. Skeletal muscle utilization of free fatty
acids in women with visceral obesity. J Clin Invest. 1995;95:1846–53.
42. Horber FF, Kohler SA, Lippuner K, Jaeger P. Effect of regular physical training on age-
associated alteration of body composition in men. Eur J Clin Investig. 1996;26:279–85.
43. Toth MJ, Tchernof A. Lipid metabolism in the elderly. Eur J Clin Nutr. 2000;54(Suppl
3):S121–5.
44. Kelesidis T, Kelesidis I, Chou S, Mantzoros CS. Narrative review: the role of leptin in human
physiology: emerging clinical applications. Ann Intern Med. 2010;152:93–100.
45. Rönnemaa T, Karonen SL, Rissanen A, Koskenvuo M, Koivisto VA. Relation between plasma
leptin levels and measures of body fat in identical twins discordant for obesity. Ann Intern
Med. 1997;126:26–31.
46. Mantzoros CS, Magkos F, Brinkoetter M, Sienkiewicz E, Dardeno TA, Kim S-Y, Hamnvik
O-PR, Koniaris A. Leptin in human physiology and pathophysiology. Am J Physiol
Endocrinol Metab. 2011;301:E567–84.
47. Beaufrère B, Morio B. Fat and protein redistribution with aging: metabolic considerations.
Eur J Clin Nutr. 2000;54(Suppl 3):S48–53.
420 M. P. Aparisi Gómez et al.
48. Coon PJ, Rogus EM, Drinkwater D, Muller DC, Goldberg AP. Role of body fat distribution
in the decline in insulin sensitivity and glucose tolerance with age. J Clin Endocrinol Metab.
1992;75:1125–32.
49. Cefalu WT, Wang ZQ, Werbel S, Bell-Farrow A, Crouse JR, Hinson WH, Terry JG,
Anderson R. Contribution of visceral fat mass to the insulin resistance of aging. Metabolism.
1995;44:954–9.
50. Cefalu WT, Werbel S, Bell-Farrow AD, Terry JG, Wang ZQ, Opara EC, Morgan T, Hinson
WH, Crouse JR. Insulin resistance and fat patterning with aging: relationship to metabolic
risk factors for cardiovascular disease. Metabolism. 1998;47:401–8.
51. Barzilai N, She L, Liu BQ, Vuguin P, Cohen P, Wang J, Rossetti L. Surgical removal of vis-
ceral fat reverses hepatic insulin resistance. Diabetes. 1999;48:94–8.
52. Barzilai N, Banerjee S, Hawkins M, Chen W, Rossetti L. Caloric restriction reverses hepatic
insulin resistance in aging rats by decreasing visceral fat. J Clin Invest. 1998;101:1353–61.
53. Mourier A, Gautier JF, De Kerviler E, Bigard AX, Villette JM, Garnier JP, Duvallet A,
Guezennec CY, Cathelineau G. Mobilization of visceral adipose tissue related to the improve-
ment in insulin sensitivity in response to physical training in NIDDM. Effects of branched-
chain amino acid supplements. Diabetes Care. 1997;20:385–91.
54. Girard J. [Role of free fatty acids in the insulin resistance of non-insulin-dependent diabetes].
Diabete Metab. 1995; 21:79–88.
55. Boden G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes.
1997;46:3–10.
56. Petroni ML, Caletti MT, Dalle Grave R, Bazzocchi A, Aparisi Gómez MP, Marchesini
G. Prevention and treatment of sarcopenic obesity in women. Nutrients. 2019;11:E1302.
57. Abenavoli L, Peta V. Role of adipokines and cytokines in non-alcoholic fatty liver disease.
Rev Recent Clin Trials. 2015;9:134–40.
58. Pires A, Martins P, Pereira AM, Marinho J, Vaz Silva P, Marques M, Castela E, Sena C, Seiça
R. Pro-inflammatory triggers in childhood obesity: correlation between leptin, adiponectin
and high-sensitivity C-reactive protein in a group of obese Portuguese children. Rev Port
Cardiol. 2014;33:691–7.
59. Isidori AM, Strollo F, Morè M, Caprio M, Aversa A, Moretti C, Frajese G, Riondino G,
Fabbri A. Leptin and aging: correlation with endocrine changes in male and female healthy
adult populations of different body weights. J Clin Endocrinol Metab. 2000;85:1954–62.
60. Gordeladze JO, Drevon CA, Syversen U, Reseland JE. Leptin stimulates human osteoblastic
cell proliferation, de novo collagen synthesis, and mineralization: impact on differentiation
markers, apoptosis, and osteoclastic signaling. J Cell Biochem. 2002;85:825–36.
61. Karsenty G, Ferron M. The contribution of bone to whole-organism physiology. Nature.
2012;481:314–20.
62. Motyl KJ, Rosen CJ. The skeleton and the sympathetic nervous system: it’s about time! J Clin
Endocrinol Metab. 2012;97:3908–11.
63. Upadhyay J, Farr OM, Mantzoros CS. The role of leptin in regulating bone metabolism.
Metabolism. 2015;64:105–13.
64. Hubbard RE, O’Mahony MS, Calver BL, Woodhouse KW. Nutrition, inflammation, and
leptin levels in aging and frailty: nutrition, inflammation, and leptin levels. J Am Geriatr Soc.
2008;56:279–84.
65. Luo X-H, Guo L-J, Xie H, Yuan L-Q, Wu X-P, Zhou H-D, Liao E-Y. Adiponectin stimulates
RANKL and inhibits OPG expression in human osteoblasts through the MAPK signaling
pathway. J Bone Miner Res. 2006;21:1648–56.
66. Lewerin C, Johansson H, Lerner UH, Karlsson MK, Lorentzon M, Barrett-Connor E, Smith
U, Ohlsson C, Mellström D. High serum adiponectin is associated with low blood haemoglo-
bin in elderly men: the Swedish MrOS study. J Intern Med. 2015;278:68–76.
67. Oshima K, Nampei A, Matsuda M, Iwaki M, Fukuhara A, Hashimoto J, Yoshikawa H,
Shimomura I. Adiponectin increases bone mass by suppressing osteoclast and activating
osteoblast. Biochem Biophys Res Commun. 2005;331:520–6.
15 Body Composition in Geriatric Patients 421
68. Williams GA, Wang Y, Callon KE, et al. In vitro and in vivo effects of adiponectin on bone.
Endocrinology. 2009;150:3603–10.
69. Ghoshal K. Adiponectin: probe of the molecular paradigm associating diabetes and obesity.
World J Diabetes. 2015;6:151.
70. Fiaschi T, Giannoni E, Taddei ML, Chiarugi P. Globular adiponectin activates motility and
regenerative traits of muscle satellite cells. PLoS One. 2012;7:e34782.
71. Newman AB, Lee JS, Visser M, Goodpaster BH, Kritchevsky SB, Tylavsky FA, Nevitt M,
Harris TB. Weight change and the conservation of lean mass in old age: the health, aging and
body composition study. Am J Clin Nutr. 2005;82:872–8.
72. Koster A, Patel KV, Visser M, et al. Joint effects of adiposity and physical activity on incident
mobility limitation in older adults: adiposity, physical activity, and mobility limitation. J Am
Geriatr Soc. 2008;56:636–43.
73. Visser M, Langlois J, Guralnik JM, Cauley JA, Kronmal RA, Robbins J, Williamson JD,
Harris TB. High body fatness, but not low fat-free mass, predicts disability in older men and
women: the cardiovascular health study. Am J Clin Nutr. 1998;68:584–90.
74. Zoico E, Di Francesco V, Mazzali G, Zivelonghi A, Volpato S, Bortolani A, Dioli A, Coin
A, Bosello O, Zamboni M. High baseline values of fat mass, independently of appendicular
skeletal mass, predict 2- year onset of disability in elderly subjects at the high end of the
functional spectrum. Aging Clin Exp Res. 2007;19:154–9.
75. Marsh AP, Rejeski WJ, Espeland MA, et al. Muscle strength and BMI as predictors of major
mobility disability in the lifestyle interventions and independence for elders pilot (LIFE-P). J
Gerontol Ser A Biol Med Sci. 2011;66A:1376–83.
76. Houston DK, Ding J, Nicklas BJ, Harris TB, Lee JS, Nevitt MC, Rubin SM, Tylavsky FA,
Kritchevsky SB, for the Health ABC Study. Overweight and obesity over the adult life course
and incident mobility limitation in older adults: the health, aging and body composition
study. Am J Epidemiol. 2009;169:927–36.
77. Arnold AM, Newman AB, Cushman M, Ding J, Kritchevsky S. Body weight dynamics and
their association with physical function and mortality in older adults: the cardiovascular
health study. J Gerontol Ser A Biol Med Sci. 2010;65A:63–70.
78. Flegal KM, Kit BK, Orpana H, Graubard BI. Association of all-cause mortality with over-
weight and obesity using standard body mass index categories: a systematic review and meta-
analysis. JAMA. 2013;309:71.
79. Shaukat A, Dostal A, Menk J, Church TR. BMI is a risk factor for colorectal cancer mortality.
Dig Dis Sci. 2017;62:2511–7.
80. Leigh L, Byles JE, Jagger C. BMI and healthy life expectancy in old and very old women. Br
J Nutr. 2016;116:692–9.
81. Ramsay SE, Whincup PH, Shaper AG, Wannamethee SG. The relations of body composition
and adiposity measures to ill health and physical disability in elderly men. Am J Epidemiol.
2006;164:459–69.
82. Meadows R, Bower JK. Associations of anthropometric measures of obesity with physical
limitations in older adults. Disabil Rehabil. 2020;42:1101–6.
83. Fantin F, Francesco VD, Fontana G, Zivelonghi A, Bissoli L, Zoico E, Rossi A, Micciolo R,
Bosello O, Zamboni M. Longitudinal body composition changes in old men and women: inter-
relationships with worsening disability. J Gerontol Ser A Biol Med Sci. 2007;62:1375–81.
84. Visser M, Goodpaster BH, Kritchevsky SB, Newman AB, Nevitt M, Rubin SM, Simonsick
EM, Harris TB, for the Health ABC Study. Muscle mass, muscle strength, and muscle fat
infiltration as predictors of incident mobility limitations in well-functioning older persons. J
Gerontol Ser A Biol Med Sci. 2005;60:324–33.
85. Reinders I, Murphy RA, Koster A, et al. Muscle quality and muscle fat infiltration in rela-
tion to incident mobility disability and gait speed decline: the age, gene/environment
susceptibility-Reykjavik Study. J Gerontol A Biol Sci Med Sci. 2015;70:1030–6.
86. Sui X, LaMonte MJ, Laditka JN, Hardin JW, Chase N, Hooker SP, Blair SN. Cardiorespiratory
fitness and adiposity as mortality predictors in older adults. JAMA. 2007;298:2507–16.
422 M. P. Aparisi Gómez et al.
87. Leitzmann MF, Moore SC, Koster A, Harris TB, Park Y, Hollenbeck A, Schatzkin A. Waist
circumference as compared with body-mass index in predicting mortality from specific
causes. PLoS One. 2011;6:e18582.
88. Kuk JL, Katzmarzyk PT, Nichaman MZ, Church TS, Blair SN, Ross R. Visceral fat is an
independent predictor of all-cause mortality in men*. Obesity. 2006;14:336–41.
89. Auyeung TW, Lee JSW, Leung J, Kwok T, Leung PC, Woo J. Survival in older men may
benefit from being slightly overweight and centrally obese--a 5-year follow-up study in 4,000
older adults using DXA. J Gerontol Ser A Biol Med Sci. 2010;65A:99–104.
90. Thinggaard M, Jacobsen R, Jeune B, Martinussen T, Christensen K. Is the relationship
between BMI and mortality increasingly U-shaped with advancing age? A 10-year follow-up
of persons aged 70-95 years. J Gerontol A Biol Sci Med Sci. 2010;65:526–31.
91. Hughes VA, Frontera WR, Wood M, Evans WJ, Dallal GE, Roubenoff R, Fiatarone Singh
MA. Longitudinal muscle strength changes in older adults: influence of muscle mass, physi-
cal activity, and health. J Gerontol A Biol Sci Med Sci. 2001;56:B209–17.
92. Frontera WR, Reid KF, Phillips EM, Krivickas LS, Hughes VA, Roubenoff R, Fielding
RA. Muscle fiber size and function in elderly humans: a longitudinal study. J Appl Physiol.
2008;105:637–42.
93. Song M-Y, Ruts E, Kim J, Janumala I, Heymsfield S, Gallagher D. Sarcopenia and increased
adipose tissue infiltration of muscle in elderly African American women. Am J Clin Nutr.
2004;79:874–80.
94. Bazzocchi A, Diano D, Ponti F, Andreone A, Sassi C, Albisinni U, Marchesini G, Battista
G. Health and ageing: a cross-sectional study of body composition. Clin Nutr. 2013;32:569–78.
95. Diano D, Ponti F, Guerri S, Mercatelli D, Amadori M, Aparisi Gómez MP, Battista G,
Guglielmi G, Bazzocchi A. Upper and lower limbs composition: a comparison between
anthropometry and dual-energy X-ray absorptiometry in healthy people. Arch Osteoporos.
2017;12:78.
96. Bua EA, McKiernan SH, Wanagat J, McKenzie D, Aiken JM. Mitochondrial abnormalities
are more frequent in muscles undergoing sarcopenia. J Appl Physiol. 2002;92:2617–24.
97. Payette H, Roubenoff R, Jacques PF, Dinarello CA, Wilson PWF, Abad LW, Harris T. Insulin-
like growth factor-1 and interleukin 6 predict sarcopenia in very old community-living men
and women: the Framingham Heart Study. J Am Geriatr Soc. 2003;51:1237–43.
98. Kolovou G, Katsiki N, Pavlidis A, Bilianou H, Goumas G, Mikhailidis DP. Ageing mecha-
nisms and associated lipid changes. Curr Vasc Pharmacol. 2014;12:682–9.
99. van den Beld AW, Carlson OD, Doyle ME, Rizopoulos D, Ferrucci L, van der Lely AJ, Egan
JM. IGFBP-2 and aging: a 20-year longitudinal study on IGFBP-2, IGF-I, BMI, insulin sen-
sitivity and mortality in an aging population. Eur J Endocrinol. 2019;180:109–16.
100. Hamrick MW, McGee-Lawrence ME, Frechette DM. Fatty infiltration of skeletal muscle:
mechanisms and comparisons with bone marrow adiposity. Front Endocrinol. 2016;7:69.
https://doi.org/10.3389/fendo.2016.00069.
101. Kohrt WM, Holloszy JO. Loss of skeletal muscle mass with aging: effect on glucose toler-
ance. J Gerontol A Biol Sci Med Sci. 1995;50 Spec No:68–72.
102. Genaro PS, Pinheiro MM, Szejnfeld VL, Martini LA. Secondary hyperparathyroidism and
its relationship with sarcopenia in elderly women. Arch Gerontol Geriatr. 2015;60:349–53.
103. on behalf of the IOF Committee of Scientific Advisors (CSA) Nutrition Working Group,
Mithal A, Wahl DA, et al. Global vitamin D status and determinants of hypovitaminosis
D. Osteoporos Int. 2009;20:1807–20.
104. Baczynski R, Massry SG, Magott M, el-Belbessi S, Kohan R, Brautbar N. Effect of parathy-
roid hormone on energy metabolism of skeletal muscle. Kidney Int. 1985;28:722–7.
105. Bird M-L, El Haber N, Batchelor F, Hill K, Wark JD. Vitamin D and parathyroid hormone
are associated with gait instability and poor balance performance in mid-age to older aged
women. Gait Posture. 2018;59:71–5.
15 Body Composition in Geriatric Patients 423
106. El Hilali J, de Koning EJ, van Ballegooijen AJ, Lips P, Sohl E, van Marwijk HWJ, Visser
M, van Schoor NM. Vitamin D, PTH and the risk of overall and disease-specific mortal-
ity: results of the Longitudinal Aging Study Amsterdam. J Steroid Biochem Mol Biol.
2016;164:386–94.
107. Suzuki Y, Murakami T, Haruna Y, Kawakubo K, Goto S, Makita Y, Ikawa S, Gunji A. Effects
of 10 and 20 days bed rest on leg muscle mass and strength in young subjects. Acta Physiol
Scand Suppl. 1994;616:5–18.
108. Nelson ME, Layne JE, Bernstein MJ, et al. The effects of multidimensional home-based
exercise on functional performance in elderly people. J Gerontol A Biol Sci Med Sci.
2004;59:154–60.
109. Roubenoff R. Sarcopenia: a major modifiable cause of frailty in the elderly. J Nutr Health
Aging. 2000;4:140–2.
110. Dennison EM, Sayer AA, Cooper C. Epidemiology of sarcopenia and insight into possible
therapeutic targets. Nat Rev Rheumatol. 2017;13:340–7.
111. Reginster J-Y, Beaudart C, Buckinx F, Bruyère O. Osteoporosis and sarcopenia: two diseases
or one? Curr Opin Clin Nutr Metab Care. 2016;19:31–6.
112. Chase PB, Szczypinski MP, Soto EP. Nuclear tropomyosin and troponin in striated muscle:
new roles in a new locale? J Muscle Res Cell Motil. 2013;34:275–84.
113. Kalinkovich A, Livshits G. Sarcopenia – the search for emerging biomarkers. Ageing Res
Rev. 2015;22:58–71.
114. Abreu EL, Cheng A-L, Kelly PJ, Chertoff K, Brotto L, Griffith E, Kinder G, Uridge T, Zachow
R, Brotto M. Skeletal muscle troponin as a novel biomarker to enhance assessment of the
impact of strength training on fall prevention in the older adults. Nurs Res. 2014;63:75–82.
115. Janssen I. Skeletal muscle cutpoints associated with elevated physical disability risk in older
men and women. Am J Epidemiol. 2004;159:413–21.
116. Jankowski CM, Gozansky WS, Van Pelt RE, Schenkman ML, Wolfe P, Schwartz RS, Kohrt
WM. Relative contributions of adiposity and muscularity to physical function in community-
dwelling older adults. Obesity. 2008;16:1039–44.
117. Rolland Y, Lauwers-Cances V, Cristini C, van Kan GA, Janssen I, Morley JE, Vellas
B. Difficulties with physical function associated with obesity, sarcopenia, and sarcopenic-
obesity in community-dwelling elderly women: the EPIDOS (EPIDemiologie de
l’OSteoporose) study. Am J Clin Nutr. 2009;89:1895–900.
118. Hairi NN, Cumming RG, Naganathan V, Handelsman DJ, Le Couteur DG, Creasey H, Waite
LM, Seibel MJ, Sambrook PN. Loss of muscle strength, mass (sarcopenia), and quality (spe-
cific force) and its relationship with functional limitation and physical disability: the concord
health and ageing in men project: age-related muscle changes and physical function. J Am
Geriatr Soc. 2010;58:2055–62.
119. Bouchard DR, Dionne IJ, Brochu M. Sarcopenic/obesity and physical capacity in older
men and women: data from the nutrition as a determinant of successful aging (NuAge)-the
Quebec Longitudinal Study. Obesity. 2009;17:2082–8.
120. Zoico E, Di Francesco V, Guralnik JM, Mazzali G, Bortolani A, Guariento S, Sergi G, Bosello
O, Zamboni M. Physical disability and muscular strength in relation to obesity and different
body composition indexes in a sample of healthy elderly women. Int J Obes. 2004;28:234–41.
121. Woo J, Leung J, Sham A, Kwok T. Defining sarcopenia in terms of risk of physical limita-
tions: a 5-year follow-up study of 3,153 Chinese men and women: definition of sarcopenia by
incident physical limitation. J Am Geriatr Soc. 2009;57:2224–31.
122. Visser M, Pahor M, Tylavsky F, Kritchevsky SB, Cauley JA, Newman AB, Blunt BA, Harris
TB. One- and two-year change in body composition as measured by DXA in a population-
based cohort of older men and women. J Appl Physiol. 2003;94:2368–74.
123. Santanasto AJ, Glynn NW, Newman MA, Taylor CA, Brooks MM, Goodpaster BH, Newman
AB. Impact of weight loss on physical function with changes in strength, muscle mass, and
muscle fat infiltration in overweight to moderately obese older adults: a randomized clinical
trial. J Obes. 2011;2011:1–10.
424 M. P. Aparisi Gómez et al.
124. Newman AB, Kupelian V, Visser M, Simonsick EM, Goodpaster BH, Kritchevsky SB,
Tylavsky FA, Rubin SM, Harris TB, on Behalf of the Health, Aging and Body Composition
Study Investigators. Strength, but not muscle mass, is associated with mortality in the health,
aging and body composition study cohort. J Gerontol Ser A Biol Med Sci. 2006;61:72–7.
125. Cesari M, Pahor M, Lauretani F, Zamboni V, Bandinelli S, Bernabei R, Guralnik JM, Ferrucci
L. Skeletal muscle and mortality results from the InCHIANTI study. J Gerontol Ser A Biol
Med Sci. 2009;64A:377–84.
126. Lee CG, Boyko EJ, Nielson CM, et al. Mortality risk in older men associated with changes
in weight, lean mass, and fat mass: body composition changes and mortality in older men. J
Am Geriatr Soc. 2011;59:233–40.
127. Santoro A, Guidarelli G, Ostan R, et al. Gender-specific association of body composition with
inflammatory and adipose-related markers in healthy elderly Europeans from the NU-AGE
study. Eur Radiol. 2019;29:4968–79.
128. Ross R. Advances in the application of imaging methods in applied and clinical physiology.
Acta Diabetol. 2003;40:s45–50.
129. Andreoli A, Scalzo G, Masala S, Tarantino U, Guglielmi G. Body composition assessment by
dual-energy X-ray absorptiometry (DXA). Radiol Med. 2009;114:286–300.
130. Buckinx F, Landi F, Cesari M, et al. Pitfalls in the measurement of muscle mass: a need
for a reference standard: measurement of muscle mass. J Cachexia Sarcopenia Muscle.
2018;9:269–78.
131. Toombs RJ, Ducher G, Shepherd JA, De Souza MJ. The impact of recent technological
advances on the trueness and precision of DXA to assess body composition. Obesity (Silver
Spring). 2012;20:30–9.
132. Misra A, Vikram NK. Clinical and pathophysiological consequences of abdominal adiposity
and abdominal adipose tissue depots. Nutrition. 2003;19:457–66.
133. Kaul S, Rothney MP, Peters DM, Wacker WK, Davis CE, Shapiro MD, Ergun DL. Dual-
energy X-ray absorptiometry for quantification of visceral fat. Obesity. 2012;20:1313–8.
134. Guglielmi G, Ponti F, Agostini M, Amadori M, Battista G, Bazzocchi A. The role of DXA in
sarcopenia. Aging Clin Exp Res. 2016;28:1047–60.
135. Janssen I, Heymsfield SB, Ross R. Low relative skeletal muscle mass (sarcopenia) in older
persons is associated with functional impairment and physical disability. J Am Geriatr Soc.
2002;50:889–96.
136. Shepherd JA, Baim S, Bilezikian JP, Schousboe JT. Executive summary of the 2013
International Society for Clinical Densitometry Position Development Conference on Body
Composition. J Clin Densitom. 2013;16:489–95.
137. Päivi M, Mirja H, Terttu P. Changes in physical activity involvement and attitude to physical
activity in a 16-year follow-up study among the elderly. J Aging Res. 2010;2010:1–7.
138. Kyle UG, Genton L, Slosman DO, Pichard C. Fat-free and fat mass percentiles in 5225
healthy subjects aged 15 to 98 years. Nutrition. 2001;17:534–41.
139. Kelly TL, Wilson KE, Heymsfield SB. Dual energy X-ray absorptiometry body composition
reference values from NHANES. PLoS One. 2009;4:e7038.
140. Bazzocchi A, Ponti F, Albisinni U, Battista G, Guglielmi G. DXA: technical aspects and
application. Eur J Radiol. 2016;85:1481–92.
141. Genant HK, Grampp S, Glüer CC, Faulkner KG, Jergas M, Engelke K, Hagiwara S, van
Kuijk C. Universal standardization for dual X-ray absorptiometry: patient and phantom
cross-calibration results. J Bone Miner Res. 2009;9:1503–14.
142. Saarelainen J, Hakulinen M, Rikkonen T, Kröger H, Tuppurainen M, Koivumaa-Honkanen
H, Honkanen R, Hujo M, Jurvelin JS. Cross-calibration of GE healthcare lunar prodigy
and iDXA dual-energy X-ray densitometers for bone mineral measurements. J Osteoporos.
2016;2016:1–11.
143. Bazzocchi A, Filonzi G, Ponti F, Albisinni U, Guglielmi G, Battista G. Ultrasound: which
role in body composition? Eur J Radiol. 2016;85:1469–80.
15 Body Composition in Geriatric Patients 425
164. Tyagi A, Yeganeh O, Levin Y, et al. Intra- and inter-examination repeatability of magnetic
resonance spectroscopy, magnitude-based MRI, and complex-based MRI for estimation
of hepatic proton density fat fraction in overweight and obese children and adults. Abdom
Imaging. 2015;40:3070–7.
165. Eggers H, Börnert P. Chemical shift encoding-based water-fat separation methods: Dixon
Methods. J Magn Reson Imaging. 2014;40:251–68.
166. Fischer Michael A, Pfirrmann CWA, Espinosa N, Raptis DA, Buck FM. Dixon-based MRI for
assessment of muscle-fat content in phantoms, healthy volunteers and patients with achillo-
dynia: comparison to visual assessment of calf muscle quality. Eur Radiol. 2014;24:1366–75.
167. Yang YX, Chong MS, Lim WS, Tay L, Yew S, Yeo A, Tan CH. Validity of estimating muscle
and fat volume from a single MRI section in older adults with sarcopenia and sarcopenic
obesity. Clin Radiol. 2017;72:427.e9–427.e14.
168. Macaluso A, Nimmo MA, Foster JE, Cockburn M, McMillan NC, De Vito G. Contractile
muscle volume and agonist-antagonist coactivation account for differences in torque between
young and older women. Muscle Nerve. 2002;25:858–63.
169. Wong AKO, Beattie KA, Min KKH, Gordon C, Pickard L, Papaioannou A, Adachi JD,
Canadian Multicentre Osteoporosis Study (CaMos) Research Group. Peripheral quantitative
computed tomography-derived muscle density and peripheral magnetic resonance imaging-
derived muscle adiposity: precision and associations with fragility fractures in women. J
Musculoskelet Neuronal Interact. 2014;14:401–10.
170. Lorbergs AL, Noseworthy MD, Adachi JD, Stratford PW, MacIntyre NJ. Fat infiltration in the
leg is associated with bone geometry and physical function in healthy older women. Calcif
Tissue Int. 2015;97:353–63.
171. Addison O, Drummond MJ, LaStayo PC, Dibble LE, Wende AR, McClain DA, Marcus
RL. Intramuscular fat and inflammation differ in older adults: the impact of frailty and inac-
tivity. J Nutr Health Aging. 2014;18:532–8.
172. Gloor M, Fasler S, Fischmann A, Haas T, Bieri O, Heinimann K, Wetzel SG, Scheffler K,
Fischer D. Quantification of fat infiltration in oculopharyngeal muscular dystrophy: compari-
son of three MR imaging methods. J Magn Reson Imaging. 2011;33:203–10.
173. Napolitano A, Miller SR, Murgatroyd PR, Coward WA, Wright A, Finer N, De Bruin TW,
Bullmore ET, Nunez DJ. Validation of a quantitative magnetic resonance method for measur-
ing human body composition. Obesity (Silver Spring). 2008;16:191–8.
174. Erlandson MC, Lorbergs AL, Mathur S, Cheung AM. Muscle analysis using pQCT, DXA and
MRI. Eur J Radiol. 2016;85:1505–11.
Myeloid and Lymphoid Disorders
in Geriatric Patients 16
Patrizia Toia, Massimo Galia, Giuseppe Filorizzo,
Ludovico La Grutta, Federico Midiri, Pierpaolo Alongi,
Emanuele Grassedonio, and Massimo Midiri
16.1 Introduction
Hematological malignancies (HMs) are a type of blood cancer with a large variety
of incidence, etiology, prognosis, and survival.
HMs are the fourth most frequent type of cancer in the world with a high impact
on elderly patients, resulting in considerable morbidity and mortality. Half of all
HMs are diagnosed in patients 65 years of age and older, and this group accounts for
70% of cancer fatalities [1].
An increase in total cancer incidence among older adults is expected [1, 2].
Population aging will have a significant impact on the prevalence of HMs. The
prognosis for older patients with HMs varies widely, depending on personal fea-
tures and treatment options [1].
Aging is a complicated and multifaceted process influenced by both hereditary
and environmental factors; older people are more susceptible to infections, and
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 427
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_16
428 T. Patrizia et al.
The limited literature about imaging features of these disorders may result in under-
diagnosis; however there are some imaging findings related to complications of
MPN, like thrombotic disease, marrow fibrosis, and extramedullary hematopoiesis
(EMH). Although there are no guidelines on the importance of imaging in disease
monitoring, radiologists must be aware of the most common imaging findings.
16.2.2.3 Thrombosis
Thrombosis is a common complication of MPNs that is often detected on imaging.
ET and PV both increase the risk of thrombotic events, which can manifest in a
variety of ways. Arterial thrombi, Budd-Chiari syndrome (BCS), and portal vein
thrombosis can all impair splanchnic circulation [13, 14].
Budd-Chiari syndrome describes the clinical signs of hepatic venous outflow
obstruction, which can occur anywhere between the hepatic veins and the cavoatrial
junction.
BCS is a problem in MPN patients. Intraluminal venous obstruction, such as
venous thrombosis, causes primary BCS, whereas extraluminal venous obstruction,
such as that caused by extrinsic compression, causes secondary BCS. The sharpness
430 T. Patrizia et al.
a b c
Fig. 16.1 Postcontrast CT axial images in chronic lymphocytic leukemia. (a–c) show multiple
abdominal pathological lymph nodes
on imaging, but they are not specific to the condition, instead lymphomas are char-
acterized by many typical radiological features [15].
16.3.1 Lymphomas
16.3.1.2 CT
Computed tomography plays a crucial role in lymphoma imaging.
Normal lymph nodes have an elongated shape and a fatty hilum. Lymph nodes
infiltrated by lymphoma cells are frequently visualized incidentally or during a tar-
geted ultrasonography examination.
At ultrasound, pathologic lymph nodes are often rounded, with a hypoechoic,
pseudocystic appearance caused by the replacement of the node with lymphoma-
tous tissue.
432 T. Patrizia et al.
At computed tomography, involved lymph nodes are often larger and of homo-
geneous density.
The location and subtype of lymphoma determine the imaging features.
Nodal lymphoma is a type of lymphoma affecting only lymph nodes. Extranodal
lymphoma refers to lymphoma that has spread to other tissues. Primary extranodal
lymphoma is a type of lymphoma that affects only one organ, though it can affect
many organs.
Lymphoma can affect many sites at the thoracic level (Fig. 16.3).
Pulmonary lymphoma might present as pulmonary masses or extranodal expan-
sion of thoracic nodal masses and is usually diffuse large B-cell lymphoma or
16 Myeloid and Lymphoid Disorders in Geriatric Patients 433
a b
Fig. 16.4 CT of a 84-year-old man with lymphoma of the left breast in external quadrants (non-
contrast acquisition in a and portal venous phase in b)
marginal zone lymphoma developing from bronchial lymphoid tissue, both are
uncommon. Multiple ill-defined solid or ground glass nodules or masses, consolida-
tion with air bronchograms, and interlobular septal thickening are all common CT
findings in pulmonary lymphoma [23].
Lymphoma can also be found in the breast (Fig. 16.4).
Lymphoma of the abdominal solid organs typically appears on CT scans as solid
masses enhancing at contrast-enhanced CT [24].
Diffuse organ involvement is also conceivable, typically in the liver and spleen,
resulting in organomegaly and different CT attenuation [25].
Most occurrences of splenic involvement in lymphoma are caused by diffuse
large B-cell lymphoma, Hodgkin lymphoma [26], or indolent B-cell lymphomas.
Splenomegaly is the most common imaging sign, but a normal spleen does not rule
out lymphoma involvement.
In the liver, periportal infiltration has been reported in association with hepatic
masses or porto-caval adenopathy. Pancreatic lymphoma is possible even if uncom-
mon, furthermore it may be encased by peripancreatic adenopathy [27].
Renal involvement can manifest as diffuse enlargement or focal renal masses on
imaging.
Gastrointestinal tract lymphoma is frequent in non-Hodgkin lymphoma with dif-
ferent CT scan appearance. Stomach is the most usually affected organ, followed by
small bowel and colon.
Small bowel lymphoma shows a prevalence in the terminal ileum, due to the
enormous proportion of lymphoid tissue at this region. Findings on CT imaging
include localized or multifocal intestinal wall or fold thickening, polyps, ulcers, and
aneurysmal dilatation [27].
Some examples are shown in Figs. 16.5 and 16.6.
Diffuse large B-cell lymphoma or follicular lymphoma are the most common
types of primary lymphoma of the bone. The CT appearance of osseous lymphoma
varies; isolated lesions are often lytic, but they can also be sclerotic, as seen in a
classic “ivory vertebra,” or they might have a mixed lytic/sclerotic appearance [27].
434 T. Patrizia et al.
a b
Fig. 16.5 Post-contrast CT of a 76-year-old man with lymphoma involving ileum (axial image in
a and coronal multiplanar reconstruction in b). In the mesenteric fat a mass with inhomogeneous
density due to the presence of peripheral tissue quota and central colliquate component is observed,
with hydro-air level
The Lugano classification is a lymphoma staging system that applies to both non-
Hodgkin and Hodgkin lymphoma. Presently, it is the most used staging system and
provides criteria for therapy response determined by PET/CT or CT alone.
16 Myeloid and Lymphoid Disorders in Geriatric Patients 435
A universally accepted and reproducible staging system is essential for the stan-
dardized management of patients with malignant lymphomas, in fact clinical staging
plays a larger role in the selection of patients’ treatment than any other clinical factor,
and clinical stage is one of the factors that can be used to predict disease prognosis.
A shared classification also allows a clear response evaluation, guiding therapies.
In this staging system, 18F-fluorodeoxyglucose (18F-FDG) PET/CT has been fully
integrated into the staging and response assessment of FDG-avid lymphoma. CT should
still be used to stage lymphomas with low or variable FDG uptake. Although 18F-FDG
PET/CT is strongly recommended for staging FDG-avid lymphomas, a diagnostic con-
trast-enhanced CT examination should still be included at initial staging for optimal ana-
tomic assessment [28].
CT identifies four categories: (1) complete radiologic response, all lymph nodes
less than or equal to 1.5 cm in longest diameter, and disappearance of all lymphoma
CT findings; (2) partial remission, 50% or greater reduction in disease burden; (3)
stable disease, less than 50% reduction in disease burden; and (4) progressive dis-
ease, new or increased adenopathy or new extranodal lymphoma.
Response assessment with 18F-FDG PET/CT is based on metabolic activity,
indicated by FDG uptake. The International Work Group criteria for reviewing PET
scans were based on visual interpretation and intended for end-of-treatment evalua-
tion, using mediastinal blood pool as the comparator [29].
The Deauville 5-point scale is used by the Lugano classification for documenting
response by 18F-FDG PET/CT ranging from no uptake or residual uptake to signifi-
cantly increased uptake or any new lesion.
Table 16.1 summarizes the Lugano criteria according to CT and 18F-FDG PET/
CT [29].
Recent evidences suggest that CT examination may be useful in patients with HL
who have a favorable interim or post-treatment PET-CT, with a smaller tumor mass
corresponding to a better result [30, 31]. Response evaluation based on CT is
favorite for histologies with low or variable FDG avidity, as well as in regions where
PET-CT is not available [31].
Other recommendations are included in the Lugano classification.
Particularly, although 18F-FDG PET/CT is widely accepted as the gold standard
for FDG-avid lymphomas, the Lugano classification recognizes the relevance of CT
for anatomic assessment, recommending contrast-enhanced CT for initial staging
and radiation therapy planning.
At the time of baseline staging, the tumor burden will be estimated. Up to six
lymphoma nodes, nodal complexes, or other lymphoma deposits are selected. The
lesions picked must be able to be measured accurately in two dimensions.
Although the Lugano classification considers detectable an adenopathy with longest
nodal diameter of more than 1.5 cm, radiologists must be aware that a lymph node
smaller than 1.0 cm with avid FDG uptake could be connected with lymphoma [29].
Splenomegaly is defined when spleen is >13 cm.
Changes in metabolic activity, expressed by SUV, can be used to quantify
response on 18F-FDG PET/CT scans. The highest SUV in any lesion on the base-
line and follow-up scan is usually measured.
Furthermore, radiologists must be aware that CT and 18F-FDG PET/CT findings
may be discordant, as in case of a significant reduction in tumor burden at CT and
increased SUV at 18F-FDG PET/CT [7].
In conclusion, the Lugano classification is used as a unified guideline for all cli-
nicians involved in lymphoma diagnosis and care because it reflects a consensus
statement of clinical experts in lymphoma. Therefore, radiologists and nuclear med-
icine specialists have a better chance of guiding clinical management based on
imaging findings thanks to these criteria [7].
Figures 16.7 and 16.8 show an example of an 18F-FDG PET/CT performed in an
old patient with a diffuse large B-cell NHL.
a b
c d
Fig. 16.7 Maximum intensity projection (MIP) representation of 18F-FDG PET (a); axial section
of PET images corrected for attenuation (b); axial section of low-dose CT used for attenuation
correction of PET images (c); axial section hybrid PET/CT images showing multiple retroperito-
neal and mesenteric pathological lymph nodes and diffuse pathological metabolic activity of the
lower pole of the spleen (d)
16 Myeloid and Lymphoid Disorders in Geriatric Patients 437
a b c
d e f
Fig. 16.8 Coronal and sagittal section of low-dose CT used for correction attenuation (a, d);
Coronal and sagittal section hybrid PET/CT images showing multiple supra and subdiaphragmatic
pathological lymph nodes, splenic and bone disease with high metabolic activity (b, e); coronal
and sagittal sections of PET images corrected for attenuation (c, f)
The functional assessment is based on DWI, which involves the study of random
Brownian motion of water molecules in biological tissues, as measured by the
apparent diffusion coefficient (ADC) [34].
Lymphoma is characterized by increased cellularity and an elevated nuclear-to-
cytoplasm ratio, resulting in restricted water molecule transport compared to normal
tissues, high signal intensity on DWI, and low ADC values [35].
In addition to standard dimension criteria, different WB-MRI criteria have been
established for the evaluation of lymph node involvement [36] such as DWI signal
greater than that of the spinal cord or muscles, high signal intensity at higher b val-
ues with restriction confirmed by low ADC or in the presence of central necrosis,
regardless of dimension; coalesces into a large nodal mass [37].
Despite this, there are no clear established ADC values to distinguish normal
lymph nodes from pathological ones; furthermore, no consensus has been reached
about mean or minimum ADC values to use in clinical practice.
WB-MRI has also been suggested by some authors as the best imaging tool for
monitoring indolent lymphomas (i-NHLs) and aggressive lymphomas in complete
remission [38].
Due to artifacts on DWI caused by heart pulse and respiration, WB-MRI has
demonstrated some issues in evaluating tiny mediastinal and pulmonary hilar lymph
nodes, with ADC values being miscalculated [39]. Furthermore, due to the anisotro-
pic physiologically constrained pattern of diffusion of normal splenic parenchyma
on DWI, characterization of focal splenic lesions by WB-MRI can be difficult, so
DWI must be combined with standard morphologic WB-MRI images for the evalu-
ation of the spleen [40].
In lymphoma, gadolinium-based contrast agents may increase the accuracy in
identifying parenchymal lesions during WB-MRI; it appears to be especially useful
in case of high probability of extranodal localization [40, 41].
The time required for a WB-MRI depends on the MRI unit and imaging tech-
nique and might take from 30 minutes to more than an hour.
The entire procedure takes less time than 18F-FDG PET/CT [42, 43].
WB-MRI, 18F-FDG PET/CT, and CT all have a high level of patient acceptance
when it comes to patient compliance. Claustrophobia, caused by the anxiety of
being in the enclosed area of a MRI machine for an extended period of time, is a
serious issue for patients undergoing WB-MRI [44].
WB-MRI can be used in HL patients as a complementary imaging modality to
replace contrast-enhanced CT in diagnostic work-up and lymphoma surveillance,
but it is unlikely to replace 18F-FDG PET/CT for HL staging and response assess-
ment at this time.
T2 signal intensity and contrast enhancement of lymphomatous lesions tend to
decrease following treatment, T2 signal reduction may be linked to fibrotic stroma
and collagen; however, immature fibrotic tissue, necrosis, or edema might cause an
increase in T2 signal, limiting its use in this setting [43].
WB-MRI has also been demonstrated to be useful in detecting certain recently
documented sequelae, such as osteonecrosis, in HL patients receiving chemother-
apy regimens that include large doses of corticosteroids.
16 Myeloid and Lymphoid Disorders in Geriatric Patients 439
a b c
Fig. 16.9 Axial postcontrast 3D-GRE T1-weighted fat suppressed (a), axial DWI b-value 800 s/
mm2 (b) and coronal maximum intensity projection (MIP) diffusion-weighted imaging (DWI) (c):
multiple lymph nodes increased in size with a tendency to confluence are shown, the largest in the
superior mediastinum and right axillary region
a b c
Fig. 16.10 (a–c) A 66-year-old man with non-Hodgkin’s lymphoma. Axial postcontrast 3D-GRE
T1-weighted fat-suppressed images: multiple lymph nodes increased in size are shown, in the right
axillary site; in the sub-diaphragmatic site there are multiple confluent lymph nodes in the paraca-
val, interaortocaval, and left para-aortic site
440 T. Patrizia et al.
16.3.3.2 CT
Unenhanced low-dose CT is commonly used to diagnose bone involvement in MM
because of the strong intrinsic contrast of bony structures. The slightly higher radia-
tion dosage is tolerable in view of the much higher sensitivity and improved patient
comfort in the typically older patient population. CT has additional benefits, such as
enhanced fracture risk assessment and the ability to visualize extraosseous myeloma,
in addition to its excellent sensitivity [47].
Some osteolytic lesions in a patient with multiple myeloma are showed in
Figs. 16.11 and 16.12.
a b
c d
Fig. 16.11 (a–d) A 73-year-old woman with multiple myeloma. Low-dose CT scan images show
multiple and widespread bone lesions of a lytic character, affecting almost all the skeletal seg-
ments, between a few millimeters in size and 5 cm, the lesion at the level of the left ischium deter-
mines swollen appearance of the bone with thinning and interruption of the cortical profile
16 Myeloid and Lymphoid Disorders in Geriatric Patients 441
a b
c d
e f
Fig. 16.12 (a–f) A 77-year-old man with multiple myeloma. CT imaging shows multiple osteo-
lytic lesions and right pleural effusion
16.3.3.3 PET/CT
PET/CT imaging using 18F-fluorodeoxyglucose visualizes glucose hypermetabo-
lism in medullary and extramedullary myeloma; it also allows the morphological
detection of osteolysis as an additional functional component. Furthermore, PET/
CT allows prognostic statements during the initial diagnosis and treatment [48].
16.3.3.4 MRI
Because nearly half of all patients have focal lesions outside of the axial skeleton,
MRI is frequently conducted as a whole-body evaluation including the extremi-
ties [49].
442 T. Patrizia et al.
A routine evaluation normally includes coronal and sagittal T1w and T2w
sequences, as well as fat-saturated T2w sequences.
On MRI, five different infiltration patterns in myeloma patients can be distin-
guished (Table 16.2), and the predictive value of MRI infiltration patterns has been
demonstrated in several studies.
A normal pattern of the bone marrow, or a “salt and pepper” pattern, was corre-
lated to an early stage of the disease and a better prognosis. Table 16.2 summarizes
the radiological features of the various patterns [50].
16.4 Conclusions
The growing expectancy of life is one of the main contributors for the increasing
number of older patients with hematologic malignancies.
The role of radiologist is to stage the disease, evaluating the level of frailty, but
also to identify possible complications and to assess therapeutic response.
Universally accepted and reproducible staging system is essential for the standard-
ized response evaluation.
Magnetic resonance is becoming even more important in clinical practice, espe-
cially in lymphoid disorders.
References
1. Krok-Schoen JL, Fisher JL, Stephens JA, et al. Incidence and survival of hematological can-
cers among adults ages ≥75 years. Cancer Med. 2018;7(7):3425–33.
2. Wang H, Dwyer-Lindgren L, Lofgren KT, Rajaratnam JK, et al. Age-specific and sex-specific
mortality in 187 countries, 1970-2010: a systematic analysis for the Global Burden of Disease
Study 2010. Lancet. 2012;380(9859):2071–94.
3. Bron D, Ades L, Fulop T, et al. Aging and blood disorders: new perspectives, new challenges.
Haematologica. 2015;100(4):415–7.
4. Vardiman JW. The World Health Organization (WHO) classification of tumors of the hema-
topoietic and lymphoid tissues: an overview with emphasis on the myeloid neoplasms. Chem
Biol Interact. 2010;184(1–2):16–20.
5. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization
(WHO) classification of lymphoid neoplasms. Blood. 2016;127:2375.
16 Myeloid and Lymphoid Disorders in Geriatric Patients 443
6. Scheepers ERM, Vondeling AM, Thielen N, et al. Geriatric assessment in older patients with a
hematologic malignancy: a systematic review. Haematologica. 2020;105(6):1484–93.
7. Johnson SA, Kumar A, Matasar MJ, et al. Imaging for staging and response assessment in
lymphoma. Radiology. 2015;276(2):323–38.
8. Liput J, Smith DA, Beck R, et al. Myeloproliferative neoplasms: a primer for radiologists. J
Comput Assist Tomogr. 2019;43:652–63.
9. Extermann M, Aapro M, Bernabei R, et al. Use of comprehensive geriatric assessment in older
cancer patients: recommendations from the task force on CGA of the International Society of
Geriatric Oncology (SIOG). Crit Rev Oncol Hematol. 2005;55(3):241–25.
10. Murphy IG, Mitchell EL, Raso-Barnett L, et al. Imaging features of myeloproliferative neo-
plasms. Clin Radiol. 2017;72:801–9.
11. Hanrahan CJ, Shah LM. MRI of spinal bone marrow: part 2, T1-weighted imaging-based dif-
ferential diagnosis. AJR Am J Roentgenol. 2011;197:1309e21.
12. Orphanidou-Vlachou E, Tziakouri-Shiakalli C, Georgiades CS. Extramedullary hemopoiesis.
Semin Ultrasound CT MR. 2014;35:255e62.
13. Potthoff A, Attia D, Pischke S, et al. Long-term outcome of liver transplant patients with Budde
Chiari syndrome secondary to myeloproliferative neoplasms. Liver Int. 2015;35:2042e9.
14. Qi X, De Stefano V, Senzolo M, et al. Splanchnic vein thrombosis: etiology, diagnosis, and
treatment. Gastroenterol Res Pract. 2015;2015:506136e2.
15. Hallek M. Chronic lymphocytic leukemia: 2017 update on diagnosis, risk stratification, and
treatment. Am J Hematol. 2017;92(9):946–65.
16. Matasar MJ, Zelenetz AD. Overview of lymphoma diagnosis and management. Radiol Clin N
Am. 2008;46(2):175–98, vii.
17. Costello P, Mauch P. Radiographic features of recurrent intrathoracic Hodgkin’s disease fol-
lowing radiation therapy. AJR Am J Roentgenol. 1979;133:201–6.
18. Lee CKK, Bloomfield CD, Goldman AI, et al. Prognostic significance of mediastinal involve-
ment in Hodgkin’s disease treated with curative chemotherapy. Cancer. 1980;46:2403–9.
19. Anderson H, Jenkins JPR, Brigg DJ, et al. The prognostic significance of mediastinal bulk
in patients with stage IA-IVB Hodgkin’s disease: a report from the Manchester Lymphoma
Group. Clin Radiol. 1985;36:449–54.
20. Lister TA, Crowther D, Sutcliffe SB, et al. Report of a committee convened to discuss the
evaluation and staging of patients with Hodgkin’s disease: Cotswolds meeting. J Clin Oncol.
1989;7:1630–6.
21. Thar TL, Millon RR, Hausner RJ, et al. Hodgkin’s disease, stages I and II: relationship
of recurrence to size of disease, radiation dose, and the number of sites involved. Cancer.
1979;43:1101–5.
22. Castellino RA, Hilton S, O’Brien JP, et al. Non Hodgkin’s lymphoma: contribution of chest CT
in the initial staging evaluation. Radiology. 1996;199:129–32.
23. Hare SS, Souza CA, Bain G, et al. The radiological spectrum of pulmonary lymphoprolifera-
tive disease. Br J Radiol. 2012;85(1015):848–64.
24. Fishman EK, Kuhlman JE, Jones RJ. CT of lymphoma: spectrum of disease. RadioGraphics.
1991;11(4):647–69.
25. Anis M, Irshad A. Imaging of abdominal lymphoma. Radiol Clin N Am. 2008;46(2):265–85,
viii–ix.
26. Saboo SS, Krajewski KM, O’Regan KN, et al. Spleen in haematological malignancies: spec-
trum of imaging findings. Br J Radiol. 2012;85(1009):81–92.
27. Chua SC, Rozalli FI, O’Connor SR. Imaging features of primary extranodal lymphomas. Clin
Radiol. 2009;64(6):574–88.
28. Cheson BD, Pfistner B, Juweid ME, et al. Revised response criteria for malignant lymphoma.
J Clin Oncol. 2007;25:579–86.
29. Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging,
and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification.
J Clin Oncol. 2014;32(27):3059–68.
444 T. Patrizia et al.
30. Sattarivand M, Caldwell C, Poon I, et al. Effects of ROI placement on PET based assessment
of tumor response to therapy. Int J Mol Imag. 2013;2013:132804.
31. Rossi C, Kanoun S, Berriolo-Riedinger A, et al. Interim 18F-FDG PET SUVmax reduction is
superior to visual analysis in predicting outcome early in Hodgkin lymphoma patients. J Nucl
Med. 2014;55(4):569–73.
32. Albano D, Bruno A, Patti C, et al. Whole body magnetic resonance imaging (WB-MRI) in
lymphoma: state of the art. Hematol Oncol. 2020;38:12–21.
33. Galia M, Albano D, Narese D, et al. Whole-body MRI in patients with lymphoma: collateral
findings. Radiol Med. 2016;121(10):793–800.
34. Chianca V, Albano D, Messina C, et al. Diffusion tensor imaging in the musculoskeletal
and peripheral nerve systems: from experimental to clinical applications. Eur Radiol Exp.
2017;1(1):12.
35. Kwee TC, Basu S, Torigian DA, Nievelstein RA, Alavi A. Evolving importance of diffusion-
weighted magnetic resonance imaging in lymphoma. PET Clin. 2012;7(1):73–82.
36. Stecco A, Buemi F, Iannessi A, et al. Current concepts in tumor imaging with whole-body
MRI with diffusion imaging (WB-MRI-DWI) in multiple myeloma and lymphoma. Leuk
Lymphoma. 2018;12(11):1–11.
37. Albano D, Patti C, Lagalla R, et al. Whole-body MRI, FDG-PET/CT, and bone marrow biopsy,
for the assessment of bone marrow involvement in patients with newly diagnosed lymphoma.
J Magn Reson Imaging. 2017;45(4):1082–9.
38. Mayerhoefer ME, Karanikas G, Kletter K, et al. Evaluation of diffusion-weighted MRI for
pretherapeutic assessment and staging of lymphoma: results of a prospective study in 140
patients. Clin Cancer Res. 2014;20(11):2984–93.
39. Chavhan GB, Babyn PS. Whole-body MR imaging in children: principles, technique, current
applications, and future directions. Radiographics. 2011;31(6):1757–72.
40. Plathow C, Walz M, Lichy MP, et al. Cost considerations for whole body MRI and PET/CT as
part of oncologic staging. Radiologe. 2008;48(4):384–96.
41. Albano D, Agnello F, Patti C, et al. Whole-body magnetic resonance imaging and FDG-
PET/CT for lymphoma staging: assessment of patient experience. Egypt J Radiol Nucl Med.
2017;48(4):1043–7.
42. Adams HJ, Kwee TC, Vermoolen MA, Ludwig I, Bierings MB, Nievelestein RA. Whole-body
MRI vs. CT for staging lymphoma: patient experience. Eur J Radiol. 2014;83(1):163–6.
43. Galia M, Albano D, Tarella C, et al. Whole body magnetic resonance in indolent lymphomas
under watchful waiting: the time is now. Eur Radiol. 2018;28(3):1187–93.
44. Rahmouni A, Divine M, Kriaa S, Haioun C, Anglade MC, Kobelter H. Lymphoma: imaging in
the evaluation of residual masses. Cancer Imaging. 2002;2(2):93–5.
45. Moreau P, San Miguel J, Sonneveld P, et al. Multiple myeloma: ESMO clinical practice guide-
lines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28:iv52–61.
46. Terpos E, Kleber M, Engelhardt M, et al. European myeloma network guidelines for the man-
agement of multiple myeloma-related complications. Haematologica. 2015;100:1254–66.
47. Gleeson TG, Moriarty J, Shortt CP, et al. Accuracy of whole-body low dose multidetector CT
(WBLDCT) versus skeletal survey in the detection of myelomatous lesions, and correlation of
disease distribution with whole-body MRI (WB-MRI). Skelet Radiol. 2009;38:225–36.
48. Dimopoulos MA, Hillengass J, Usmani S, et al. Role of magnetic resonance imaging in
the management of patients with multiple myeloma: a consensus statement. J Clin Oncol.
2015;33:657–64.
49. Kosmala A, Bley T, Petritsch B, et al. Bildgebende diagnostik des multiplen myeloms. Fortschr
Röntgenstr. 2019;191:805–16.
50. Spinnato P, Bazzocchi A, Brioli A, et al. Contrast enhanced MRI and 18F FDG PET-CT in the
assessment of multiple myeloma: a comparison of results in different phases of the disease.
Eur J Radiol. 2012;81:4013–8.
The Role of Artificial Intelligence (AI)
in the Management of Geriatric Patients 17
Salvatore Claudio Fanni, Sherif Mohsen Shalaby,
and Emanuele Neri
17.1 Introduction
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 445
G. Guglielmi, M. Maas (eds.), Imaging in Geriatrics, Practical Issues in
Geriatrics, https://doi.org/10.1007/978-3-031-14877-4_17
446 S. C. Fanni et al.
AL GRAWANY
17 The Role of Artificial Intelligence (AI) in the Management of Geriatric Patients 447
Feng et al. changed the target for radiomics features extraction to the corpus callo-
sum, as it is a major site of disease in the late stage of AD. A logistic regression
model was implemented to analyze 385 texture features, with a modest accuracy of
70% in the detection of AD versus controls [21].
As AD is a neurodegenerative disorder with a widespread involvement of the
entire brain, the next natural step was the application of radiomics features extrac-
tion to a series of brain’s region. Nanni et al. achieved an AUC of 94% in differen-
tiating AD versus controls and an interesting 70% in detecting the patients with
MCI who faster convert to AD, combining texture and volume features of AD
brain [22].
PD is the second most common neurodegenerative disorder and, as AD, an
increasing social and economic burden is expected on our society. Shinde et al. in
2019 implemented a CNN to detect PD subject analyzing neuromelanin-sensitive
MR images. The CNN showed an interesting performance, locating the most impor-
tant site involved in the disease, differentiating PD from healthy subject with an
accuracy of 80% and even from other parkinsonian syndrome with accuracy of
85.7%. The CNN has outperformed the radiomics classifier with an accuracy of
about 60.3% [23].
As for the neurodegenerative disorders, even stroke-related burden on healthcare
services is going to increase due to aging of population, with an upcoming and wor-
rying rise in terms of mortality, disability, and economics costs [24].
Neuroimaging, using CT and MRI, plays a pivotal role even in stroke manage-
ment, as they are crucial to select subject for the most appropriate therapy. Radiomics
and machine learning algorithm are expected to manage the incoming increased
workload and to improve early diagnosis, prediction of early outcomes, and evalua-
tion of long-term prognosis [25]. As ischemic stroke accounts for approximately
85% of all stroke cases [26], we will focus on applications of AI in management of
this entity. Nonenhanced computed tomography (NECT) is the first imaging choice
in patients with episode of neurologic deficit. As far as early diagnosis is vital to
rapidly treat the patients with the thrombolytic agents, it still remains extremely
challenging because the changes in ischemic area often are not easily detectable.
Texture analysis has been implemented in order to differentiate healthy tissue from
ischemic lesion. With this purpose, Peter et al. built a machine learning model with
six texture features extracted from NECT, achieving an AUC of 0.82 [27].
Texture features were also evaluated as a predictor for secondary intracranial
hemorrhage in patients with acute ischemic stroke. In a study of Kassner et al. tex-
ture parameters extracted from MR T1-weighted images achieved an AUC > 0.75,
resulting in a great predictive ability for early outcomes and outperforming visual
enhancement score (AUC < 0.6) [28]. Furthermore, radiomics features were ana-
lyzed to evaluate the long-term prognosis. Betrouni et al. demonstrated a correlation
between the 6-month cognitive impairment and radiomics features extracted at 72 h
after stroke from MR images of hippocampus and entorhinal cortex. The support
vector machine algorithm achieved an interesting AUC of 0.9 [29].
17 The Role of Artificial Intelligence (AI) in the Management of Geriatric Patients 449
In this section, we will be addressing lung and cardiovascular diseases which can
benefit from the potential role of AI in the imaging management of these disease.
Chest CT is a medical imaging technique widely performed in elder people,
mostly to diagnose, stage, or assess therapy response of lung cancer, to evaluate
other pulmonary disease such as chronic obstructive pulmonary disease (COPD) or
cardiovascular disease. Building upon this assumption, and in order to take advan-
tage of the large amounts of hidden data not fully exploited in these chest CTs,
Carneiro et al. developed a radiomics and a deep-learning approach to predict the
5-year all-cause mortality. The following structures were segmented to extract
radiomic features from CT images: lungs, heart, epicardial fat, aorta, spinal column,
body fat, and muscles. The deep learning approach, based on two different types of
CNNs, achieved a mean classification AUC of 69%, outperforming the radiomics
approach, which still presented a good performance (64.6%). Furthermore, the pre-
diction accuracy was similar to currently used clinical risk scores despite the small
cohort of patients enrolled and the exclusion from the models of strongly predictive
variable as gender or age [30].
Among all the lung disease, COPD has received a great deal of interest, as it is
one of the most important cause of death in elder people, and in the next two
decades, it is expected to become the leading cause [31]. Furthermore, COPD is a
known major risk factor for lung cancer, as they share similar physiopathology and
etiology, such as smoking cigarettes [32]. COPD has a heterogeneous CT pattern
resulting from the variable combinations of centrilobular emphysema and airway
disease [33]. This complexity and the ongoing increase of incidence make it neces-
sary to investigate new tools, in order to improve patient’s outcome through an early
diagnosis. The first step was represented by quantitative CT (QCT), which is defined
as the study of computer-aided methods able to quantify handcrafted features on
CT, as the amount of emphysema or airway abnormalities [34]. Though effective,
these methods are often time-consuming, consider only a few handcrafted features,
and are prone to variability [35]; therefore, more dedicated effort need to be done to
explore new hands-on and less time-consuming methods, such as radiomics and
machine learning algorithm. Ginsburg et al. in 2012 extracted texture features to
train a multiple logistic regression classifier able to differentiate effectively between
smokers even without emphysema and never-smokers’ lungs [36]. Lafata et al.
extracted 39 radiomics features to quantify lung function from CT images. The
radiomics signature was then compared to spirometry test as a reference standard,
demonstrating an interesting correlation in particular with FEV1 [37].
Another medical imaging technique progressively more used over the years is
cardiac computed tomography angiography (CCTA), which is extensively per-
formed to rule out coronary artery disease (CAD) [38]. AI may have a potential role
in reducing the time of image analysis, lightening the workload of radiologists,
ranging from diagnostic to prognostic tasks [39]. For instance, Muscogiuri et al.
450 S. C. Fanni et al.
investigated the potential role of a CNN for the classification of CAD-RADS, which
is a standardized method to describe coronary artery disease ranging from 0 (absence
of stenosis) to 5 (at least one totally occluded coronary artery) [40]. The algorithm
effectively distinguishes CAD-RADS ≥1 from 0 with an average time of analysis
much shorter compared to radiologists, which is an important result assuming an
increase of CCTA in the next few years [41]. Another important quantitative param-
eter and predictor for adverse cardiovascular events is the coronary artery calcium
score (CACS) [42]. Wolterink et al. described the implementation of a CNN to
evaluate the CACS on CCTA, achieving an interclass correlation of 0.94 compared
to the reference standard [43].
Additionally, CCTA could be source of parameters to build prognostication
model. Motwani et al. in 2016 developed an ML model for prognostic stratification
in patients followed up for 5 years combining 44 parameters extracted from CCTA
and 25 clinical parameters. The severity score computed by the model achieved an
AUC of 0.79, outperforming traditional prognostic score such as the Framingham
(0.61) and Duke Index (0.62) [44].
17.2.3 Abdomen
Colorectal cancer (CRC) is the third most frequently diagnosed malignancy in both
genders [45]. In spite of the availability of screening programs today, about 60–70%
of CRC are still diagnosed at advanced stages [46].
Therefore, AI could be implemented to improve the diagnostic performance of
routinary screening, such as colonoscopy or computed tomographic colonography
(CTC) [47]. One of the challenges of CTC is the detection of flat colorectal ade-
noma. In 2008 Taylor et al. developed a computer-aided detection system to seek the
flat early-stage CRC on supine and prone CTC images. The algorithm achieved a
sensitivity of 83.3% and 54.1%, respectively, with 0 and 1 sphericity values [48].
Apart from the detection, AI could improve colorectal lesion classification into neo-
plastic and non-neoplastic findings. Song et al. in 2014 demonstrated a significant
improvement in classification of colorectal lesion by adding to image intensity a
texture features analysis, with the AUC rising up from 0.74 to 0.85 [49].
Recently, Grosu et al. implemented an ML method to distinguish more specifi-
cally benign and precancerous lesions detected on CTC of asymptomatic patients,
with an extremely interesting AUC of 0.91 [50].
Another abdominal oncologic condition whose incidence is expected to increase
in the next few years is the hepatocellular carcinoma (HCC). Contrary to CRC,
HCC develops from a specific pathologic substrate, the cirrhotic liver, thus leading
to screening not of the entire population but only in selected patients. Ultrasound
(US) is the main imaging tool to detect new lesion in cirrhotic patients.
To determine the presence of cirrhosis, Liu et al. developed an algorithm based
on the analysis of liver capsule. Using the analysis of liver capsule contour, they
were able to determine the presence or absence of cirrhosis, with an area under the
curve of 0.97 [51]. Once cirrhosis is identified, the challenge is to distinguish benign
from malignant lesions. Bharti et al. proposed an ANN model to differentiate four
17 The Role of Artificial Intelligence (AI) in the Management of Geriatric Patients 451
stages of liver disease using data obtained from US images: normal liver, chronic
liver disease, cirrhosis, and HCC. The classification accuracy of the model was
96.6% [52]. Further, images of contrast-enhanced US were used to develop more
accurate models. Streba et al. implemented an ANN model to classify HCC, liver
metastases, hemangioma, and focal fatty changes with a 94.5% accuracy [53].
When a follow-up ultrasound demonstrates a new lesion suspicious for HCC, other
imaging studies like CT or MRI are required to achieve a characterization. AI could
play a role in the hepatic nodule with an indeterminate behavior which could help
to avoid invasive biopsy. For this purpose, Mokrane et al. retrospectively collect 178
patients with indeterminate nodules subjected to biopsy and developed a DL algo-
rithm to classify nodules as HCC or non-HCC lesion, with an optimistic AUC
of 0.70.
17.2.4 Prostate
Prostate carcinoma represents the most common cancer in men worldwide and,
despite its low aggressivity, is the third cause of death cancer-related [54].
Overdiagnosis is usually a common issue in the diagnostic process of prostatic ade-
nocarcinoma, which leads to an increased burden on the healthcare system. Accurate
prostate segmentation as well as its volume estimation is considered to provide
invaluable information for the process of diagnosis and clinical management of
benign prostatic hyperplasia (BPH) versus the prostatic carcinoma. Therefore, that
can improve BPH treatment, surgical planning, and predictions of PCa prognosis
[55]. There are multiple use case domains of using AI or ML in prostate cancer as
the detection of prostate cancer with high accuracy both in peripheral and transi-
tional zone, characterization of cancer according to its biological aggressiveness
into clinically significant and nonsignificant disease, identification of patients with
metastatic prostate cancer as early as possible, establishing the radiologic–histo-
pathologic correlation to provide biology-based validation of AI models, prediction
of the risk of disease recurrence and prediction of treatment response in case of
radiation therapy or post-prostatectomy as well as the possibility of using
AI-powered patient stratification tools for enrollment in Active Surveillance
programs.
Accuracy of prostate lesion detection, segmentation, and volume estimation is
important at different stages of PCa management. Lesion detection identifies regions
for biopsy. Accurate segmentation is crucial for improved fusion biopsy yields as
well as improving radiotherapy delivery. Volume estimation can predict prognosis
after prostatectomy [56].
The current challenge is the differentiation between aggressive and nonaggres-
sive disease to selectively treat only aggressive type and avoid overdiagnosis and
overtreatment. As prostate cancer is a typical elderly disease, its prevalence and
incidence are expected to dramatically increase exacerbating the challenge already
mentioned. Artificial intelligence could be a solution to achieve the full potential of
multiparametric prostate MRI (mpMRI) as a screening, diagnostic, and prognos-
tic tool.
452 S. C. Fanni et al.
17.2.5 Musculoskeletal
A further type of fracture common in the elderly are the vertebral fractures [64].
When a vertebral fracture is diagnosed, it is challenging to differentiate the benign
from malignant causes, requiring a different pathway of management.
Li et al. trained a CNN with CT images from 433 patients with benign or malig-
nant fracture. The CNN achieved a good result, with an accuracy of 85% [65].
17.3 Conclusion
It is expected that in the upcoming years the old people will increasingly represent
the typical patients accessing healthcare services. Therefore, there is an emerging
need for handling that increased workload on the healthcare systems. With the
advent of many AI algorithms, the potential of AI applications in geriatric patient
management shows a promising spectrum of clinically beneficial uses. The applica-
tions can be employed in diverse practical domains as patients’ screening, imaging
acquisition, pre-reporting, and the further diagnostic process.
References
1. Corselli-Nordblad L, Strandell H, European Commission, Statistical Office of the European
Union. Ageing Europe: looking at the lives of older people in the EU. Brussels: European
Commission; 2020.
2. EUROSTAT. Population structure and ageing. Luxembourg: EUROSTAT; 2020.
3. Bień B, et al. Disabled older people’s use of health and social care services and their unmet
care needs in six European countries. Eur J Pub Health. 2013;23(6):1032–8. https://doi.
org/10.1093/eurpub/cks190.
4. Martin Prince A, et al. World Alzheimer report 2015. The global impact of dementia an
analysis of prevalence, incidence, cost and trends. London: ADI; 2015. www.alz.co.uk/
worldreport2015corrections.
5. Cesario A, et al. The role of artificial intelligence in managing multimorbidity and cancer. J
Personal Med. 2021;11(4):314. https://doi.org/10.3390/jpm11040314.
6. Tinetti ME, Fried TR, Boyd CM. Designing health care for the most common chronic condi-
tion - multimorbidity. JAMA. 2012;307(23):2493–4. https://doi.org/10.1001/jama.2012.5265.
7. Micera S, Bonato P, Tamura T. Gerontechnology. IEEE Eng Med Biol Mag. 2008;27(4):10–4.
https://doi.org/10.1109/MEMB.2008.925213.
8. Nevedal AL, Ayalon L, Briller SH, Heyn PC. A qualitative evidence synthesis review of longi-
tudinal qualitative research in gerontology. Gerontologist. 2019;59(6):E791–801. https://doi.
org/10.1093/geront/gny134.
9. Kok J, Boers EJW, Kosters WA, van der Putten P, Poel M. Artificial intelligence: definition,
trends, techniques and cases. Paris: EOLSS Publishers/UNESCO; 2009.
10. Goldenberg SL, Nir G, Salcudean SE. A new era: artificial intelligence and machine
learning in prostate cancer. Nat Rev Urol. 2019;16(7):391–403. https://doi.org/10.1038/
s41585-019-0193-3.
11. Kohavi R, Provost F. Glossary of terms. Mach Learn. 1998;2:271–4. https://doi.org/10.102
3/A:1017181826899.
12. Brodie A, Dai N, Teoh JYC, Decaestecker K, Dasgupta P, Vasdev N. Artificial intelligence
in urological oncology: an update and future applications. Urol Oncol Semin Orig Investig.
2021;39(7):379–99. https://doi.org/10.1016/j.urolonc.2021.03.012.
454 S. C. Fanni et al.
13. Deng L, Yu D. Deep learning: methods and applications. Found Trends Signal Process.
2013;7(3–4):197–387. https://doi.org/10.1561/2000000039.
14. Javadi-Pashaki N, Ghazanfari MJ, Karkhah S. Machine learning for geriatric clinical care:
opportunities and challenges. Ann Geriatric Med Res. 2021;25(2):137–8. https://doi.
org/10.4235/agmr.21.0054.
15. Ho A. Are we ready for artificial intelligence health monitoring in elder care? BMC Geriatr.
2020;20(1):358. https://doi.org/10.1186/s12877-020-01764-9.
16. Aerts HJWL, et al. Decoding tumour phenotype by noninvasive imaging using a quantitative
radiomics approach. Nat Commun. 2014;5:4006. https://doi.org/10.1038/ncomms5006.
17. Wimo A, Ljunggren G, Winblad B. Costs of dementia and dementia care: a review. Int J Geriatr
Psychiatry. 1997;12:841.
18. Salvatore C, Castiglioni I, Cerasa A. Radiomics approach in the neurodegenerative brain.
Aging Clin Exp Res. 2021;33(6):1709–11. https://doi.org/10.1007/s40520-019-01299-z.
19. Sørensen L, et al. Early detection of Alzheimer’s disease using MRI hippocampal texture.
Hum Brain Mapp. 2016;37(3):1148–61. https://doi.org/10.1002/hbm.23091.
20. Luk CC, et al. Alzheimer’s disease: 3-dimensional MRI texture for prediction of con-
version from mild cognitive impairment. Alzheimer’s Dement Diagn Assess Dis Monit.
2018;10:755–63. https://doi.org/10.1016/j.dadm.2018.09.002.
21. Feng Q, et al. Hippocampus radiomic biomarkers for the diagnosis of amnestic mild cogni-
tive impairment: a machine learning method. Front Aging Neurosci. 2019;11:323. https://doi.
org/10.3389/fnagi.2019.00323.
22. Nanni L, Brahnam S, Salvatore C, Castiglioni I. Texture descriptors and voxels for the early
diagnosis of Alzheimer’s disease. Artif Intell Med. 2019;97:19–26. https://doi.org/10.1016/j.
artmed.2019.05.003.
23. Shinde S, et al. Predictive markers for Parkinson’s disease using deep neural nets on neu-
romelanin sensitive MRI. NeuroImage Clin. 2019;22:101748. https://doi.org/10.1016/j.
nicl.2019.101748.
24. Johnson CO, et al. Global, regional, and national burden of stroke, 1990–2016: a systematic
analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(5):439–58.
https://doi.org/10.1016/S1474-4422(19)30034-1.
25. Chen Q, Xia T, Zhang M, Xia N, Liu J, Yang Y. Radiomics in stroke neuroimaging: tech-
niques, applications, and challenges. Aging Dis. 2021;12(1):143–54. https://doi.org/10.14336/
AD.2020.0421.
26. Musuka TD, Wilton SB, Traboulsi M, Hill MD. Diagnosis and management of acute ischemic
stroke: speed is critical. CMAJ. 2015;187(12):887–93. https://doi.org/10.1503/cmaj.140355.
27. Peter R, et al. A quantitative symmetry-based analysis of hyperacute ischemic stroke lesions
in noncontrast computed tomography. Med Phys. 2017;44(1):192–9. https://doi.org/10.1002/
mp.12015.
28. Kassner A, Liu F, Thornhill RE, Tomlinson G, Mikulis DJ. Prediction of hemorrhagic trans-
formation in acute ischemic stroke using texture analysis of postcontrast T1-weighted MR
images. J Magn Reson Imaging. 2009;30(5):933–41. https://doi.org/10.1002/jmri.21940.
29. Betrouni N, et al. Texture features of magnetic resonance images: an early marker of post-
stroke cognitive impairment. Transl Stroke Res. 2020;11(4):643. https://doi.org/10.1007/
s12975-019-00746-3.
30. Carneiro G, Oakden-Rayner L, Bradley AP, Nascimento J, Palmer L. Automated 5-year mor-
tality prediction using deep learning and radiomics features from chest computed tomography.
In Proceedings of the International Symposium on Biomedical Imaging. Washington, DC:
IEEE; 2017. p. 130–4. https://doi.org/10.1109/ISBI.2017.7950485.
31. Quaderi SA, Hurst JR. The unmet global burden of COPD. Glob Health Epidemiol Genom.
2018;3:e4. https://doi.org/10.1017/gheg.2018.1.
32. Durham AL, Adcock IM. The relationship between COPD and lung cancer. Lung Cancer.
2015;90(2):121–7. https://doi.org/10.1016/j.lungcan.2015.08.017.
33. Washko GR. Diagnostic imaging in COPD. Sem Respir Crit Care Med. 2010;31(3):276–85.
https://doi.org/10.1055/s-0030-1254068.
17 The Role of Artificial Intelligence (AI) in the Management of Geriatric Patients 455
34. Fanni SC, et al. Role of quantitative imaging and deep learning in interstitial lung diseases. J
Radiol Rev. 2021;8(2):152. https://doi.org/10.23736/s2723-9284.21.00127-9.
35. Refaee T, et al. The emerging role of radiomics in COPD and lung cancer. Respiration.
2020;99(2):99–107. https://doi.org/10.1159/000505429.
36. Ginsburg SB, Lynch DA, Bowler RP, Schroeder JD. Automated texture-based quantification
of centrilobular nodularity and centrilobular emphysema in chest CT images. Acad Radiol.
2012;19(10):1241–51. https://doi.org/10.1016/j.acra.2012.04.020.
37. Lafata KJ, Zhou Z, Liu JG, Hong J, Kelsey CR, Yin FF. An exploratory radiomics approach
to quantifying pulmonary function in CT images. Sci Rep. 2019;9(1):11509. https://doi.
org/10.1038/s41598-019-48023-5.
38. Neumann FJ, et al. ESC Guidelines for the diagnosis and management of chronic coronary
syndromes. Eur Heart J. 2020;41(3):407–77. https://doi.org/10.1093/eurheartj/ehz425.
39. van Assen M, Muscogiuri G, Caruso D, Lee SJ, Laghi A, de Cecco CN. Artificial intelli-
gence in cardiac radiology. Radiol Med. 2020;125(11):1186–99. https://doi.org/10.1007/
s11547-020-01277-w.
40. Cury RC, et al. Coronary artery disease - reporting and data system (CAD-RADS): an expert
consensus document of SCCT, ACR and NASCI: endorsed by the ACC. JACC Cardiovasc
Imaging. 2016;9(9):1099.
41. Muscogiuri G, et al. Performance of a deep learning algorithm for the evaluation of CAD-
RADS classification with CCTA. Atherosclerosis. 2020;294:25–32. https://doi.org/10.1016/j.
atherosclerosis.2019.12.001.
42. Greenland P, et al. ACCF/AHA 2007 Clinical Expert Consensus Document on Coronary
artery calcium scoring by computed tomography in global cardiovascular risk assessment and
in evaluation of patients with chest pain. A report of the American College of Cardiology
Foundation Clinical Expert Consensus Task Force (ACCF/AHA Writing Committee to Update
the 2000 Expert Consensus Document on Electron Beam Computed Tomography). J Am Coll
Cardiol. 2007;49(3):378–402. https://doi.org/10.1016/j.jacc.2006.10.001.
43. Wolterink JM, Leiner T, de Vos BD, van Hamersvelt RW, Viergever MA, Išgum I. Automatic
coronary artery calcium scoring in cardiac CT angiography using paired convolutional neural
networks. Med Image Anal. 2016;34:123–36. https://doi.org/10.1016/j.media.2016.04.004.
44. Motwani M, et al. Machine learning for prediction of all-cause mortality in patients with sus-
pected coronary artery disease: a 5-year multicentre prospective registry analysis. Eur Heart
J. 2017;38(7):500–7. https://doi.org/10.1093/eurheartj/ehw188.
45. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics
2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185
countries. CA Cancer J Clin. 2018;68(6):394–424. https://doi.org/10.3322/caac.21492.
46. Mandel JS, et al. Reducing mortality from colorectal cancer by screening for fecal occult
blood. N Engl J Med. 1993;328(19):1365.
47. Mitsala A, Tsalikidis C, Pitiakoudis M, Simopoulos C, Tsaroucha AK. Artificial intel-
ligence in colorectal cancer screening, diagnosis and treatment. A new era. Curr Oncol.
2021;28(3):1581–607. https://doi.org/10.3390/curroncol28030149.
48. Robinson C, et al. CT colonography: computer-assisted detection of colorectal cancer. Br J
Radiol. 2011;84(1001):435–40. https://doi.org/10.1259/bjr/17848340.
49. Song B, et al. Volumetric texture features from higher-order images for diagnosis of colon
lesions via CT colonography. Int J Comput Assist Radiol Surg. 2014;9(6):1021–31. https://doi.
org/10.1007/s11548-014-0991-2.
50. Grosu S, et al. Machine learning-based differentiation of benign and premalignant
colorectal polyps detected with CT colonography in an asymptomatic screening popula-
tion: a proof-of-concept study. Radiology. 2021;299(2):326–35. https://doi.org/10.1148/
RADIOL.2021202363.
51. Liu X, Song J, Hong Wang S, Zhao J, Chen Y. Learning to diagnose cirrhosis with liver cap-
sule guided ultrasound image classification. Sensors. 2017;17(1):149. https://doi.org/10.3390/
s17010149.
456 S. C. Fanni et al.
52. Bharti P, Mittal D, Ananthasivan R. Preliminary study of chronic liver classification on ultra-
sound images using an ensemble model. Ultrason Imaging. 2018;40(6):357–79. https://doi.
org/10.1177/0161734618787447.
53. Streba CT, et al. Contrast-enhanced ultrasonography parameters in neural network diagno-
sis of liver tumors. World J Gastroenterol. 2012;18(32):4427–34. https://doi.org/10.3748/wjg.
v18.i32.4427.
54. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.
https://doi.org/10.3322/caac.21387.
55. Bardis MD, et al. Applications of artificial intelligence to prostate multiparametric MRI
(mpMRI): current and emerging trends. Cancers. 2020;12(5):1204. https://doi.org/10.3390/
cancers12051204.
56. Steenbergen P, et al. Prostate tumor delineation using multiparametric magnetic reso-
nance imaging: inter-observer variability and pathology validation. Radiother Oncol.
2015;115(2):186–90. https://doi.org/10.1016/j.radonc.2015.04.012.
57. Davood K, Ruan D. Synergistic combination of learned and hand-crafted features for prostate
lesion classification in multiparametricmagnetic resonance imaging. In: Descoteaux M, et al.,
editors. Lecture notes in computer science. Cham: Springer; 2017. p. 391–8.
58. Zhong X, et al. Deep transfer learning-based prostate cancer classification using 3 Tesla
multi-parametric MRI. Abdom Radiol. 2019;44(6):2030–9. https://doi.org/10.1007/
s00261-018-1824-5.
59. Li M, et al. Radiomics prediction model for the improved diagnosis of clinically significant
prostate cancer on biparametric MRI. Quantitat Imag Med Surg. 2020;10(2):368–79. https://
doi.org/10.21037/qims.2019.12.06.
60. Rundo L, et al. Automated prostate gland segmentation based on an unsupervised fuzzy
C-means clustering technique using multispectral T1w and T2w MR imaging. Information.
2017;8(2):49. https://doi.org/10.3390/info8020049.
61. Kannus P, Parkkari J, Siev H, Heinonen A, Vuori I. Epidemiology of hip fractures. Bone.
1996;18:57S.
62. Rizzo PF, Gould ES, Lyden JP, Asnis SE. Diagnosis of occult fractures about the hip. J Bone
Joint Surg. 1993;75-A(3):395.
63. Mawatari T, et al. The effect of deep convolutional neural networks on radiologists’ performance
in the detection of hip fractures on digital pelvic radiographs. Eur J Radiol. 2020;130:109188.
https://doi.org/10.1016/j.ejrad.2020.109188.
64. Kendler DL, et al. Vertebral fractures: clinical importance and management. Am J Med.
2016;129(2):221.e1–221.e10. https://doi.org/10.1016/j.amjmed.2015.09.020.
65. Li Y, et al. Differential diagnosis of benign and malignant vertebral fracture on CT using deep
learning. Eur Radiol. 2021;31(12):9612–9. https://doi.org/10.1007/s00330-021-08014-5.
AL GRAWANY