• No results found

University of Groningen Exploring new molecular imaging concepts of prostate cancer Wondergem, Maurits

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Exploring new molecular imaging concepts of prostate cancer Wondergem, Maurits"

Copied!
25
0
0

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

Hele tekst

(1)

University of Groningen

Exploring new molecular imaging concepts of prostate cancer

Wondergem, Maurits

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2017

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Wondergem, M. (2017). Exploring new molecular imaging concepts of prostate cancer. University of

Groningen.

Copyright

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

Take-down policy

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

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

(2)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 9PDF page: 9PDF page: 9PDF page: 9

1

(3)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

(4)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 11PDF page: 11PDF page: 11PDF page: 11

(5)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 12PDF page: 12PDF page: 12PDF page: 12 12

PROSTATE CANCER

In Western and Northern Europe, North America, Australia and New Zealand prostate cancer is the most common cancer in men. A lower incidence is reported in other parts of the world, however in recent years the incidence and mortality rate of prostate cancer in some economically developing Asian countries have grown rapidly (1). Incidence of prostate cancer in the Netherlands doubled in two decades, from 4300 per year in 1990 to approximately 10.000 newly diagnosed patients in 2010 (2). In terms of mortality prostate cancer is the third most lethal cancer in man after lung cancer and colorectal cancer with over 2500 deaths in the Netherlands every year since 2010. Both incidence and mortality rate remained more or less stable since 2010.

Except for its high incidence and mortality rate, prostate cancer is also challenging because of its heterogeneous character. This heterogeneity results in a wide spectrum of clinical presentations ranging from patients that are diagnosed with prostate cancer for which they will never need intervention during their life time to patients with rapidly progressive disease, who die within a year after diagnosis (3, 4). For clinicians it is challenging to apply optimal treatment at the right moment to the right patient. Expected profit of treatment options is highly dependent from the stage of the disease. Different tools, including clinical characteristics, biochemical measurements, pathological examinations and medical imaging, are available to get insight in the stage of the disease and help clinicians’ decision-making.

The main tools to diagnose prostate cancer include digital rectal exam, serum concentration of prostate specific antigen (PSA), and transrectal ultrasound guided biopsy (5-8). As for most other cancers, after diagnosis prostate cancer is staged by the TNM-classification, for which the 7th edition is currently in use (9) (Table 1). The

T-status is used to characterize the primary tumour, which may grow locally within the prostate or may directly invade structures and organs in the direct vicinity. In common clinical practice T-status is determined by one or more tools, including: digital rectal exam, needle biopsy and magnetic resonance imaging (MRI) (10). The N-status indicates presence or absence of lymph node metastases in the true pelvis, which are considered loco-regional metastases. Presence of distant lymph node metastases out-side the true pelvis or other metastases are reflected in the M-status. Clinical T-status, serum PSA levels and Gleason score of the primary tumour are used to estimate the risk on presence of local-regional and distant metastases. Patients with Gleason 7 tumours or PSA between 10-20 ng/ml have intermediate risk prostate cancer. Those with cT3 tumours or Gleason ≥8 tumours or PSA ≥20 ng/ml and those with Gleason 7 tumours and PSA between 10-20 ng/ml are considered high risk. All others are considered low risk. High risk may indicate the necessity for further diagnostic tests, including imaging modalities such as Magnetic Resonance Imaging (MRI), Computed Tomography (CT), different nuclear medicine imaging techniques including planar scintigraphy or Single Photon Emission Computed Tomography (SPECT) or Positron Emission Tomography (PET) and hybrid imaging modalities which combine SPECT or PET with CT or MRI (11-16).

(6)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 13PDF page: 13PDF page: 13PDF page: 13

1

13

Also pelvic lymphadenectomy may be performed in intermediate and high-risk patients and this surgical approach combines diagnosis and treatment of pelvic lymph node metastases (10).

Table 1. American Joint Committee on Cancer Prostate Cancer Staging, TNM 7th Edition

Primary Tumour (T) Clinical

TX Primary tumour cannot be assessed T0 No evidence of primary tumour

T1 Clinically inapparent tumour neither palpable nor visible by imaging

T1a Tumour incidental histologic finding in 5% or less of prostate tissue resected (for other

reasons)

T1b Tumour incidental histologic finding in more than 5% of prostate tissue resected T1c Tumour identified by needle biopsy (for example, because of elevated PSA) T2 Tumour confined within prostate

T2a Tumour involves one-half of one-lobe or less

T2b Tumour involves more than one-half of one lobe but not both lobes T2c Tumour involves both lobes

T3 Tumour extends through the prostate capsule

T3a Extracapsular extension (unilateral or bilateral) T3b Tumour invades seminal vesicle(s)

T4 Tumour is fixed or invades adjacent structures other than seminal vesicles, such as external

sphincter, rectum, bladder, levator muscles, and/or pelvic wall

Regional Lymph Nodes (N) Clinical

NX Regional lymph nodes were not assessed N0 No regional lymph node metastasis N1 Metastasis in regional lymph node(s) Distant Metastasis (M)

MX Distant metastases were not assessed M0 No distant metastasis

M1 Distant metastasis

M1a Non regional lymph node(s) M1b Bone(s)

M1c Other site(s) with or without bone disease

Detection and localisation of metastases is an important aspect in guidance of prostate cancer treatment at different stages of the disease. The presence of advanced metastasis at clinical presentation will withhold the patient from strategies with curative intent, while local, loco-regional or oligometastatic disease may be targeted with aggressive therapies with curative potential. Although most patients diagnosed with prostate cancer in economically developed countries have clinically localised prostate cancer and are treated with treatments with curative intent such as radical prostatectomy or external beam radiation therapy, many of those patients will eventually have biochemical recurrence (10). At this stage imaging modalities, especially PET/CT with different tracers including 11C-choline, 18F-fluorocholine or PSMA targeted tracers, are used in present

clinical practice to indicate the extent of disease. In case of localised, locoregional or limited distant metastases salvage therapeutic options with curative intent, such as salvage radiation therapy or salvage lymphadenectomy, may be indicated (10, 17).

(7)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 14PDF page: 14PDF page: 14PDF page: 14

14

Table 2. Overview of radiopharmaca studied in this thesis. Chemical name, Molecular formula,

2D-structure and 3D-model

Tracers of bone remodelling Tracers of tumour cells

3D-model legend. White: Hydrogen, black: Carbon, blue: Nitrogen, red: Oxygen, green: Fluorine, purple: Sodium, orange: Phosphorus and dark cyan blue: Technetium.

Subsequently most of them will progress and receive androgen deprivation therapy in a palliative setting (17). Serum PSA and imaging modalities, most commonly bone scintigraphy with 99mTc-phosphonates (BS) or PET/CT with 18F-sodiumfluoride (NaF) are

(8)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 15PDF page: 15PDF page: 15PDF page: 15

1

15

castrate levels of testosterone, most patients will progress to develop castrate resistant prostate cancer. At this stage, different systemic agents such as docetaxel, abiraterone, enzalutamide and carbazitaxel and the alpha-emitting radioisotope 223Ra may be used

to prolong life expectancy and treat symptoms of which bone pain is most prevalent

(18-21). Other symptomatic treatments include radioactive bone tracers as 188Re-HEDP, 153Sm-EDTMP or 89Sr-strontiumchloride and external radiation therapy. At this stage

serum PSA and imaging, most commonly imaging of bone metastases, are used for treatment planning and follow-up (22, 23). Progression of disease may result in a switch between available systemic agents or 223Ra.

Early metastatic disease most commonly comprises haematogenous spread to the skeleton and/or lymphogenic spread to lymph nodes. Therefore diagnosis of prostate cancer metastases is most frequently targeted at detection of bone and/or lymph node metastases (24, 25). In more advanced disease metastases in lungs, liver, pleura and adrenals may be encountered and in rare occasions metastases are found in the brain, breasts, eyes, kidneys, muscles, pancreas, salivary glands and spleen.

This thesis is aimed at nuclear medicine techniques that are useful in detection of prostate cancer lesions. Amongst others those techniques include BS, NaF PET/CT,

11C-choline or 18F-fluorocholine PET/CT and PET/CT with tracers that target the prostate

specific membrane antigen (PSMA) which are commonly labelled with 68Ga or 18F

(Table 2).

NUCLEAR MEDICINE IMAGING

In short, nuclear medicine uses non-invasive imaging techniques that can visualize the distribution of radioactive tracers within a living body. A tracer is a molecule or ligand that can be measured and which resembles a molecule that is biologically active in physiological or pathological processes. The administered dosage of a tracer is very low and interference with biological processes in the body does not occur. In nuclear medicine tracers for imaging are labelled with isotopes that either emit gamma-rays (γ-rays) or positrons (β+). Gamma-rays can be measured by an Anger gamma camera,

which provides planar images or SPECT images (26). Positrons are emitted from the nucleus of a radioactive isotope with a certain velocity; first they will lose all their kinetic energy by interactions with their surroundings after which the positron annihilates with a nearby electron. At annihilation the mass of both the positron and electron are transformed into two gamma photons of 511 keV, which will be emitted at almost 180 degrees of each other (27). Those photons may be measured using Anger gamma cameras, however PET-cameras are normally used since those cameras can localise the positron source along a straight line of coincidence. Moreover, the crystals used in these cameras are more appropriate for detection of photons with relatively high energies such as 511 keV (27). The use of PET cameras results in higher photon detection yield,

(9)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 16PDF page: 16PDF page: 16PDF page: 16 16

which in combination with better localisation of the source of the emitted photons, results in higher quality images as compared to planar scintigraphy or SPECT. Hybrid imaging, which synergises the information of distribution of biologically active tracers from PET or SPECT with anatomical information of CT, has become standard of care for many disorders in oncology, cardiology, orthopaedics and neurology in the past decade (28, 29).

For prostate cancer different nuclear imaging techniques and tracers can be used to assess the extent and localisation of the disease. Depending on the course of the disease and the estimated risk on lymph node or bone metastases, different tracers are used. Some of them only show the secondary process of bone remodelling due to metastatic infiltration in the bone matrix while others show the localisation of tumour cells themselves.

Imaging bone remodelling

From a historical point of view it makes sense to discuss tracers that enable imaging of bone matrix remodelling first. Already in the early 1920s it was recognised that certain radionuclides had the property to accumulate in bone. It was demonstrated that ingested salts by painters of luminous watch dials were concentrated in the bone. Metabolic studies of a wide variety of radionuclides were commenced in the 1930s, including 32P orthophosphate, 18F, 45Ca and 89Sr and 87mSr. Clinical studies indicated

that bone imaging with 89Sr or 87mSr was capable of detecting bone lesions well before

radiological changes became apparent (30-32). Better results were found with 18F in

the 1960s, due to faster blood clearance and improved skeletal visualisation (33). As a result of the ideal imaging properties of 99mTc for imaging with an Anger gamma-camera,

which at that time was widely used for other nuclear medicine procedures, and the known effect of phosphonates in the treatment of bone disorders, the search for a bone-seeking tracer was centred on a 99mTc-labelled phosphate complex agent. Many

bisphosphonates have undergone extensive evaluation and comparison, but because of more rapid blood clearance and higher skeletal affinity 99mTc-methylene diphosphonate

(99mTc-MDP) and later 99mTc-hydroxymethylene diphosphonate (99mTc-HDP) became the

most widely used agents in routine clinical bone scintigraphy (34).

The wide availability and technical improvements of PET/CT cameras after the introduction of 2-deoxy-2-(18F)fluoro-D-glucose (FDG) in standard medical imaging from

the late 1980s and the increased knowledge of the only moderate accuracy of 99mTc-MDP

and 99mTc-HDP bone scans resulted in renewed interest in 18F, which is a positron emitter,

as a bone tracer in medical imaging. Improvement of sensitivity was expected due to better image quality of PET techniques including higher spatial resolution, and favourable pharmacokinetics of 18F with higher target-to-non-target ratios. Implementation of

hybrid PET/CT cameras, which enable better anatomical identification of lesions and as a result better discrimination between benign and malignant processes, were expected to improve specificity, but did not reach the market before the turn of the millennium.

(10)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 17PDF page: 17PDF page: 17PDF page: 17

1

17

99mTc-diphosphonates

For decades 99mTc-MDP and 99mTc-HDP have been the most commonly used tracers for

bone imaging (35). Those tracers consist of 99mTc-diphosphonates complexes (Table 2),

which are administered intravenously. A substantial degree of those complexes binds to plasma-proteins, increasing in time from around 25% immediately after injection to 50% at 1 hour (36-38). This results in relatively slow bloodpool clearance, slow accumulation in the bone matrix and slow renal excretion. Four hours after injection about 50-60% of the injected amount is accumulated and bound in the bone matrix of the skeleton, 34% is excreted in the urine and only 6% remains in the circulation. Delayed images for detection of bone metastases are commonly acquired from 2 up to 5 hours after intravenous injection, which fits with the half-life of 6.02h of 99mTc emitting one 140.5

keV gamma ray per decay (35). Typically administered activities usually range between 8 – 10 MBq/kg.

Skeletal uptake of 99mTc-MDP and 99mTc-HDP depends on local blood flow and osteoblastic

activity but does not reflect the true tumour burden in the bone marrow. Prostate cancer cells in the bone matrix induce a complex biochemically interaction with osteoblasts and osteoclasts resulting in remodelling of the bone matrix and the formation of new hydroxyapatite (39-41). Increased osteoblastic activity results in increased accumulation of 99mTc-MDP and 99mTc-HDP in the vicinity of prostate cancer cells. The nature of the

actual complex that reacts with bone and the mechanism of uptake into the bone mineral remain unclear. It is postulated that 99mTc-biphosphonate complexes adhere

to the bone mineral surface of newly formed hydroxyapatite intact or that they may fall apart to allow separate binding of the diphosphonate component and the reduced pertechnetate (42).

Physiologic uptake of 99mTc-MDP and 99mTc-HDP is seen in osseous structures, kidney

and bladder. A wide spectrum of both malignant and benign lesions may be diagnosed by increased uptake of the tracer. Also soft tissue uptake is seen in a variety of non-osseous disorders including neoplastic, hormonal, inflammatory, ischemic and traumatic entities (43).

18F-sodiumfluoride

NaF is administered intravenously as a solution of the saline sodium fluoride (Na+ 18F-) (Table 2) and only the 18F- ion is the tracer of bone remodelling. However in the

continuation of this thesis the tracer will be addressed as NaF, as is common in recent medical literature. Rapidly around 30% of the injected NaF is found in erythrocytes due to free diffusion across membranes (44). Almost all NaF is retained in the bone after a single pass of blood (45). The initial distribution of NaF therefore represents blood flow, which varies among different bones (46). NaF is rapidly cleared from the plasma and harboured within bone or excreted by the kidneys. Only 10% of the injected amount is found in the plasma one hour after injection (47). As a result image acquisition of

(11)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 18PDF page: 18PDF page: 18PDF page: 18 18

the axial skeleton may start about 30-45 minutes after administration, which is ideal concerning the 110 minutes half-life of 18F (48). The administered activity for adults

commonly ranges between 185-370 MBq.

The mechanism of skeletal uptake of NaF is based on ion exchange, probably in the same manner as 99mTc-MDP and 99mTc-HDP is incorporated in the bone matrix. 18F- ions

exchange with hydroxyl ions (OH-) on the surface of newly formed hydroxyapatite by

osteoblasts (47, 49, 50). Concentrations of NaF can be up to 10 times higher in areas of bone remodelling, compared with areas of normal bone (33).

Physiologic NaF uptake resembles uptake of 99mTc-MDP and 99mTc-HDP and is found in

osseous structures, kidney and bladder. Normal physiologic osseous uptake in adults is generally uniform. Mechanisms leading to increased uptake are not limited to neoplastic processes and include any process of bone remodelling (51). As a consequence NaF can visualise the same variety of osseous diseases as 99mTc-MDP and 99mTc-HDP. Non-osseous

uptake of NaF can be observed in the arterial vasculature, gastrointestinal tract and genitourinary tract. Vascular activity is mainly due to atherosclerotic calcification (52,

53). Lesions containing calcifications such as calcified visceral metastases may show NaF

uptake (54). Occasionally NaF activity is seen in the bowel. The exact uptake mechanism is unclear, however protein losing enteropathy has been suggested as a cause of bowel activity (55).

Imaging tumour cells

Different cellular pathways or characteristics may be upregulated in prostate cancer cells and serve as a target for diagnostic imaging. At present 18F-fluorodeoxyglucose (FDG) is

most commonly used in oncology. The use of glucose is increased in a large number of malignancies as a result of an increased metabolic rate of the tumour cells (56, 57). However the role of FDG for detection of primary disease and metastases in prostate cancer is limited, due to relatively low avidity of prostate tumours for FDG resulting in low sensitivity and due to overlap of FDG uptake intensity with benign entities including benign prostatic hypertrophy and prostatitis (58-60). As a consequence tracers that target other cellular pathways, cellular receptors or transmembrane proteins have been developed, including lipid metabolism, amino acid metabolism, androgen receptor, prostate specific membrane antigen inhibitors/antibodies, cellular proliferation, gastrin releasing peptide receptor expression and gene-mediated imaging (Table 3) (61-71). At present the majority of those tracers have no or limited clinical application and are beyond the scope of this thesis. 11C-choline, 18F-fluorocholine and tracers that target

the prostate specific membrane antigen are becoming integrated in standard clinical practice, and are further introduced in the next paragraphs.

(12)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 19PDF page: 19PDF page: 19PDF page: 19

1

19 Table 3. Potential PET tracers for prostate cancer

PET tracer Mechanism

11C-choline, 18F-fluorocholine, 11C-acetate or

18F-fluoroacetate Lipid metabolism 18F-FACBC, 11C-methionine Amino acid transport

18F-FDHT Androgen receptor

18F-DCFPyL, 18F-DCFBC, 68Ga-PSMA-compounds:

for example 68Ga-PSMA-HBED-CC PSMA inhibitors/antibodies 18F-FLT, 18F-FMAU Cellular proliferation

18F-NaF Bone remodeling

68Ga-bombesin analogues Gastrin releasing peptide receptor expression 18F-FACBC: anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid

18F-FDHT: 16β-18F-fluoro-5α-dihydrotestosterone

18F-DCFPyL: 2-(3-(1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl)-ureido)-pentanedioic

acid

18F-DCFBC: N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-4-18F-fluorobenzyl-L-cysteine 68Ga-PSMA-HBED-CC: Glu-NH-CO-NH-Lys-(Ahx)-[68Ga(HBED-CC)]

18F-FLT: 18F-3’-deoxy-3’-fluorothymidine

18F-FMAU: 18F-20-fluoro-5-methyl-1-b-D-arabinofuranosyluracil 18F-NaF: 18F-sodiumfluoride

11C-choline and 18F-fluorocholine

In the 1970s research had been directed to the development of low molecular weight tracers for imaging of biochemically active substrates that play an active role in the biosynthesis of cell constituents, such as proteins, nucleic acids, and lipid derivatives. Choline, a small molecule involved in many cellular functions, was identified as a substrate of interest. Choline homeostasis is altered in different malignancies, probably as a result of increased proliferation of membrane lipids in actively replicating tumours. Increased choline kinase activity and increased choline concentration was found in brain tumours and later on also in other tumours such as lung, colorectal and prostate tumours (72-78).

Radiolabelled choline was first synthesised as 14C-choline in the early 1980s, however

due to unfavourable emission properties, including the 14C half-life of approximately

5730 years, 14C-choline was unsuitable for medical imaging (79). As a result 11C-choline

PET/CT was designed and synthesised for imaging of brain tumours and a few years later 18F labelled choline tracers were synthesised, which allowed widespread use

of choline tracers, also in medical centres without access to a cyclotron facility (Table 2) (80-82).

In prostate cancer choline cellular uptake is a result of both an active transporter-mediated and a passive diffusion-like component (83). The definite mechanism leading to intracellular accumulation and trapping of choline is still unknown. Whether tracer uptake is related to increased synthesis of membrane phosphocholine in tumour cells with high turnover or to cellular proliferation has been investigated, which yielded inconclusive results (84-87).

(13)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 20PDF page: 20PDF page: 20PDF page: 20 20

The biodistribution of 11C- choline and 18F-fluorocholine show great resemblance and

both show a rapid blood clearance and rapid uptake in prostate cancer cells (88). Physiological accumulation is seen in lacrimal glands, salivary glands, liver, spleen, pancreas, small intestine, colon, muscle and bone marrow (89, 90). A larger portion of 18F-flurocholine, as compared to 11C-choline, is excreted in the urine, which may

interfere with detection of the primary tumour (81, 82). Increased choline uptake is seen in different malignancies including hepatocellular cancer, lung cancer, breast cancer, lymphomas, brain tumours, melanoma, colon cancer, bladder cancer and multiple myeloma (91). Also benign processes may show increased choline uptake such as a wide variety of infections and inflammations as well as meningioma, benign thyroid nodule, parathyroid adenoma, thymoma, adrenal adenoma, Morbus Paget and posttraumatic bone injury (89, 91, 92).

Prostate specific membrane antigen targeted tracers

Lack of specificity of conventional imaging techniques for prostate cancer has encouraged screening of prostate cancer cells for possible antigens to develop agents capable of specific binding. This resulted in the development of monoclonal antibodies (mAbs) to target PSA and prostatic acid phosphatase (PAP) (93). However, secretion of those antigens preclude cell-associated binding and presence of PSA and PAP in the plasma effectively blocks specific antibody binding at the tumour site. Later PSMA was discovered, which is a 750 amino acid transmembrane protein and a highly specific prostate epithelial cell membrane antigen (94-96). Physiological expression of PSMA is 100-1000 fold less than typical expression in prostate cancer, and expression increases as tumour grade increases with concurrent increase in metastatic sites and castrate refractory prostate cancer (CRPC) (97, 98). Furthermore, PSMA is internalised and endosomally recycled, which increases the deposition of these radiopharmaceuticals into the cell over time (99).

In 2006 111In-capromab, a mAb for targeting PSMA, was reported. However this tracer

has a poor efficacy associated with binding to the intracellular domain of PSMA, which results only in binding to nonviable cells that have damaged cell membranes (100). A few years later mAbs targeting the extracellular domain of PSMA were reported. Due to their relatively large mass these ligands show slow clearance from background and slow target recognition, prohibiting their success as radiopharmaceuticals for imaging, since these are preferably administered and subsequently used for imaging on the same day. Furthermore radiopharmaceuticals require superior safety profiles. mAbs have potential side effects including allergic reactions, which is another disadvantage

(101-103). From the late 2000s small molecule PSMA inhibitors, which are approximately 350

fold smaller than mAbs, have been reported (104-108). Those tracers have rapid target recognition and background clearance and no adverse effects have been reported. Most of these tracers targeting PSMA are glutamate-urea-lysine analogues (109).

(14)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 21PDF page: 21PDF page: 21PDF page: 21

1

21

PSMA-tracer kinetics show that, after intravenous injection, the tracer accumulates in prostate cancer cells over time while background activity decreases (110-114). Physiological accumulation of PSMA tracers occurs in lacrimal glands, salivary glands, liver, gall bladder, spleen, small intestine, colon, paravertebral ganglia and coeliac ganglia. Besides prostate cancer increased PSMA expression is demonstrated by other malignant diseases including renal cell carcinoma, adenoid cystic carcinoma of the salivary gland, differentiated thyroid cancer, urothelial cancer, lung cancer, colon cancer, oesophageal cancer and brain tumours (115-120). In some of those malignancies PSMA may be expressed directly at the tumour cell membranes, however in most cases PSMA expression in found in the neovasculature of the tumour. Increased accumulation of PSMA may also be seen in benign entities such as degenerative bone diseases, bone fractures, Morbus Paget, Schwannoma, haemangioma, adrenal adenomas, benign thyroid nodules, sub-acute brain infarction and pleural fluid (121-126).

At the moment, a large variety of PSMA tracers are available for clinical use, most of which are labelled with 68Ga. However there is an increasing interest in 18F labelled

PSMA-tracers (Table 2) as a result of favourable physical and imaging characteristics. Positrons emitted by 18F decay have lower kinetic energies as compared to those

emitted by 68Ga, which results in higher resolution PET images acquired using 18F-tracers.

Furthermore the 110-minute half-life of 18F compared to 68 minutes for 68Ga enables

imaging at later time points without significant deterioration of image quality or the need for administration of higher dosages.

NUCLEAR MEDICINE IMAGING IN PROSTATE CANCER ACCORDING TO PRESENT

GUIDELINES

Primary staging

According to present guidelines use of nuclear medicine imaging is, at this stage, only recommended for detection of bone metastases and only in cases with significant risk on bone metastases. BS, which has been applied for decades, is indicated at initial staging in a selection of patients with a high pre-test likelihood of having bone metastases. Those include patients with PSA ≥ 20 ng/ml, clinical stage 3 tumour (T3) or Gleason score ≥ 8 according to the Dutch and European guidelines (10, 127). The Dutch guidelines state that NaF, 11C-choline or 18F-choline PET/CT may substitute BS according to

local availability.

Biochemical relapse after treatment with curative intent

The Dutch guidelines state that 11C-choline PET/CT or 18F-fluorocholine PET/CT can be

used in this patient category provided that there is a reasonable chance on a positive scan which is indicated by a PSA > 5.0 ng/ml or a PSA > 1.0 ng/ml together with a PSA doubling time < 3 months or a Gleason score ≥ 8. 99mTc-phosphonate BS for detection of

(15)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 22PDF page: 22PDF page: 22PDF page: 22 22

bone metastases may be indicated in case of a biochemical relapse if the PSA ≥ 20 ng/ ml or in case of bone pain. BS may be substituted by NaF, 11C-choline or 18F-choline PET/

CT according to local availability (127).

The European guidelines state that imaging such as 11C-choline PET/CT should only be

performed if therapeutic consequences such as salvage lymphadenectomy or salvage RT to lymph nodes are being considered as a therapeutic option. A PSA doubling time less than 3 months can certainly be regarded as a strong predictor of PET positivity as does a PSA serum level >1.5 ng/ml (17).

Follow-up of metastasised disease

Both Dutch and European guidelines give no recommendations on the use of imaging at this stage of the disease.

AIM AND OUTLINE OF THE THESIS

As described above a lot of new tracers for use in nuclear medicine have been developed in the last decades and some of them found their way into standard clinical practice. Once available for use in patients, insights in clinical aspects including diagnostic accuracy, optimal patient preparation and optimal acquisition parameters, such as acquisition timing after tracer administration, are needed before tracers can be incorporated in standard of care. This thesis aims to elucidate some of these aspects for tracers that recently became available. In the first chapters the scope is mainly fixed at detection of bone metastases, while the remaining chapters are focussed at tracers that enable detection of both bone and soft tissue lesions.

In chapter 2 contemporary literature on the diagnostic accuracy of NaF, 11C-choline and 18F-flurocholine PET/CT for detection of bone metastases in prostate cancer patients is

reviewed systematically using the Oxford Criteria. Available data is pooled and pooled sensitivities and specificities for 11C-choline and 18F-fluorocholine together and NaF are

calculated on a per patient and a per lesion basis.

In chapter 3 the diagnostic accuracy and clinical impact of BS with 99mTc-HDP and NaF

PET/CT is studied in an observational cohort study. Both cohorts consisted of patients primarily staged for high-risk prostate cancer. Additionally, for NaF PET/CT the added value and its impact on management of CT findings, especially detection of enlarged lymph nodes, are analysed.

In chapter 4 the scope is switched to 18F-fluorocholine. The kinetics of 18F-fluorocholine

in malignant as well as normal tissues are studied in order to identify imaging characteristics that differentiate malignant from normal lymph nodes and malignant bone lesions from normal bone marrow uptake. The findings are used to optimise timing of acquisition after tracer administration.

(16)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 23PDF page: 23PDF page: 23PDF page: 23

1

23

In chapter 5 the impact of fasting for at least 6 hours prior to intravenous 18F-fluorocholine

administration on gastrointestinal uptake and detection of lymph node metastases is investigated.

In chapter 6 we studied whether early dynamic imaging of the prostatic region with

18F-fluorocholine PET/CT increased the detection rate of local prostate cancer recurrence

in patients with a biochemical relapse after radical prostatectomy.

In chapter 7 the scope is switched again to another tracer; the PSMA targeting tracer

18F-DCFPyL. Effects of activity kinetics on the detection rate of prostate cancer lesions,

image quality and biodistribution are studied at 60 and 120 minutes after intravenous administration of the tracer.

In intermezzo 1 a case is reported with high 18F-DCFPyL uptake in bilateral adrenal

adenomas in a patient with rising PSA-serum values while on Luteinizing Hormone-Releasing Hormone agonist therapy.

Future perspectives and conclusions are described in chapter 8.

(17)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 24PDF page: 24PDF page: 24PDF page: 24 24

REFERENCES

1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359-86.

2. www.cijfersoverkanker.nl Accessed February 17, 2015

3. Johansson JE, Adami HO, Andersson SO, Bergstrom R, Krusemo UB, Kraaz W. Natural history of localised prostatic cancer. A population-based study in 223 untreated patients. Lancet. 1989;1:799-803.

4. Whitmore WF,Jr. Natural history and staging of prostate cancer. Urol Clin North Am. 1984;11: 205-220.

5. Carvalhal GF, Smith DS, Mager DE, Ramos C, Catalona WJ. Digital rectal examination for detecting prostate cancer at prostate specific antigen levels of 4 ng./ml. or less. J Urol. 1999;161:835-839. 6. Thompson IM, Pauler DK, Goodman PJ, et al. Prevalence of prostate cancer among men with

a prostate-specific antigen level < or =4.0 ng per milliliter. N Engl J Med. 2004;350:2239-2246. 7. Hara R, Jo Y, Fujii T, et al. Optimal approach for prostate cancer detection as initial biopsy:

Prospective randomized study comparing transperineal versus transrectal systematic 12-core biopsy. Urology. 2008;71:191-195.

8. Takenaka A, Hara R, Ishimura T, et al. A prospective randomized comparison of diagnostic efficacy between transperineal and transrectal 12-core prostate biopsy. Prostate Cancer Prostatic Dis. 2008;11:134-138.

9. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer V. 1. 2015. Available at http://www.nccn.org/professionals/physician_gls/pdf/ prostate.pdf. Accessed: May 4, 2017.

10. Heidenreich A, Bastian PJ, Bellmunt J, et al. EAU guidelines on prostate cancer. part 1: Screening, diagnosis, and local treatment with curative intent-update 2013. Eur Urol. 2014;65:124-137. 11. Abuzallouf S, Dayes I, Lukka H. Baseline staging of newly diagnosed prostate cancer: A

summary of the literature. J Urol. 2004;171:2122-2127.

12. Dickinson L, Ahmed HU, Allen C, et al. Magnetic resonance imaging for the detection, localisation, and characterisation of prostate cancer: Recommendations from a european consensus meeting. Eur Urol. 2011;59:477-494.

13. Rorvik J, Halvorsen OJ, Albrektsen G, Haukaas S. Lymphangiography combined with biopsy and computer tomography to detect lymph node metastases in localized prostate cancer. Scand J Urol Nephrol. 1998;32:116-119.

14. Van Poppel H, Ameye F, Oyen R, Van de Voorde W, Baert L. Accuracy of combined computerized tomography and fine needle aspiration cytology in lymph node staging of localized prostatic carcinoma. J Urol. 1994;151:1310-1314.

15. Even-Sapir E, Metser U, Mishani E, Lievshitz G, Lerman H, Leibovitch I. The detection of bone metastases in patients with high-risk prostate cancer: 99mTc-MDP planar bone scintigraphy, single- and multi-field-of-view SPECT, 18F-fluoride PET, and 18F-fluoride PET/CT. J Nucl Med. 2006;47:287-297.

(18)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 25PDF page: 25PDF page: 25PDF page: 25

1

25 16. Herlemann A, Wenter V, Kretschmer A, et al. 68Ga-PSMA positron emission tomography/ computed tomography provides accurate staging of lymph node regions prior to lymph node dissection in patients with prostate cancer. Eur Urol. 2016;70:553-557.

17. Heidenreich A, Bastian PJ, Bellmunt J, et al. EAU guidelines on prostate cancer. part II: Treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol. 2014;65:467-479. 18. Parker C, Nilsson S, Heinrich D, et al. Alpha emitter radium-223 and survival in metastatic

prostate cancer. N Engl J Med. 2013;369:213-223.

19. Poorthuis MH, Vernooij RW, van Moorselaar RJ, de Reijke TM. First-line non-cytotoxic therapy in chemotherapy-naive patients with metastatic castration-resistant prostate cancer: A systematic review of 10 randomised clinical trials. BJU Int. 2017.

20. Kearns B, Pandor A, Stevenson M, et al. Cabazitaxel for hormone-relapsed metastatic prostate cancer previously treated with a docetaxel-containing regimen: An evidence review group perspective of a NICE single technology appraisal. Pharmacoeconomics. 2017;35:415-424. 21. Hurwitz M. Chemotherapy in prostate cancer. Curr Oncol Rep. 2015;17:44-015-0468-7.

22. Etchebehere EC, Araujo JC, Milton DR, et al. Skeletal tumor burden on baseline 18F-fluoride PET/CT predicts bone marrow failure after 223Ra therapy. Clin Nucl Med. 2016;41:268-273. 23. Kairemo K, Joensuu T. Radium-223-dichloride in castration resistant metastatic prostate

cancer-preliminary results of the response evaluation using F-18-fluoride PET/CT. Diagnostics (Basel). 2015;5:413-427.

24. Krishnamurthy GT, Tubis M, Hiss J, Blahd WH. Distribution pattern of metastatic bone disease. A need for total body skeletal image. JAMA. 1977;237:2504-2506.

25. Bubendorf L, Schopfer A, Wagner U, et al. Metastatic patterns of prostate cancer: An autopsy study of 1,589 patients. Hum Pathol. 2000;31:578-583.

26. Anger HO. Scintillation camera with multichannel collimators. J Nucl Med. 1964;5:515-531. 27. Phelps ME, Hoffman EJ, Mullani NA, Ter-Pogossian MM. Application of annihilation coincidence

detection to transaxial reconstruction tomography. J Nucl Med. 1975;16:210-224.

28. Akhurst T, Chisin R. Hybrid PET/CT machines: Optimized PET machines for the new millennium? J Nucl Med. 2000;41:961-963.

29. Delbeke D, Sandler MP. The role of hybrid cameras in oncology. Semin Nucl Med. 2000;30: 268-280.

30. Fleming WH, McIlraith JD, King ER. Photoscanning of bone lesions utilizing strontium 85. Radiology. 1961;77:635-636.

31. Charkes ND. Some differences between bone scans made with 87mSr and 85Sr. J Nucl Med. 1969;10:491-494.

32. Myers WG. The first radioindicator study in the life sciences with a man-made radionuclide: “Radioactive indicators in the study of phosphorus metabolism in rats, by O. chievitz and G. hevesy, reprinted from nature 136: 754-755, nov. 9, 1935.”. J Nucl Med. 1975;16:1106-1108. 33. Blau M, Nagler W, Bender MA. Fluorine-18: A new isotope for bone scanning. J Nucl Med.

(19)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 26PDF page: 26PDF page: 26PDF page: 26 26

34. Subramanian G, McAfee JG, Blair RJ, Kallfelz FA, Thomas FD. Technetium-99m-methylene diphosphonate--a superior agent for skeletal imaging: Comparison with other technetium complexes. J Nucl Med. 1975;16:744-755.

35. Van den Wyngaert T, Strobel K, Kampen WU, et al. The EANM practice guidelines for bone scintigraphy. Eur J Nucl Med Mol Imaging. 2016;43:1723-1738.

36. Hyldstrup L, McNair P, Ring P, Henriksen O. Studies on diphosphonate kinetics. part I: Evaluation of plasma elimination curves during 24 h. Eur J Nucl Med. 1987;12:581-584.

37. Blake GM, Moore AE, Park-Holohan SJ, Fogelman I. A direct in vivo measurement of 99mTc-methylene diphosphonate protein binding. Nucl Med Commun. 2003;24:829-835.

38. Moore AE, Hain SF, Blake GM, Fogelman I. Validation of ultrafiltration as a method of measuring free 99mTc-MDP. J Nucl Med. 2003;44:891-897.

39. Guise TA, Mohammad KS, Clines G, et al. Basic mechanisms responsible for osteolytic and osteoblastic bone metastases. Clin Cancer Res. 2006;12:6213s-6216s.

40. Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: A fatal attraction. Nat Rev Cancer. 2011;11:411-425.

41. Ibrahim T, Flamini E, Mercatali L, Sacanna E, Serra P, Amadori D. Pathogenesis of osteoblastic bone metastases from prostate cancer. Cancer. 2010;116:1406-1418.

42. Kanishi D. 99mTc-MDP accumulation mechanisms in bone. Oral Surg Oral Med Oral Pathol. 1993;75:239-246.

43. Peller PJ, Ho VB, Kransdorf MJ. Extraosseous tc-99m MDP uptake: A pathophysiologic approach. Radiographics. 1993;13:715-734.

44. Carlson CH, Armstong WD, Singer L. Distribution, migration and binding of whole blood fluoride evaluated with radiofluoride. Am J Physiol. 1960;199:187-189.

45. Wootton R, Dore C. The single-passage extraction of 18F in rabbit bone. Clin Phys Physiol Meas. 1986;7:333-343.

46. Frost ML, Blake GM, Cook GJ, Marsden PK, Fogelman I. Differences in regional bone perfusion and turnover between lumbar spine and distal humerus: (18)F-fluoride PET study of treatment-naive and treated postmenopausal women. Bone. 2009;45:942-948.

47. Weber DA, Greenberg EJ, Dimich A, et al. Kinetics of radionuclides used for bone studies. J Nucl Med. 1969;10:8-17.

48. Beheshti M, Mottaghy FM, Payche F, et al. (18)F-NaF PET/CT: EANM procedure guidelines for bone imaging. Eur J Nucl Med Mol Imaging. 2015;42:1767-1777.

49. Blau M, Ganatra R, Bender MA. 18 F-fluoride for bone imaging. Semin Nucl Med. 1972;2:31-37. 50. Czernin J, Satyamurthy N, Schiepers C. Molecular mechanisms of bone 18F-NaF deposition. J

Nucl Med. 2010;51:1826-1829.

51. Segall G, Delbeke D, Stabin MG, et al. SNM practice guideline for sodium 18F-fluoride PET/CT bone scans 1.0. J Nucl Med. 2010;51:1813-1820.

(20)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 27PDF page: 27PDF page: 27PDF page: 27

1

27 52. Derlin T, Toth Z, Papp L, et al. Correlation of inflammation assessed by 18F-FDG PET, active

mineral deposition assessed by 18F-fluoride PET, and vascular calcification in atherosclerotic plaque: A dual-tracer PET/CT study. J Nucl Med. 2011;52:1020-1027.

53. Dweck MR, Joshi FR, Newby DE, Rudd JH. Noninvasive imaging in cardiovascular therapy: The promise of coronary arterial (1)(8)F-sodium fluoride uptake as a marker of plaque biology. Expert Rev Cardiovasc Ther. 2012;10:1075-1077.

54. Grant FD, Fahey FH, Packard AB, Davis RT, Alavi A, Treves ST. Skeletal PET with 18F-fluoride: Applying new technology to an old tracer. J Nucl Med. 2008;49:68-78.

55. Zuckier LS, Freeman LM. Nonosseous, nonurologic uptake on bone scintigraphy: Atlas and analysis. Semin Nucl Med. 2010;40:242-256.

56. Woods M, Hunter J, Burk D. Regulation of glucose utilization in tumors by a stress-modified insulin: Anti-insulin system. J Natl Cancer Inst. 1955;16:351-404.

57. Weber G. Enzymology of cancer cells (second of two parts). N Engl J Med. 1977;296:541-551. 58. Hofer C, Laubenbacher C, Block T, Breul J, Hartung R, Schwaiger M.

Fluorine-18-fluorodeoxyglucose positron emission tomography is useless for the detection of local recurrence after radical prostatectomy. Eur Urol. 1999;36:31-35.

59. Liu IJ, Zafar MB, Lai YH, Segall GM, Terris MK. Fluorodeoxyglucose positron emission tomography studies in diagnosis and staging of clinically organ-confined prostate cancer. Urology. 2001;57:108-111.

60. Morris MJ, Akhurst T, Osman I, et al. Fluorinated deoxyglucose positron emission tomography imaging in progressive metastatic prostate cancer. Urology. 2002;59:913-918.

61. Jadvar H. Molecular imaging of prostate cancer: PET radiotracers. AJR Am J Roentgenol. 2012;199:278-291.

62. Picchio M, Messa C, Landoni C, et al. Value of [11C]choline-positron emission tomography for re-staging prostate cancer: A comparison with [18F]fluorodeoxyglucose-positron emission tomography. J Urol. 2003;169:1337-1340.

63. Mena E, Turkbey B, Mani H, et al. 11C-acetate PET/CT in localized prostate cancer: A study with MRI and histopathologic correlation. J Nucl Med. 2012;53:538-545.

64. Jambor I, Borra R, Kemppainen J, et al. Functional imaging of localized prostate cancer aggressiveness using 11C-acetate PET/CT and 1H-MR spectroscopy. J Nucl Med. 2010;51: 1676-1683.

65. Ren J, Yuan L, Wen G, Yang J. The value of anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid PET/CT in the diagnosis of recurrent prostate carcinoma: A meta-analysis. Acta Radiol. 2016;57:487-493.

66. Larson SM, Morris M, Gunther I, et al. Tumor localization of 16beta-18F-fluoro-5alpha-dihydrotestosterone versus 18F-FDG in patients with progressive, metastatic prostate cancer. J Nucl Med. 2004;45:366-373.

67. Szabo Z, Mena E, Rowe SP, et al. Initial evaluation of [(18)F]DCFPyL for prostate-specific membrane antigen (PSMA)-targeted PET imaging of prostate cancer. Mol Imaging Biol. 2015;17:565-574.

(21)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 28PDF page: 28PDF page: 28PDF page: 28 28

68. Afshar-Oromieh A, Haberkorn U, Eder M, Eisenhut M, Zechmann CM. 68Ga]gallium-labelled PSMA ligand as superior PET tracer for the diagnosis of prostate cancer: Comparison with 18F-FECH. Eur J Nucl Med Mol Imaging. 2012;39:1085-1086.

69. Oyama N, Hasegawa Y, Kiyono Y, et al. Early response assessment in prostate carcinoma by (1)(8)F-fluorothymidine following anticancer therapy with docetaxel using preclinical tumour models. Eur J Nucl Med Mol Imaging. 2011;38:81-89.

70. Sun H, Sloan A, Mangner TJ, et al. Imaging DNA synthesis with [18F]FMAU and positron emission tomography in patients with cancer. Eur J Nucl Med Mol Imaging. 2005;32:15-22.

71. Kahkonen E, Jambor I, Kemppainen J, et al. In vivo imaging of prostate cancer using [68Ga]-labeled bombesin analog BAY86-7548. Clin Cancer Res. 2013;19:5434-5443.

72. Ramirez de Molina A, Rodriguez-Gonzalez A, Gutierrez R, et al. Overexpression of choline kinase is a frequent feature in human tumor-derived cell lines and in lung, prostate, and colorectal human cancers. Biochem Biophys Res Commun. 2002;296:580-583.

73. Ackerstaff E, Pflug BR, Nelson JB, Bhujwalla ZM. Detection of increased choline compounds with proton nuclear magnetic resonance spectroscopy subsequent to malignant transformation of human prostatic epithelial cells. Cancer Res. 2001;61:3599-3603.

74. Heerschap A, Jager GJ, van der Graaf M, et al. In vivo proton MR spectroscopy reveals altered metabolite content in malignant prostate tissue. Anticancer Res. 1997;17:1455-1460.

75. Shimizu H, Kumabe T, Tominaga T, et al. Noninvasive evaluation of malignancy of brain tumors with proton MR spectroscopy. AJNR Am J Neuroradiol. 1996;17:737-747.

76. Yavin E. Regulation of phospholipid metabolism in differentiating cells from rat brain cerebral hemispheres in culture: Ontogenesis of carrier-specific transport of choline and N-methyl-substituted choline analogs. J Neurochem. 1980;34:178-183.

77. Lanks K, Somers L, Papirmeister B, Yamamura H. Choline transport by neuroblastoma cells in tissue culture. Nature. 1974;252:476-478.

78. George TP, Morash SC, Cook HW, Byers DM, Palmer FB, Spence MW. Phosphatidylcholine biosynthesis in cultured glioma cells: Evidence for channeling of intermediates. Biochim Biophys Acta. 1989;1004:283-291.

79. Friedland RP, Mathis CA, Budinger TF, Moyer BR, Rosen M. Labeled choline and phosphorylcholine: Body distribution and brain autoradiography: Concise communication. J Nucl Med. 1983;24:812-815.

80. Hara T, Kosaka N, Shinoura N, Kondo T. PET imaging of brain tumor with [methyl-11C]choline. J Nucl Med. 1997;38:842-847.

81. DeGrado TR, Coleman RE, Wang S, et al. Synthesis and evaluation of 18F-labeled choline as an oncologic tracer for positron emission tomography: Initial findings in prostate cancer. Cancer Res. 2001;61:110-117.

82. Hara T, Kosaka N, Kishi H. Development of (18)F-fluoroethylcholine for cancer imaging with PET: Synthesis, biochemistry, and prostate cancer imaging. J Nucl Med. 2002;43:187-199.

(22)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 29PDF page: 29PDF page: 29PDF page: 29

1

29 83. Muller SA, Holzapfel K, Seidl C, Treiber U, Krause BJ, Senekowitsch-Schmidtke R. Characterization of choline uptake in prostate cancer cells following bicalutamide and docetaxel treatment. Eur J Nucl Med Mol Imaging. 2009;36:1434-1442.

84. Hara T, Kosaka N, Kishi H. PET imaging of prostate cancer using carbon-11-choline. J Nucl Med. 1998;39:990-995.

85. Al-Saeedi F, Smith T, Welch A. Methyl-3H]-choline incorporation into MCF-7 cells: Correlation with proliferation, choline kinase and phospholipase D assay. Anticancer Res. 2007;27:901-906. 86. Al-Saeedi F, Welch AE, Smith TA. Methyl-3H]choline incorporation into MCF7 tumour cells:

Correlation with proliferation. Eur J Nucl Med Mol Imaging. 2005;32:660-667.

87. Breeuwsma AJ, Pruim J, Jongen MM, et al. In vivo uptake of [11C]choline does not correlate with cell proliferation in human prostate cancer. Eur J Nucl Med Mol Imaging. 2005;32:668-673. 88. Sutinen E, Nurmi M, Roivainen A, et al. Kinetics of [(11)C]choline uptake in prostate cancer: A

PET study. Eur J Nucl Med Mol Imaging. 2004;31:317-324.

89. Schillaci O, Calabria F, Tavolozza M, et al. 18F-choline PET/CT physiological distribution and pitfalls in image interpretation: Experience in 80 patients with prostate cancer. Nucl Med Commun. 2010;31:39-45.

90. Haroon A, Zanoni L, Celli M, et al. Multicenter study evaluating extraprostatic uptake of 11C-choline, 18F-methylcholine, and 18F-ethylcholine in male patients: Physiological distribution, statistical differences, imaging pearls, and normal variants. Nucl Med Commun. 2015;36:1065-1075.

91. Calabria F, Chiaravalloti A, Schillaci O. (18)F-choline PET/CT pitfalls in image interpretation: An update on 300 examined patients with prostate cancer. Clin Nucl Med. 2014;39:122-130. 92. Michaud L, Balogova S, Burgess A, et al. A pilot comparison of 18F-fluorocholine PET/CT,

ultrasonography and 123I/99mTc-sestaMIBI dual-phase dual-isotope scintigraphy in the preoperative localization of hyperfunctioning parathyroid glands in primary or secondary hyperparathyroidism: Influence of thyroid anomalies. Medicine (Baltimore). 2015;94:e1701. 93. Ghosh A, Heston WD. Tumor target prostate specific membrane antigen (PSMA) and its

regulation in prostate cancer. J Cell Biochem. 2004;91:528-539.

94. Rajasekaran AK, Anilkumar G, Christiansen JJ. Is prostate-specific membrane antigen a multifunctional protein? Am J Physiol Cell Physiol. 2005;288:C975-81.

95. Pinto JT, Suffoletto BP, Berzin TM, et al. Prostate-specific membrane antigen: A novel folate hydrolase in human prostatic carcinoma cells. Clin Cancer Res. 1996;2:1445-1451.

96. Israeli RS, Powell CT, Corr JG, Fair WR, Heston WD. Expression of the prostate-specific membrane antigen. Cancer Res. 1994;54:1807-1811.

97. Laidler P, Dulinska J, Lekka M, Lekki J. Expression of prostate specific membrane antigen in androgen-independent prostate cancer cell line PC-3. Arch Biochem Biophys. 2005;435:1-14. 98. Sokoloff RL, Norton KC, Gasior CL, Marker KM, Grauer LS. A dual-monoclonal sandwich assay

for prostate-specific membrane antigen: Levels in tissues, seminal fluid and urine. Prostate. 2000;43:150-157.

(23)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 30PDF page: 30PDF page: 30PDF page: 30 30

99. Rajasekaran SA, Anilkumar G, Oshima E, et al. A novel cytoplasmic tail MXXXL motif mediates the internalization of prostate-specific membrane antigen. Mol Biol Cell. 2003;14:4835-4845. 100. Elsasser-Beile U, Wolf P, Gierschner D, Buhler P, Schultze-Seemann W, Wetterauer U. A new

generation of monoclonal and recombinant antibodies against cell-adherent prostate specific membrane antigen for diagnostic and therapeutic targeting of prostate cancer. Prostate. 2006;66:1359-1370.

101. Tagawa ST, Beltran H, Vallabhajosula S, et al. Anti-prostate-specific membrane antigen-based radioimmunotherapy for prostate cancer. Cancer. 2010;116:1075-1083.

102. Wolf P, Freudenberg N, Buhler P, et al. Three conformational antibodies specific for different PSMA epitopes are promising diagnostic and therapeutic tools for prostate cancer. Prostate. 2010;70:562-569.

103. Regino CA, Wong KJ, Milenic DE, et al. Preclinical evaluation of a monoclonal antibody (3C6) specific for prostate-specific membrane antigen. Curr Radiopharm. 2009;2:9-17.

104. Foss CA, Mease RC, Fan H, et al. Radiolabeled small-molecule ligands for prostate-specific membrane antigen: In vivo imaging in experimental models of prostate cancer. Clin Cancer Res. 2005;11:4022-4028.

105. Mease RC, Dusich CL, Foss CA, et al. N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-4-[18F] fluorobenzyl-L-cysteine, [18F]DCFBC: A new imaging probe for prostate cancer. Clin Cancer Res. 2008;14:3036-3043.

106. Chen Y, Pullambhatla M, Foss CA, et al. 2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pen tanedioic acid, [18F]DCFPyL, a PSMA-based PET imaging agent for prostate cancer. Clin Cancer Res. 2011;17:7645-7653.

107. Afshar-Oromieh A, Zechmann CM, Malcher A, et al. Comparison of PET imaging with a (68)ga-labelled PSMA ligand and (18)F-choline-based PET/CT for the diagnosis of recurrent prostate cancer. Eur J Nucl Med Mol Imaging. 2014;41:11-20.

108. Eder M, Neels O, Muller M, et al. Novel preclinical and radiopharmaceutical aspects of [68Ga] ga-PSMA-HBED-CC: A new PET tracer for imaging of prostate cancer. Pharmaceuticals (Basel). 2014;7:779-796.

109. Hillier SM, Maresca KP, Femia FJ, et al. Preclinical evaluation of novel glutamate-urea-lysine analogues that target prostate-specific membrane antigen as molecular imaging pharmaceuticals for prostate cancer. Cancer Res. 2009;69:6932-6940.

110. Szabo Z, Mena E, Rowe SP, et al. Initial evaluation of [(18)F]DCFPyL for prostate-specific membrane antigen (PSMA)-targeted PET imaging of prostate cancer. Mol Imaging Biol. 2015;17:565-574.

111. Afshar-Oromieh A, Malcher A, Eder M, et al. PET imaging with a [68Ga]gallium-labelled PSMA ligand for the diagnosis of prostate cancer: Biodistribution in humans and first evaluation of tumour lesions. Eur J Nucl Med Mol Imaging. 2013;40:486-495.

112. Herrmann K, Bluemel C, Weineisen M, et al. Biodistribution and radiation dosimetry for a probe targeting prostate-specific membrane antigen for imaging and therapy. J Nucl Med. 2015;56:855-861.

(24)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 31PDF page: 31PDF page: 31PDF page: 31

1

31 113. Afshar-Oromieh A, Hetzheim H, Kratochwil C, et al. The theranostic PSMA ligand PSMA-617 in

the diagnosis of prostate cancer by PET/CT: Biodistribution in humans, radiation dosimetry, and first evaluation of tumor lesions. J Nucl Med. 2015;56:1697-1705.

114. Afshar-Oromieh A, Hetzheim H, Kubler W, et al. Radiation dosimetry of (68)ga-PSMA-11 (HBED-CC) and preliminary evaluation of optimal imaging timing. Eur J Nucl Med Mol Imaging. 2016;43:1611-1620.

115. Demirci E, Ocak M, Kabasakal L, et al. (68)ga-PSMA PET/CT imaging of metastatic clear cell renal cell carcinoma. Eur J Nucl Med Mol Imaging. 2014;41:1461-1462.

116. Rowe SP, Gorin MA, Hammers HJ, et al. Imaging of metastatic clear cell renal cell carcinoma with PSMA-targeted (1)(8)F-DCFPyL PET/CT. Ann Nucl Med. 2015;29:877-882.

117. Gupta M, Choudhury PS, Gupta G, Gandhi J. Metastasis in urothelial carcinoma mimicking prostate cancer metastasis in ga-68 prostate-specific membrane antigen positron emission tomography-computed tomography in a case of synchronous malignancy. Indian J Nucl Med. 2016;31:222-224.

118. Shetty D, Loh H, Bui C, Mansberg R, Stevanovic A. Elevated 68Ga prostate-specific membrane antigen activity in metastatic non-small cell lung cancer. Clin Nucl Med. 2016;41:414-416. 119. Verburg FA, Krohn T, Heinzel A, Mottaghy FM, Behrendt FF. First evidence of PSMA expression

in differentiated thyroid cancer using [(6)(8)ga]PSMA-HBED-CC PET/CT. Eur J Nucl Med Mol Imaging. 2015;42:1622-1623.

120. Lutje S, Sauerwein W, Lauenstein T, Bockisch A, Poeppel TD. In vivo visualization of prostate-specific membrane antigen in adenoid cystic carcinoma of the salivary gland. Clin Nucl Med. 2016;41:476-477.

121. Demirci E, Sahin OE, Ocak M, Akovali B, Nematyazar J, Kabasakal L. Normal distribution pattern and physiological variants of 68Ga-PSMA-11 PET/CT imaging. Nucl Med Commun. 2016;37:1169-1179.

122. Gykiere P, Goethals L, Everaert H. Healing sacral fracture masquerading as metastatic bone disease on a 68Ga-PSMA PET/CT. Clin Nucl Med. 2016;41:e346-7.

123. Rowe SP, Deville C, Paller C, et al. Uptake of 18F-DCFPyL in paget’s disease of bone, an important potential pitfall in clinical interpretation of PSMA PET studies. Tomography. 2015;1:81-84. 124. Krohn T, Verburg FA, Pufe T, et al. (68)ga]PSMA-HBED uptake mimicking lymph node metastasis

in coeliac ganglia: An important pitfall in clinical practice. Eur J Nucl Med Mol Imaging. 2015;42:210-214.

125. Rischpler C, Maurer T, Schwaiger M, Eiber M. Intense PSMA-expression using (68)ga-PSMA PET/ CT in a paravertebral schwannoma mimicking prostate cancer metastasis. Eur J Nucl Med Mol Imaging. 2016;43:193-194.

126. Noto B, Vrachimis A, Schafers M, Stegger L, Rahbar K. Subacute stroke mimicking cerebral metastasis in 68Ga-PSMA-HBED-CC PET/CT. Clin Nucl Med. 2016;41:e449-51.

127. Integraal Kankercentrum Nederland. Richtlijn prostaat carcinoom version 2.1. 2014. Available at: http://www.oncoline.nl/prostaatcarcinoom, accessed May 11, 2017. 

(25)

514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem 514137-L-bw-Wondergem Processed on: 27-9-2017 Processed on: 27-9-2017 Processed on: 27-9-2017

Processed on: 27-9-2017 PDF page: 32PDF page: 32PDF page: 32PDF page: 32

Referenties

GERELATEERDE DOCUMENTEN

For both cohorts the number of positive, negative and equivocal findings, calculated diagnostic performance of the imaging modality in terms of sensitivity and specificity, as well

On the static images, a significant higher 18 F-fluorocholine uptake was measured in malignant lymph nodes compared to normal lymph nodes, adipose tissue and muscle

Intestinal activity that possibly interferes with the detection of lymph node metastases is encountered in all prostate cancer patients receiving 18 F-fluorocholine

The number of patients for which a histological biopsy was present in this study is unclear and, to our opinion, a decreasing PSA below 0.1 ng/ml after salvage radiation therapy

A recent case report showed faint accumulation of 68 Ga PSMA HBED-CC in an adrenal adenoma (8), but there are no reports of adrenal masses with high uptake of PSMA ligands...

Although, especially according to the standards for reporting diagnostic accuracy studies (STARD 2015), the currently available evidence for use of PSMA targeted tracers instead

Additionally a second cohort, which included patients that received 18 F-fluorocholine PET/CT for primary staging, was studied to determine whether increased tracer uptake on

Op laesie niveau werd een gewogen gepoolde sensitiviteit en specificiteit van respectievelijk 84% en 98% gevonden voor PET/CT met op choline-gebaseerde radiofarmaca en 89% en 91%