Vol 11, No 4 (2015)
Guidelines / Expert consensus
Published online: 2015-08-27

open access

Page views 900
Article views/downloads 5766
Get Citation

Connect on Social Media

Connect on Social Media

_01_OiCP_2015_4__Krzakowski

EXPERTS` OPINION

Maciej Krzakowski1, Piotr Rutkowski1, Jacek Jassem2, Renata Zaucha2, Jacek Fijuth3,Janusz Słuszniak4, Barbara Jarząb5, Wojciech Zegarski6, Bogdan Małkowski6,Andrzej Kawecki1, Witold Rzyman2, Andrea D’Amico5, Andrzej Stelmach7,Wojciech Polkowski8, Radosław Mądry9, Leszek Królicki10

1Centrum Onkologii — Instytut im. Marii Skłodowskiej-Curie w Warszawie

2Gdański Uniwersytet Medyczny

3Uniwersytet Medyczny w Łodzi

4Świętokrzyskie Centrum Onkologii w Kielcach

5Centrum Onkologii — Instytut im. Marii Skłodowskiej-Curie, Oddział w Gliwicach

6Centrum Onkologii im. Prof. F. Łukaszczyka w Bydgoszczy

7Centrum Onkologii — Instytut im. Marii Skłodowskiej-Curie, Oddział w Krakowie

8Uniwersytet Medyczny w Lublinie

9Uniwersytet Medyczny w Poznaniu

10Warszawski Uniwersytet Medyczny

Recommendations on the application of positron emission tomography in oncology

Under the auspices of the national consultants in clinical oncology, nuclear medicine and Polish Society of Oncology, Polish Society of Surgical Oncology, Polish Society of Clinical Oncology and Polish Society of the Nuclear Medicine

ABSTRACT

Positron emission tomography (PET) is a modern functional imaging method with a proven value in diagnosing, staging, evaluating responses to anticancer therapy and detecting relapses in numerous neoplasms. The utility, sensitivity and specificity of PET has increased by its use in combination with computed tomography (CT) or magnetic resonance in the form of fusion PET-CT or PET-MR and the introduction of new radiotracers. This paper, based on scientific evidence by a multidisciplinary group of authors, presents the utility and clinical recommendations for the application of PET-CT in oncology.

PET-CT is particularly useful for:

  • appropriate diagnosis and initial staging of patients with head and neck, lung, pancreatic and esophageal cancers, as well as lymphomas, advanced melanomas and tumors of unknown primary site;
  • detection of relapses in patients with colorectal, thyroid, ovarian, head and neck, and breast cancers, as well as lymphomas;
  • monitoring of response to treatment in patients with testicular and lung cancers, lymphomas and some types of sarcomas.

PET-MR is particulary useful in pediatrics.

Key words: positron emission tomography, oncology, diagnosis, staging, response to treatment, recommendations

Oncol Clin Pract 2015; 11, 4: 155171

Introduction

Positron emission tomography (PET) is a modern, functional imaging method used to evaluate many biological processes. The introduction of hybrid techniques such as a combination of PET with computed tomography (CT) PET-CT imaging and with the magnetic resonance imaging (MRI) PET-MRI imaging has increased the applicability and sensitivity of PET. The simultaneous application of CT or MRI and PET with the subsequent fusion of both images allows for the correction of the phenomenon of the absorption of the radiation emitted by the radioisotope combined with a radiotracer and the precise positioning of the site of abnormal radiotracer accumulation. Another factor influencing the importance of PET imaging is the introduction of novel radiotracers, allowing one to evaluate a range of phenotypic changes characteristic for different neoplastic processes. 18F-phosphatydyloclucose (18F-FDG) is still a basic radiotracer in PET imaging. PET imaging with the use of this radiotracer allows the evaluation of glucose metabolic processes, as well as those responsible for the Warburg phenomenon. An increase in 18F-FDG intake is observed not only in malignant neoplasms but also in many inflammatory changes (e.g. tuberculosis, ulcers, fungal infections, sarcoidosis). Despite the limitation mentioned above, this method has a confirmed clinical value in establishing the diagnosis, in the initial cancer staging, in the assessment of the response to anticancer therapy and in the relapse detection in the course of many cancers.

However, the introduction of the PET-MRI method has not changed the indications for performing PET-CT imaging. PET-MRI should be considered mostly in children, especially when repeated PET imaging will be necessary to control treatment results. PET-MTI imaging permits an important reduction of absorbed radiation dose and consequently decreases the risk of secondary neoplasms induced by ionizing radiation. PET-MRI has also been proven to be more effective in diagnosing central nervous system (CNS) neoplasms, breast cancer, head and neck and pelvic neoplasms.

The aim of this paper is to analyse the utility and to define clinical recommendations to use PET-CT imaging in oncology. The authors have considered and present both in descriptive form and in the tables (Table 1) current, detailed clinical indications for using PET-CT imaging. The list of indications that should be reimbursed from public payment sources has also been proposed. The recommendations have been worked out based on evidence-based medicine principles (EBM). Three sets of indications are specified, namely those clinically unequivocally confirmed and absolutely useful in clinical practice (category A), probable and potentially useful (category B) and individually accessed indications (category C).

Table 1. Summary of the recommendations for PET-CT imaging in oncology

Recommendation

Initial PET-CT in order to define treatment strategy (making diagnosis and/or staging of disease)

Monitoring and treatment results evaluation, re-staging after the anticancer treatment, relapse of disease detection, further PET-CT imaging in disease follow-up

Non-small cell lung cancer

Yes

Staging before planned radical treatment A

(except from patients with neuroendocrine cancer and patients with distant metastases)

Yes

Staging of residual disease after anticancer therapy A

Differentiation of the changes in case of the suspicion of disease relapse A

Small-cell lung cancer

Yes

Staging before radical treatment of limited disease A

No

Malignant pleural mesothelioma

Yes

Staging before radical combined treatment A

No

Single lung tumour of unknown character

Yes

Differentiation of benign and malignant tumours smaller than 5 mm if histopathological testing cannot be performed A

No

Neoplasms in the mediastinum (thymus, heart, mediastinum)

Yes

Staging before radical surgical or combined treatment B

No

Lymphomas

Yes

Staging before treatment A

Yes

Final evaluation of treatment efficacy, re-staging of disease during post-treatment follow-up, staging of relapse A

Evaluation of early response to treatment B

Radiotherapy field planning C

Multiple myeloma

Yes

Initial disease staging B

Detection of extra-skeletal changes C

Yes

Detection of disease relapse B

Colorectal cancer

Yes

To define possibility of surgical treatment in patients with potentially resectable metastases A

Staging in pre-operative evaluation in patients with rectal carcinoma and suspicion on distant metastases B

Yes

Confirmation or exclusion of disease relapse in cases of rising CEA levels or of unequivocal results of imaging test A

Oesophageal cancer

Yes

Staging at qualification for primary radical treatment B

Yes

Detection of distant metastases in cases of local relapse post radical treatment C

Table 1. Summary of the recommendations for PET-CT imaging in oncology

Pancreatic neoplasms

Yes

Differentiation of changes in pancreas in initial diagnostics B

Staging of pancreatic cancer C

No

Liver and intrahepatic biliary ducts neoplasms

Yes

Defining character of focal change in liver (only with use of acetate tracer) C*

Staging of hepatocellular cancer (only with use of carbon isotope-labelled acetate) B*

Yes

Defining staging after treatment and at relapse (with use of fluorine-labelled choline analogues, carbon-labelled acetates) B*

Gastric cancer

Yes

Pre-operative staging in patients with ambiguous results of radiological exams (only with use of 18FLT) B*

No

Gall bladder and extrahepatic biliary ducts neoplasms

Yes

Staging prior to making therapeutic decisions in cases when all other imaging exams do not supply exact diagnosis B

No

Breast cancer

Yes

Exclusion of distant metastases in locally-advanced disease if results of standard imaging tests are unequivocal B

Yes

Confirmation or exclusion of relapse of disease after radical treatment if results of other imaging exams are ambiguous and PET-CT may change treatment method B

Evaluation of response to induction chemotherapy C

Evaluation of results of chemotherapy applied due to disease dissemination C

Neoplasms of the uterus

Yes

To define possibility of radical treatment in locally advanced cervical cancer and endometrial cancer if results of other imaging exams suggest metastatic changes (especially in lymph nodes) B

Yes

Confirmation or exclusion of disease relapse in patients treated radically A

Ovarian and ovary duct neoplasms

Yes

To define disease extension in patients with potentially resectable metastases A

Yes

Confirmation or exclusion of disease relapse in patients with elevated Ca125 levels and unequivocal results of imaging exams A

Prostate cancer (PET-CT performed with labelled choline, acetate, carbon and PSMA)

Yes

Diagnosing of changes in cases of high PSA levels indicating presence of malignant neoplasm and negative results of repeated biopsies B

Disease staging before radical treatment when there is a suspicion of metastases to lymph nodes and to bones and when results of other imaging exams are ambiguous A

Yes

Confirmation or exclusion of relapse in the cases of rising PSA levels and negative results of all other imaging exams A

Table 1. Summary of the recommendations for PET-CT imaging in oncology

Testicular cancer

Yes

Neoplasm staging (with exception of mature teratomas) A

Yes

Detection and evaluation of residual changes extension, detection of relapse B

Evaluation of response to treatment B

Urinary bladder cancer

Yes

Staging of disease in patients with potentially resectable metastases B

Yes

Detection of relapse of disease after radical treatment if results of other imaging exams are unequivocal and PET-CT result may change treatment strategy B

Melanomas

Yes

Evaluation of disease extension in patients with clinical metastases to regional lymph nodes (stage IIIBC) A

Evaluation of neoplasm extension in patients with potentially resectable metastases to distant organs (stage IV) A

Neoplasms diagnosis in patients with metastasis with unknown primary site A

No

Merkel cell cancer

Yes

Staging before treatment A

Yes

Confirmation or exclusion of neoplasm relapse A

Connective tissue/other soft tissues neoplasms (including gastrointestinal stromal tumour)

Yes

Differentiation of benign and malignant changes B

Planning of tumour biopsy C

Initial staging C

Yes

Evaluation of disease response to treatment and detection of relapse on treatment in patients with gastrointestinal stromal tumours A

Evaluation of response to treatment in other sarcomas (especially in small cell sarcomas) B

Neoplasm of bone/cartilage

Yes

Differentiating of benign and malignant changes B

Yes

Detection of relapse and distant metastases A

Head and neck neoplasms

Yes

Detection of primary site of the neoplasm in patients with metastases in cervical lymph nodes from unknown primary site A

Evaluation of primary site condition and of changes in the lymph nodes in loco-regionally advanced neoplasms B

Radiotherapy planning in case of loco-regionally advance disease B

Yes

Detection of local or regional relapse when results of other imaging exams are ambiguous A

Neoplasms of central nervous system (imaging examination indicated with the use of fluorine labelled tyrosine,carbon-labelled methionine or of fluorine-labelled DOPA)

Yes

Planning of stereotactic biopsy in the case of suspicion of central nervous system neoplasm B*

Detection of regions of higher histological grade within central nervous system neoplasms B*

Yes

Detection of neoplasm relapse C*

Table 1. Summary of the recommendations for PET-CT imaging in oncology

Thyroid cancer

No

Yes

Differentiation of changes in course of mature thyroid cancer after total thyroidectomy in cases of elevated thyroglobulin levels ( 10 ng/ml) and negative result of the 131I scintigraphy A

Other neuroendocrine neoplasms

Yes

Evaluation of somatostatin receptors expression B*

Evaluation of biologic malignancy of primary and metastatic sites B*

No

Neoplasms of unknown primary site

Yes

Evaluation of primary site if results of other imaging exams are negative or unequivocal (value of exam depends on metastases localization) A

No

Radical radiotherapy

Yes

If results of another exams cannot be used in radiotherapy planning A

No

*Radiotracer other than FDG; CEA carcinoembryonic antigen; PSA prostate specific antigen; FDG fluorodeoxyglucose; PSMA prostate-specific membrane antigen

Recommendations for performing PET-CT imaging have been divided based on the EBM rules with the specification of the categories listed below regarding utility in clinical practice:

A utility equivocally confirmed; B potential utility; C utility in individual indications.

The proposed categories presents the recommendation to perform the PET-CT imaging. In all other clinical situations the value of the PET-CT imaging was not proven or there are no EBM based data evaluating it utility.

In the majority of cases, the recommendations are based on the use of fusion PET-CT with 18F-FDG while some marked separately refer to the use of other radiotracers.

The authors of this paper are specialists in different fields of medicine (included: clinical oncologists, radiotherapists, surgeons and nuclear medicine specialists).

Lung cancer

Lung cancer is one of the main indications for the use of PET-CT imaging in oncology. PET-CT imaging is applied to diagnose and to access the stage of the disease prior and post anticancer treatment. The majority of clinical trials testing PET-CT applicability refer to the non-small cell lung cancer (NSCLC). There is an increasing clinical evidence for the utility of PET-CT also in the diagnostic process of small cell lung cancer (SCLC). Present data prove the cost effectiveness of PET-CT imaging in lung cancer.

However, PET-CT imaging is not recommended as a screening tool for the early detection of lung cancer.

PET-CT imaging with 18F-FDG is not recommended in NSCLC patients with previously detected metastasis. In the neuroendocrine neoplasms the aim of PET-CT imaging is to assess the biological malignancy of the tumour/tumours (the increased uptake 18F-FDG within the tumour indicates more aggressive disease and helps to choose the appropriate treatment method).

A negative result of the PET-CT of the mediastinal lymph nodes imaging proves limited disease. A positive result of PET-CT in mediastinal lymph nodes may be related to an inflammatory process and demands a further invasive diagnostics (ultrasound controlled trans-bronchial or trans-oesophageal biopsy, mediastinoscopy or thoracoscopy).

Initial diagnostics and pre-treatment staging

The pre-treatment indications for PET-CT diagnostic include:

  • staging in patients with the diagnosis of NSCLC who are candidates for radical surgery or radiotherapy;
  • characterization of an suspected pulmonary nodules of diameter exceeding 5 mm (nodules of diameter below 5 mm may be an indication for PET-CT imaging only when recommended by a multidisciplinary team with the participation of a nuclear medicine specialist);
  • radiotherapy planning in selected NSCLC patients (e.g. extensive atelectasis of the lung) who are candidates for radical radio-chemotherapy or radiotherapy;
  • initial staging in a selected group of patients with limited SCLC being candidates for radio-chemotherapy.

Post-treatment follow-up

Post-treatment PET-CT imaging is indicated in patients only if CT indicates the possibility of relapse (to differentiate between treatment side-effects and neoplasm relapse).

Mesothelioma

PET-CT imaging is a clinically useful method in the initial diagnosis of patients with suspicion of mesothelioma.

The indications include:

  • selection of the most suitable site for a the effective biopsy in patients with suspicion of mesothelioma (this concerns patients with pleural thickening; PET-CT imaging is not useful in those patients who present only pleural effusion or who underwent pleurodesis);
  • exclusion of neoplastic sites beyond the thorax in patients being candidates for combined treatment including surgery (pleurectomy/decortication/pleuro-pneumonectomy).

PET-CT imaging is not indicated during monitoring after the anticancer treatment in patients with mesothelioma.

Thymus neoplasms

PET-CT imaging with the use of 18F-FDG is a valuable method in the diagnostics of thymus cancer. The value of PET-CT in remaining thymus neoplasms has not been proven. Due to the persistent risk of thymus cancer, the PET-CT imaging is recommended before the surgical treatment of the thymus tumour.

PET-CT imaging has no value in monitoring patients post-thymus neoplasm treatment.

Lymphoma

Precise initial disease staging is crucial for selecting an optimal treatment strategy in lymphoma patients. PET-CT imaging is currently recommended for patients with diagnosed lymphoma that is potentially curable and 18F-FDG avid, which applies mostly to Hodgkin lymphoma (HL), diffused large B cell lymphoma (DLBCL) and follicular lymphoma (FL). The utility of PET-CT imaging depends on histological type, location and size of pathological changes (the sensitivity and specificity of the test decreases with a pathological site diameter below 10 mm) and in evaluation of the extra-nodal changes (e.g. eye socket, skin, spleen, stomach or intestines).

Initial diagnostics and pre-treatment staging

PET-CT imaging with the use of 18F-FDG, compared to another imaging methods, is characterized by the highest sensitivity and specificity in diagnosing pathological changes, except for changes in the CNS (MRI imaging as the highest value in the diagnostics of focal changes in the CNS). Therefore CT imaging should not be performed before PET-CT.

PET-CT imaging is recommended in the initial staging of early and advanced stages of HL and DLBCL. PET-CT imaging is also useful in cases of diagnosis of the FL or mantle cell lymphoma (MCL). The result of PET-CT imaging with the use of 18F-FDG modifies the primary staging in about 2530% of patients with HL and/or DLBCL and in about 60% of patients with the early stages of FL, which in consequence leads to a treatment plan modification in almost 30% of patients. PET-CT imaging allows for the more accurate measurement of the neoplastic lymph nodes, differentiation of the lymph nodes from intestinal loops or to detect the compression or thrombosis of the large mediastinal vessels. PET-CT is also a method used to detect bone marrow involvement. In patients with HL performing PET-CT imaging, regardless of its result, permits one to avoid bone marrow trepanobiopsy. In patients with DLBCL positive bone marrow involvement PET-CT imaging result permits to avoid trepanobiopsy, but in case of negative PET-CT result performing trepanobiopsy is obligatory (especially when the bone marrow infiltration may influence the treatment schedule). The interpretation of the images should be performed visually and with the use of a five degree rating scale (the Deauville system).

The initial staging of the lymphomas with differentiated accumulation of the radiotracer should be based on the CT scans with the use of an intravenous contrast. In cases of negative 18F-FDG PET-CT images of the lymphoma sites, the initial staging should be evaluated based on the CT scans obtained by PET-CT testing and the efficacy of the treatment based on the CT scans. PET-CT imaging has a limited use in cutaneous lymphomas, extra nodular marginal zone lymphomas (MZL) and small B cell lymphomas (SLL).

Treatment response evaluation

If the initial staging has been assessed by a PET-CT imaging result it is recommended to perform an early PET-CT in order to evaluate the response to treatment (so-called interim PET-CT) and another one at the end of treatment. An early PET-CT should be performed after the second chemotherapy cycle (directly before the next cycle, so that the time between the former chemotherapy administration and the PET-CT imaging was the longest possible). The aim of the interim PET-CT imaging is an early evaluation of the treatment efficacy. According to the current evidence, modification of the treatment method based on the interim PET-CT imaging result is acceptable only when the result unequivocally indicates the disease progression. In other cases, modification of the primary treatment plan is acceptable only as part of the controlled clinical trials. The interim PET-CT imaging must be evaluated both qualitatively and quantitatively.

PET-CT imaging at the treatment completion should be performed after 68 weeks after the last chemotherapy administration and 12 weeks post radiotherapy. Complete metabolic remission is equivalent with a complete response to treatment even if CT scans reveal residual masses. If PET-CT imaging detects metabolically active disease and any second line treatment can be considered it is recommended to perform a residual mass biopsy in order to obtain pathologic confirmation of the neoplasm before treatment administration. In patients receiving combined treatment, the PET-CT imaging should be performed 23 weeks after the last chemotherapy cycle in order to avoid any delay in radiotherapy onset.

The results of currently published studies show that as a negative result of the early PET-CT imaging does not guarantee the achievement of the complete remission, one cannot terminate or limit the treatment without risking treatment failure. Likewise, a positive result does not currently ensure primary resistance of the disease demanding treatment intensification.

It is particularly difficult to interpret the post-treatment PET-CT images of foci/sites of a moderately increased radiotracer 18F-FDG uptake (minimal residual uptake MRU) which some authors would consider a risk factor for disease relapse.

Post-treatment follow-up

It is not recommended to perform PET-CT imaging in patients without any clinical symptoms and who have achieved complete remission after treatment. In cases of an ambiguous result, it is recommended to repeat the PET-CT not earlier than after four weeks. If the result is still uncertain one should consider performing a biopsy or, in cases of further morphologic regression, to continue observation.

PET-CT imaging is indicated to assess an extent of relapse.

Colorectal cancer

Although PET-CT imaging has no use in the initial diagnostics of colorectal cancer, it is a valuable method to evaluate the metastases to the other organs (especially to the liver) and in the diagnostics of the disease’s relapse.

Initial diagnostics and pre-treatment staging

It is not recommended to use PET-CT imaging for the early detection and early diagnosis of colorectal cancer. The only exception is for patients with a high risk of disseminated disease (especially rectal cancer) being a contraindication for surgical treatment.

The detailed indications for the use of PET-CT imaging in the initial diagnostics of colorectal cancer are:

  • liver metastases detection in patients being candidates for surgery;
  • the detection of the dissemination beyond the liver as a part of the pre-operative diagnostics in the patients being potential candidates for recession of the liver metastases.

Post-treatment staging

PET-CT imaging is useful in the patients being candidates for local treatment of residual changes (resection, thermo-ablation, chemo-embolization).

The indications for PET-CT imaging in a post-treatment follow-up are:

  • relapse detection (especially in cases of the elevated CEA level);
  • differentiation between relapse and treatment induced changes (e.g. post radiation changes);
  • differentiation between malignant and benign changes (e.g. in the lymph nodes).

Oesophageal cancer

PET-CT imaging constitutes a supplementary method of staging oesophageal cancer and detecting distant metastases (evaluation prior to the treatment and in cases of local reoccurrence).

The PET-CT imaging is less precise than endoscopic ultrasonography in evaluating the loco-regional disease.

The value of PET-CT imaging in the treatment efficacy evaluation has not been proven.

Gastric cancer

In advanced gastric cancer, PET-CT imaging with the use of 18F-fluorothymidine (FLT) is useful in the pre-operative staging.

Hepatocellular cancer

In a case of suspected or diagnosed hepatocellular cancer, PET-CT imaging should be performed with the use of a carbon 11C isotope-labelled acetate. The method is useful in the initial staging prior to the making of therapeutic decisions. PET-CT imaging with the use of 11C traced acetate is also recommended to exclude potential distant metastases.

Pancreatic cancer

PET-CT imaging is useful in the initial differential diagnostics of suspicious changes in the pancreas and in the staging of pancreatic cancer at the different treatment stages.

There is no clinical evidence for the usefulness of PET-CT imaging in the evaluating of the treatment efficacy of pancreatic neoplasms or during post-therapy follow-up.

Liver metastases

PET-CT imaging is used in the differential diagnostics of the focal liver lesions when the neoplasm cannot be excluded by any other imaging method.

Breast cancer

PET-CT imaging in breast cancer is indicated in disease staging and in relapse detection.

Initial diagnostics and pre-treatment staging

Limited sensitivity in detecting primary neoplastic lesions of diameter less than 10 mm and lymph node axillary metastases, as well as relatively high risk of false positive results, does not allow to recommend PET-CT imaging in the initial diagnostics of breast cancer. In contrast, PET-CT imaging should be considered in patients with locally advanced disease and unequivocal results of the standard imaging methods.

Post-treatment staging

Although PET-CT imaging may be helpful for the evaluation of response to initial chemotherapy (especially in cases when clinical and mammographic evaluation is difficult), performing the initial PET-CT imaging prior to the onset of treatment is mandatory. The clinical value of the above-mentioned indication has not yet been confirmed in the prospective trials.

Post-treatment follow-up

The PET-CT imaging is useful in detecting cancer relapse in patients receiving radical treatment when the results of other tests are unequivocal and the PET-CT imaging result may contribute to changing the treatment method.

Uro-genital tract cancer

Initial diagnostics and pre-treatment staging

PET-CT imaging is useful in patients diagnosed with locally advanced cervical cancer before planning radical combined treatment (radio-chemotherapy, radiotherapy).

Although PET-CT imaging has no proved clinical value in the initial diagnostics of endometrial cancer and ovarian neoplasms, it is highly useful in disease staging.

The PET-CT imaging has no use in the primary diagnostics of testicular neoplasms. In contrast it is highly useful in disease staging (with exception of mature teratomas).

PET-CT imaging with the use of choline, fluorine (18F), carbon (11C) and prostate specific membrane antigen (PSMA)-labelled acetate is a useful test in prostate cancer staging. One should also consider performing a PET-CT test in the initial diagnostics of prostate cancer when the results of the repeated prostate biopsies are negative and, simultaneously, the prostate specific antigen level (PSA) is rising, or when one suspects the presence of metastases in the lymph nodes or bones in patients who are candidates for radical treatment.

PET-CT has no use in the initial diagnostics of the urinary bladder. The value of PET-CT in the initial diagnostics of renal cancer is limited.

Post-treatment staging

Although PET-CT imaging is useful to evaluate the efficacy of chemotherapy in patients with seminoma (also in the detection of the residual changes), it is mandatory to perform an initial PET-CT prior to the onset of treatment. PET-CT imaging cannot differentiate the character of the residual changes in a non-seminoma.

Post-treatment follow-up

PET-CT imaging is useful in detecting a relapse of the ovarian carcinoma (especially in cases of the elevated CA-125 levels) and in patients with potentially resectable metastases. There are no indications to perform a PET-CT imaging in the patients with ovarian cancer without any suspicion of a relapse.

PET-CT imaging is useful for detecting the reccurrence of cervical cancer and of endometrial cancer (especially in patients with potentially metastatic disease). PET-CT imaging is not recommended in the patients with cervical and endometrial neoplasms when there is no suspicion of a relapse.

PET-CT imaging can be used in cases of the suspicion of the local spread or distant metastases of the urinal bladder cancer in patients treated radically and when other imaging techniques have failed to distinguish the character of the changes.

PET-CT imaging with the use of choline and fluorine (18F), carbon (11C)-labelled acetate and gallium (68Ga)-labelled PSMA is useful in patients with a suspicion of prostate cancer relapse.

PET-CT imaging has no use in post-treatment monitoring of patients with uro-genital neoplasms.

Melanoma and skin neoplasms

The recommendations for use of the PET-CT imaging in patients with skin melanoma without clinical signs of regional lymph node involvement (stage: I, II and IIIa/N1a and N2a) are limited.

The PET-CT imaging cannot substitute a sentinel lymph node biopsy and is not useful in staging the disease in patients with unresectable metastases or in evaluating the response to systemic treatment. PET-CT imaging is not recommended for post-treatment observation.

PET-CT imaging with the use of 18F-FDG is characterized by a high accuracy in diagnosing Merkle cell carcinoma. In contrast, there are no recommendations for PET-CT in patients with basocellular cancer at any stage of the disease.

Initial diagnostics and pre-treatment staging

PET-CT imaging is useful in qualifying for surgery patients with melanoma in the II clinical stage (IIb/IIIc) with clinical symptoms of the metastases to the lymph modes in 30% of patients, PET-CT imaging detects distant extra-nodal metastases which results in changing the treatment method. The early detection of distant metastases is actually important due to lack of any effective systemic treatment in the IV stage of the disease.

A second but rarely used indication for performing PET-CT imaging is the initial staging in patients with potentially resectable distant organ metastases (stage IV). In the former group of patients, the result of the PET-CT imaging may lead to the substitution of surgery with systemic treatment or to a more extensive resection.

A third indication is to evaluate disease stage in patients with melanoma with diagnosed potentially resectable metastases (e.g. the lymph nodes) without a defined primary disease site.

Sarcomas

PET-CT imaging is especially useful in gastrointestinal stromal tumours (GIST) and in small-cell sarcomas (mainly Ewing sarcoma). In patients with GIST, PET-CT is the most sensitive method of monitoring the response to tyrosine kinase inhibitor treatment and of detecting the progression of the disease. PET-CT imaging differentiates, earlier than CT, the cases of disease sensitive to treatment from those which are resistant (the response can be evaluated after 12 weeks and after 26 months, respectively). In the majority of cases, an adequate evaluation of response and progression of the disease can be made by a careful interpretation of CT scans with the use of the Choi criteria to evaluate changes in size and density of the neoplastic sites.

In some individual cases, the PET-CT can be used to differentiate the primary sarcomas from benign changes in the bones and soft tissues (e.g. in von Recklinghausen’s disease).

PET-CT imaging is also used to evaluate the response to systemic treatment in some types of sarcomas (an efficacy evaluation after 13 chemotherapy cycles compared to the initial PET-CT imaging result).

Head and neck cancer

The value of the PET-CT imaging in the initial diagnostics and in the evaluation of the response to the treatment in the patients with the epithelial head and neck neoplasms is becoming systematically greater.

Initial diagnostics and pre-treatment staging

PET-CT imaging is a diagnostic method of choice for the diagnosis of lymph node metastases from an unknown primary site and enables in many cases the localization of the primary site of neoplasm in the head and neck region. However, PET-CT imaging cannot substitute a precise endoscopy of the upper respiratory tracts and of the laryngeal part of the throat with a biopsy of the clinically suspicious cites.

PET-CT should be also considered in selected patients with the diagnosis of locally advanced cancer in order to get a reliable evaluation of the lymph nodes metastases and to assess the extension of the infiltration in the primary site (this is especially important in poorly differentiated neoplasms with submucosal infiltrations). PET-CT imaging has an advantage over other imaging methods, especially in the evaluation of the regional advancement of the disease since it detects the metastases to the regular lymph nodes (the sensitivity and the specificity to detect the metastases respectively 8287% and 94100%).

PET-CT imaging is useful in radiotherapy planning (mostly in more locally or regionally advanced neoplasms) since it permits precise definition of target volume relative to the normal tissues.

Post-treatment staging

PET-CT imaging is useful in detecting residual changes after treatment and local and regional relapses. However, one must consider the possibility of false positive results (inflammatory changes especially after radiotherapy) and false negative (especially residual changes in the lymph nodes). It is obligatory to confirm the neoplastic character of the residual changes or relapse, based on the histopathological evaluation of the excision biopsy prior to qualifying any patient for salvage surgery. PET-CT imaging should not be performed earlier than after three months post radiotherapy or chemo-radiotherapy, due to the above-mentioned risk of false positive results, which is especially high in the early period after treatment.

Recently presented results justify the use of PET-CT imaging in locally advanced head and neck neoplasms after completing radiotherapy or radio-chemotherapy, as it may allow one to avoid performing a lymphadenectomy in nearly 80% of patients.

Neoplasms of the central nervous system

The aminoacid radiotracers 11C labelled methionine, 18F labelled tyrosine, 18F labelled dihydroxyphenylalanine (DOPA) play an important role in central nervous system imaging. These radiotracers show a low uptake in the normal brain tissues and in benign changes (fibrosis, necrosis, oedema). PET-CT imaging is recommended in the differential diagnostics of neoplastic and non-neoplastic changes. The sensitivity of this test in the diagnostics of primary brain tumours reaches 7894% while its specificity reaches 93100%. PET-CT imaging may be used in the intraoperative navigating technique. The results of surgery using this method suggest the possibility of radicalizing the operation and prolonging survival in this group of patients compared to those operated on based only an MRI image.

PET-CT imaging may be indicated to perform the non-invasive grading of primary CNS tumours, to define the site of a stereotactic biopsy and in cases of the suspicion of the neoplasm’s relapse.

Defining the biopsy site and tumour grading is based on the localization of the change in the PET-CT imaging and on the kinetics of the 18FET uptake in the neoplastic tumour. In high grade tumours, according to the World Health Organization (WHO) classification, positive kinetics of the radiotracer uptake is observed. This method is used to evaluate tumours placed in the eloquent regions.

The vast majority of primary CNS neoplasms are characterised by isolated local growth and the risk of dissemination into the lymphatic system or the distant metastases is extremely low. Performing a whole body PET-CT in disease staging is not applicable. The only exceptions are low-differentiated, small-cell and embryonic neoplasms, occurring mostly in children and youths (e.g. medulloblastoma, low-differentiated neuroectodermal neoplasms, germ cell tumours). In this group of patients, one should consider performing a PET-CT imaging with the use of fluorine labelled tyrosine (18F FET) or of carbon-labelled methionine (11C MET). There are no evidence-based data for the role of PET-CT imaging in the differential diagnostics of the histological types of brain tumours.

PET-CT imaging with the use of the 18F-FDG should be performed in patients with suspicion of a metastatic site in the brain of an unknown primary site.

It is indicated to perform PET-CT imaging with the use of the following: the 18F FET, 18F DOPA or 11C MET in differentiating between a resistant or recurrent and active neoplasm and a post-radiotherapy necrosis or the so-called “pseudo-progression” of the tumour. PET-CT imaging combined with the MRI spectroscopy are the methods of choice in the localization of the sites of increased cellular proliferation and higher metabolic activity. For this reason, the test should be performed before a planned stereotactic biopsy of the tumour in patients with suspected CNS neoplasm or of disease relapse in order to exclude the presence of active sites or regions of higher histological grading.

Performing PET-CT imaging with the use of the methionine tyrosine or DOPA after the neurosurgical operation is helpful in the residual tumour mass evaluation and in radiotherapy planning. PET-CT imaging with the use of methionine or fluoride-tyrosine is valuable in radiotherapy planning in patients with paragangliomas or meningiomas.

Endocrine system neoplasms

Each type of the endocrine system neoplasm has its own characteristics which can be used in PET-CT imaging. Currently, there is an access in Poland apart from 18F-FDG to gallium (68Ga)-labelled peptide radiotracers. These are very useful in diagnostics and in the monitoring of the clinical course of highly differentiated neuroendocrine neoplasms. There is a similar role for PET-CT using 18FDOPA applied in the monitoring of malignant pheochromocytoma tumours and of medullary thyroid cancer. One should expect the introduction of another radiotracers, which will enable the specific differential diagnostic of neuroendocrine system neoplasms: metomidate (in the diagnostics of the adrenal cancer), iodine (124I) (useful in dosimetry a crucial procedure prior to planned radioiodine administration to treat thyroid cancer).

PET-CT imaging with the use of 18F-FDG is not recommended in diagnostics, staging, the treatment efficacy assessment or the post-treatment follow-up in the most frequent a highly-differentiated thyroid cancer which uptakes 131I. However, this method plays a key role in the detection and localization of the metastases in patients with an increased serum level of thyreoglobulin and a negative scintigraphy result with the use of 131I. In patients with dissemination of a thyroid neoplasm with a positive iodine uptake test, the result of the 18F-FDG-PET has a prognostic value: the increased uptake of the radiotracer suggests a higher resistance to the 131I treatment.

In medullar thyroid cancer, PET-CT imaging is a reasonable method in order to localize a relapse in a case when the calcitonin level exceeds 500 pg/ml, (one should consider performing also a DOPA-PET, which is not reimbursed in Poland despite an EBM-confirmed recommendation).

In adrenal cortex carcinoma, PET-CT imaging is a complementary test in disease staging.

In some neuroendocrine neoplasms it is necessary to perform PET imaging with the use of 68Ga and 18F-FDG-labelled somatostatin derivates. The first of these radiotracers enables the evaluation of the expression of the somatostatin receptor system while the second defines the biological malignancy of the primary site and of the metastases. The results of both tests are the basis for choosing the optimal treatment method and predicting the clinical course of the disease.

Neoplasms with an unknown primary site

PET-CT imaging is useful in the diagnostics of selected patients with metastases with an unknown primary site of a neoplasm. The value of the PET-CT depends on the location of lesion (high e.g. metastases to the cervical lymph nodes, low e.g. metastases to the subdiaphragmatic lymph nodes).

Bone metastases

PET-CT (especially with the use of the 18F-NaF tracer) is characterised by a high sensitivity in the evaluation of bone metastases. This test is recommended in patients with an ambiguous result of a bone scintigraphy with the use of 99Tc-MDP. PET-CT with 18F-FDG has a slightly lower sensitivity but a higher specificity.

Proposed criteria for qualification to PET-CT imaging and list of oncological recommendations which should be reimbursed from public sources:

  • a solitary lung tumour larger than 5 mm in diameter in order to differentiate between its benign and the malignant character if the diagnosis cannot be made by any other accessible method;
  • non-small cell lung cancer in order to stage the disease prior to a planned resection or radical radiotherapy and in order to evaluate regional disease extension and to exclude distant metastases (apart from formerly detected distant metastases);
  • small-cell lung cancer in the stage of limited disease prior to radical combined treatment;
  • mesothelioma prior to radical combined treatment with the use of surgery;
  • mediastinal tumour prior to combined radical treatment with the use of surgery;
  • Hodgkin lymphoma and non-Hodgkin lymphomas in order to perform initial staging, evaluation of treatment efficacy or for early relapse detection;
  • colon cancer in order to perform pre-operative clinical staging or for early relapse detection after the radical treatment (when the results of other imaging exams are ambiguous);
  • oesophageal cancer in order to access to staging prior to radical treatment and in early relapse detection post radical treatment (when the results of other imaging exams are ambiguous);
  • gastric cancer in preoperative disease staging in cases of unequivocal results of other exams;
  • pancreatic cancer in initial disease staging prior to radical treatment (when the results of other imaging exams are ambiguous);
  • pathological changes in the pancreas and in the liver evaluation of changes suspected of a neoplastic character when a diagnosis cannot be made using other methods;
  • breast cancer in the initial diagnostics in order to exclude distant metastases (when the results of other exams are unequivocal);
  • breast cancer during the follow-up post radical treatment in cases where there is a suspicion of relapse when the results of other imaging exams are ambiguous and the PET-CT result may change the treatment method;
  • ovarian cancer in order to define the disease stage in the patients with potentially resectable metastases and for early detection of the relapse post radical treatment (in cases of increased Ca125 levels or unequivocal results of other exams);
  • cervical and endometrial cancer in order to access the disease stage prior to radical treatment and for early detection of a relapse post radical therapy (when the results of the imaging exams are unequivocal);
  • testis neoplasms (except from mature teratomas) in disease staging and in the evaluation of treatment efficacy (including residual tumour and relapse detection) if evaluation cannot be made by the other methods;
  • urinary bladder cancer in disease staging in the patient with potentially resectable metastases and in order to detect the relapse of the disease after radical treatment when the imaging exams results are unequivocal;
  • suspicion of prostate cancer in cases of elevated PSA levels and a negative result of a repeated transrectal prostate biopsy in order to localize the neoplasm the exam should be carried out with the use of acetate, choline or PSMA);
  • prostate cancer prior to radical treatment but only when the metastases and the results of other exams are unequivocal (PTE-CT imaging with the use of the acetate, choline or PSMA);
  • prostate cancer in relapse (metastases) detection post radical treatment (an exam with the use of PSMA, choline or acetate), when the diagnosis cannot be made by other exams);
  • renal cancer in relapse (metastases) detection post radical treatment (an exam with the use of acetate), when the diagnosis cannot be made by other exams);
  • melanomas in patients with clinically overt metastases to regional lymph nodes and potentially resectable metastases in the distant organs or with potentially resectable metastasis with an unknown primary site;
  • Merkel cell cancer in initial disease staging prior to the treatment and in relapse or dissemination diagnostics;
  • sarcomas in order to differentiate benign and malignant changes and in staging prior to treatment and as the reference exam in chemotherapy efficacy evaluation and during chemotherapy (after 13 chemotherapy cycles in order to compare with the initial PET-CT exam) and for the early detection of relapse;
  • gastrointestinal stromal tumours (GIST) in order to monitor the molecular response to the target treatment and to detect relapse of the disease;
  • head and neck neoplasms in order to detect the relapse of the disease early and to evaluate loco-regional staging when the results of other exams are ambiguous;
  • central nervous system in order to evaluate the areas of higher malignancy, to detect relapse early and to define the biopsy site (imaging examination indicated with the use of fluorine-labelled tyrosine, methionine or DOPA);
  • mature thyroid cancer in order to differentiate and evaluate the localization of a relapse in the cases of elevated thyroglobulin levels if this site cannot be diagnosed by other exams (it is obligatory to perform a 131I scintigraphy earlier);
  • neuroendocrine tumours in the initial disease staging; PET-CT with the use of radiotracers evaluating the expression of the receptors and with 18F-FDG in order to define the character of the metastatic changes and the grade of the malignancy prior to the planned treatment with radioisotope-labelled somatostatin analogues;
  • metastases of an unknown primary site in order to localize the primary site when it cannot be defined by other accessible exams;
  • suspicion of metastases to the bones when the result of a scintigraphy is unequivocal;
  • planning of radical radiotherapy in order to localize the foci of living neoplastic cells, of hypoxia or of the proliferation of a tumour when such an evaluation cannot be done by the other methods.

PET-MRI imaging is recommended in diagnostics of the liver, of the pelvic organs and of the brain only if the aforementioned criteria are met.

PET-MRI is recommended in children up to 18 years of age only if the aforementioned recommendations for PET-CT imaging are met.

References

I. General

  1. 1. Kumar R, Halanaik D, Malhotra A. Clinical applications of positron emission tomographycomputed tomography in oncology. Indian J Cancer 2010; 47: 100119.
  2. 2. Papathanassiou D, Bruna-Muraille C, Liehn J-C et al. Positron Emission Tomography in oncology: Present and future of PET and PET/CT. Crit Rev Oncol Hematol 2009; 72: 239254.
  3. 3. Podoloff DA, Ball DW, Ben-Josef E et al. NCCN task force: clinical utility of PET in a variety of tumor types. J Natl Compr Canc Netw 2009; 7 (Suppl 2): S126.

II. Lung and the pleura neoplasms

  1. 1. Alberts WM. Diagnosis and management of lung cancer executive summary: ACCP evidence-based clinical practice guidelines (2nd ed). Chest 2007; 132 (Suppl 3): 1S9S.
  2. 2. Berghmans T, Dusart M, Paesmans M et al. Primary tumor standardized uptake value (SUVmax) measured on fluorodeoxyglucose positron emission tomography (FDG-PET) is of prognostic value for survival in non-small cell lung cancer (NSCLC): a systematic review and meta-analysis (MA) by the European Lung Cancer Working Party for the IASLC Lung Cancer Staging Project. J Thorac Oncol 2008; 3: 612.
  3. 3. Binderup T, Knigge U, Loft A et al. Functional imaging of neuroendocrine tumors: a head-to-head comparison of somatostatin receptor scintigraphy, 123I-MIBG scintigraphy, and 18F-FDG PET. J Nucl Med 2010; 51: 704712.
  4. 4. Bradley JD, Dehdashti F, Mintun MA et al. Positron emission tomography in limited-stage small-cell lung cancer: A prospective study. J Clin Oncol 2004; 22: 32483254.
  5. 5. Bradley JD, Thorstad WL, Mutic S et al. Impact of FDG-PET on radiation therapy volume delineation in non-small-cell lung cancer. Int J Radiat Oncol Biol Phys 2004; 59: 7886.
  6. 6. Brink I, Schumacher T, Mix M et al. Impact of [18F]FDG-PET on the primary staging of small-cell lung cancer. Eur J Nucl Med Mol Imaging 2004; 31: 16141620.
  7. 7. Cheran SK, Nielsen ND, Patz EF Jr. False-negative findings for primary lung tumors on FDG positron emission tomography: staging and prognostic implications. Am J Roentgenol 2004; 182: 11291132.
  8. 8. Christensen JA, Nathan MA, Mullan BP, Hartman TE, Swensen SJ, Lowe VJ. Characterization of the solitary pulmonary nodule: 18F-FDG PET versus nodule-enhancement CT. Am J Roentgenol 2006; 187: 13611367.
  9. 9. de Geus-Oei LF, van der Heijden HF, Corstens FH, Oven WJ. Predictive and prognostic value of FDG-PET in nonsmall-cell lung cancer: a systematic review. Cancer 2007; 110: 16541664.
  10. 10. De Wever W, Stroobants S, Coolen J, Verschakelen JA. Integrated PET/CT in the staging of non-small cell lung cancer: technical aspects and clinical integration. Eur Respir J 2009; 33: 201212.
  11. 11. Dwamena MK, Kuschner WG, Rydzak CE et al. Test performance of positron emission tomography and computed tomography for mediastinal staging in patients with non-small cell lung cancer: a meta-analysis. Ann Intern Med 2003; 139: 879892.
  12. 12. Fischer B, Lassen U, Mortensen J et al. Preoperative staging of lung cancer with combined PET-CT. N Engl J Med 2009; 361: 3239 [Errata in: N Engl J Med 2011; 364: 982].
  13. 13. Fischer BM, Mortensen J, Langer SW et al. A prospective study of PET/CT in initial staging of small-cell lung cancer: comparison with CT, bone scintigraphy and bone marrow analysis. Ann Oncol 2007; 18: 338345.
  14. 14. Fletcher JW, Kymes SM, Gould M et al. Cooperative Studies Group. A comparison of the diagnostic accuracy of 18F-FDG PET and CT in the characterization of solitary pulmonary nodules. J Nucl Med 2008; 49: 179185.
  15. 15. Goodgame B, Pillot GA, Yang Z et al. Prognostic value of preoperative positron emission tomography in resected stage I non-small cell lung cancer. J Thorac Oncol 2008; 3: 130134.
  16. 16. Gould MK, Kuschner WG, Rydzak CE et al. Test performance of positron emission tomography and computed tomography for mediastinal staging in patients with non-small-cell lung cancer: a meta-analysis. Ann Intern Med 2003; 139: 879892.
  17. 17. Gould MK, Maclean CC, Kuschner WG, Rydzak CE, Owens DK. Accuracy of positron emission tomography for diagnosis of pulmonary nodules and mass lesions: a meta-analysis. JAMA 2001; 285: 914924.
  18. 18. Greco C, Rosenzweig K, Cascini GL, Tamburrini O. Current status of PET/CT for tumour volume definition in radiotherapy treatment planning for non-small cell lung cancer. Lung Cancer 2007; 57: 125134.
  19. 19. Kalff V, Hicks RJ, MacManus MP et al. Clinical impact of 18-fluorodeoxyglucose positron emission tomography in patients with non-small-cell lung cancer: a prospective study. J Clin Oncol 2001; 19: 111118.
  20. 20. Lardinois D, Weder W, Hany TF et al. Staging of non-small-cell lung cancer with integrated positron-emission tomography and computed tomography. N Engl J Med 2003; 348: 25002507.
  21. 21. Pieterman RM, van Putten JWG, Meuzelaar JJ et al. Preoperative staging of non-small cell lung cancer with positron emission tomography. N Engl J Med 2000; 343: 254261.
  22. 22. Sachs S, Bilfinger TV. The impact of positron emission tomography on clinical decision making in a university based multidisciplinary lung cancer practice. Chest 2005; 128: 698703.
  23. 23. Silvestri GA, Gould MK, Margolis ML et al. Noninvasive staging of non-small cell lung cancer: ACCP evidence-based clinical practice guidelines (2nd ed). Chest 2007; 132 (Suppl 3): 178S201S.
  24. 24. Tann M, Sandrasegaran S, Winer-Muram HT et al. Can 18FFDG- PET be used to predict growth of stage I lung cancer? Clin Radiology 2008; 63: 856863.
  25. 25. Tournoy KG, Maddens S, Gosselin R, Van Maele G, Van Meerbeeck JP, Kelles A. Integrated FDG-PET/CT does not make invasive staging of the intrathoracic lymph nodes in non-small cell lung cancer redundant: a prospective study. Thorax 2007; 62: 696701.
  26. 26. Vansteenkiste JF, Stroobants SS. PET scan in lung cancer: current recommendations and innovation. J Thorac Oncol 2006; 1: 7173.
  27. 27. Zwahlen D, Wyss MT, Stumpe KD et al. Whole-Body 18F-FDG PET improves the management of patients with small cell lung cancer. J Nucl Med 2003; 44: 19111917.

III. Lymphoma

  1. 1. Avigdor A. Staging DLBCL: Bone marrow biopsy or PET-CT? Blood 2013; 122: 45.
  2. 2. Cheson BD. Role of functional imaging in the management of lymphoma. J Clin Oncol 2011; 29: 18441854.
  3. 3. Cheson BD, Pfistner B, Juweid ME. Revised response criteria for malignant lymphoma. J Clin Oncol 2007; 25: 579586.
  4. 4. Dickinson M, Hoyt R, Roberts AW et al. Improved survival for relapsed diffuse large B cell lymphoma is predicted by a negative pre-transplant FDG-PET scan following salvage chemotherapy. Br J Haematol 2010; 150: 3945.
  5. 5. Derenzini E, Musuraca G, Fanti S et al. Pretransplantation positron emission tomography scan is the main predictor of autologous stem cell transplantation outcome in aggressive B-cell non-Hodgkin lymphoma. Cancer 2008; 113: 24962503.
  6. 6. El-Galaly T, d‘Amore F, Mylam K et al. Routine bone marrow biopsy has little or no therapeutic consequence for positron emission tomography/computed tomography-staged treatment-naive patients with Hodgkin lymphoma. J Clin Oncol 2012; 30: 45084514.
  7. 7. Gallamini A, Rigacci L, Merli F et al. The predictive value of positron emission tomography scanning performed after two courses of standard therapy on treatment outcome in advanced stage Hodgkin‘s disease. Haematologica 2006; 91: 475481.
  8. 8. Isasi CR, Lu P, Blaufox MD. A meta-analysis of 18F-2-deoxy-2-fluoro-D-glucose positron emission tomography in the staging and restaging of patients with lymphoma. Cancer 2005; 104: 10661074.
  9. 9. Itti E, Lin C, Dupuis J et al. Prognostic value of interim 18F-FDG PET in patients with diffuse large B-Cell lymphoma: SUV-based assessment at 4 cycles of chemotherapy. J Nucl Med 2009; 50: 527533.
  10. 10. Juweid ME, Stroobants S, Hoekstra OS et al. Use of positron emission tomography for response assessment of lymphoma: consensus of the Imaging Subcommittee of International Harmonization Project in Lymphoma. J Clin Oncol 2007; 25: 571578.
  11. 11. Khan A, Barrington S, Mikhaeel N et al. PET-CT staging of DLBCL accurately identifies and provides new insight into the clinical significance of bone marrow involvement. Blood 2013; 122: 6167.
  12. 12. MacManus MP, Seymour JF, Hicks RJ. Overview of early response assessment in lymphoma with FDG-PET. Cancer Imaging 2007; 7: 1018.
  13. 13. Paes FM, Kalkanis DG, Sideras PA et al. FDG PET/CT of extranodal involvement in non-Hodgkin lymphoma and Hodgkin disease. Radiographics 2010; 30: 269291.
  14. 14. Pelosi E, Pregno P, Penna D. Role of whole-body [18F] fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) and conventional techniques in the staging of patients with Hodgkin and aggressive non-Hodgkin lymphoma. Radiol Med 2008; 113: 578590.
  15. 15. Qiao W, Zhao J, Wang C et al. Predictive value of (18)F-FDG hybrid PET/CT for the clinical outcome in patients with non-Hodgkin’s lymphoma prior to and after autologous stem cell transplantation. Hematology 2010; 15: 2127.
  16. 16. Rigacci L, Vitolo U, Nassi L et al. Positron emission tomography in the staging of patients with Hodgkin’s lymphoma. A prospective multicentric study by the Intergruppo Italiano Linfomi. Ann Hematol 2007; 86: 897903.
  17. 17. Terasawa T, Nihashi T, Hotta T et al. 18F-FDG PET for posttherapy assessment of Hodgkin’s disease and aggressive Non-Hodgkin’s lymphoma: a systematic review. J Nucl Med 2008; 49: 1321.
  18. 18. Zijlstra JM, Lindauer-van der Werf G, Hoekstra OS et al. 18F-fluoro-deoxyglucose positron emission tomography for post-treatment evaluation of malignant lymphoma: a systematic review. Haematologica 2006; 91: 522529.
  19. 19. Zinzani P, Rigacci L, Stefoni V et al. Early interim 18F-FDG PET in Hodgkin‘s lymphoma: Evaluation on 304 patients. Eur J Nucl Med Mol Imaging 2012; 39: 412.

IV. Colorectal cancer

  1. 1. Domper M, Matute R, Martínez-Lázaro R et al. 18F-FDG positron emission tomography staging and restaging in rectal cancer treated with preoperative chemoradiation. Int J Radiat Oncol Biol Phys 2004; 58: 528535.
  2. 2. Hicks RJ, Drummond EG, Keck J et al. Does positron emission tomography change management in primary rectal cancer? A prospective assessment. Dis Colon Rectum 2004; 47: 451458.
  3. 3. Huebner RH, Park KC, Shepherd JE et al. A metanalysis of the literature for whole body FDG-PET Detection of Recurrent Colorectal Cancer. J Nucl Med 2000; 41: 11711189.
  4. 4. Jadvar H, Alavi A, Gambhir SS. 18F-FDG uptake in lung, breast, and colon cancers: molecular biology correlates and disease characterization. J Nucl Med 2009; 50: 18201827.
  5. 5. Kinkel K, Lu Y, Both M et al. Detection of hepatic metastases from cancers of the gastrointestinal tract by using noninvasive imaging methods (US, CT, MR imaging, PET): a meta-analysis. Radiology 2002; 224: 748756.
  6. 6. Pasetto LM, Rossi E, Monfardini S. Liver Metastases of Colorectal Cancer: Medical Treatments (Review). Anticancer Res 2003; 23: 42454256.
  7. 7. Tsunoda Y, Ito M, Fujii H, Kuwano H, Saito N. Preoperative diagnosis of lymph node metastases of colorectal cancer by FDG-PET/CT. Jpn J Clin Oncol 2008; 38: 347353.
  8. 8. Utzschneider CR, Becker R, Tiling MF et al. Whole-body MRI at 1.5 T and 3 T compared with FDG-PET-CT for the detection of tumour recurrence in patients with colorectal cancer. Eur Radiol 2009; 19: 13661378.

V. Oesophageal cancer

  1. 1. Downey RJ, Akhurst T, Ilson D et al. Whole Body 18F- FDG-PET and the response of esophageal cancer to induction therapy: results of a prospective trial. J Clin Oncol 2003; 21: 428432.
  2. 2. Heeren PAM, Jager PL, Bongaerts F et al. Detection of distant metastases in esophageal cancer with 18F-FDG PET. J Nucl Med 2004; 45: 980987.
  3. 3. Kato H, Kuwano H, Nakajima M et al. Comparison between positron emission tomography and computed tomography in the use of the assessment of esophageal carcinoma. Cancer 2002; 94: 921928.
  4. 4. Kneist W, Schreckenberger M, Bartenstein P et al. Positron emission tomography for staging esophageal cancer: Does it lead to a different therapeutic approach? World J Surg 2003; 27: 11051112.
  5. 5. Noble F, Bailey D. SWCIS Upper Gastrointestinal Tumour Panel, Tung K, Byrne JP. Impact of integrated PET/CT in the staging of oesophageal cancer: a UK population-based cohort study. Clin Radiol 2009; 64: 699705.
  6. 6. Räsänen JV, Sihvo EIT, Knuuti MJ et al. Prospective analysis of accuracy of positron emission fomography, computed tomography, and endoscopic ultrasonography in staging of adenocarcinoma of the esophagus and the esophagogastric junction. Ann Surg Oncol 2003; 10: 954960.
  7. 7. Sendler A. Metabolic response evaluation by PET during neoadjuvant treatment for adenocarcinoma of the esophagus and esophagogastric junction. Recent Results Cancer Res 2010; 182: 167177.
  8. 8. van Westreenen HL, Heeren PAM, Jager PL et al. Pitfalls of positive findings in staging esophageal cancer with F-18-fluorodeoxyglucose positron emission tomography. Ann Surg Oncol 2003; 10: 11001105.
  9. 9. Wren SM, Stijns P, Srinivas S. Positron emission tomography in the initial staging of esophageal cancer. Arch Surg 2002; 137: 10011006.
  10. 10. Young Cheol Y, Kyung Soo L, Young Mog S et al. Metastasis to regional lymph nodes in patients with esophageal squamous cell carcinoma: CT versus FDG PET for presurgical detection. Prospective study. Radiology 2003; 227: 764770.
  11. 11. Zhong X, Yu J, Zhang B et al. Using 18F-fluorodeoxyglucose positron emission tomography to estimate the length of gross tumor in patients with squamous cell carcinoma of the esophagus. Int J Radiat Oncol Biol Phys 2009; 73: 136141.

VI. Other malignant neoplasms of the gastrointestinal tract

  1. 1. Hwang KH, Choi DJ, Lee SY et al. Evaluation of patients with hepatocellular carcinomas using [(11)C]acetate and [(18)F]FDG PET/CT: a preliminary study. Appl Radiat Isot 2009; 67: 11951198.
  2. 2. Mataki Y, Shinchi H, Kurahara H et al. Clinical usefulness of FDG-PET for pancreatic cancer. Gan To Kagaku Ryoho 2009; 36: 25162520.
  3. 3. Petrowsky H, Wildbrett P, Husarik DB et al. Impact of integrated positron emission tomography and computed tomography on staging and management of gallbladder cancer and cholangiocarcinoma. J Hepatol 2006; 45: 4350.
  4. 4. Salem N, Kuang Y, Wang F et al. PET imaging of hepatocellular carcinoma with 2-deoxy-2[18F]fluoro-D-glucose, 6-deoxy-6[18F] fluoro-D-glucose, [1-11C]-acetate and [N-methyl-11C]-choline. Nucl Med Mol Imaging 2009; 53: 144156.
  5. 5. Schick V, Franzius C, Beyna T et al. Diagnostic impact of 18F-FDG PET-CT evaluating solid pancreatic lesions versus endosonography, endoscopic retrograde cholangio-pancreatography with intraductal ultrasonography and abdominal ultrasound. Eur J Nucl Med. Mol Imaging 2008; 35: 17751785.

VII. Breast cancer

  1. 1. Bourgeois AC, Warren LA, Chang TT et al. Role of positron emission tomography/computed tomography in breast cancer. Radiol Clin North Am 2013; 51: 781798.
  2. 2. Cintolo JA, Tchou J, Pryma DA. Diagnostic and prognostic application of positron emission tomography in breast imaging: emerging uses and the role of PET in monitoring treatment response. Breast Cancer Res Treat 2013; 138: 331346.
  3. 3. Cooper KL, Meng Y, Harnan S et al. Positron emission tomography (PET) and magnetic resonance imaging (MRI) for the assessment of axillary lymph node metastases in early breast cancer: systematic review and economic evaluation. Health Technol Assess 2011; 15: iiiiiv, 1134.
  4. 4. Fuster D, Duch J, Paredes P et al. Preoperative staging of large primary breast cancer with [18F]fluorodeoxyglucose positron emission tomography/computed tomography compared with conventional imaging procedures. J Clin Oncol 2008; 26: 47464751.
  5. 5. Gil-Rendo A, Zornoza G, Garcia-Velloso MJ et al. Fluorodeoxyglucose positron emission tomography with sentinel lymph node biopsy for evaluation of axillary involvement in breast cancer. Br J Surg 2006; 93: 707712.
  6. 6. Hodgson NC, Gulenchyn KY. Is there a role for positron emission tomography in breast cancer staging? J Clin Oncol 2008; 26: 712720.
  7. 7. Lee JH. Radionuclide methods for breast cancer staging. Semin Nucl Med 2013; 43: 294298.
  8. 8. Niikura N, Costelloe CM, Madewell JE et al. FDG-PET/CT compared with conventional imaging in the detection of distant metastases of primary breast cancer. Oncologist 2011; 16: 11111119.
  9. 9. Peare R, Staff RT, Heys SD. The use of FDG-PET in assessing axillary lymph node status in breast cancer: a systematic review and meta-analysis of the literature. Breast Cancer Res Treat 2010; 123: 281290.
  10. 10. Pennant M, Takwoingi Y, Pennant L et al. A systematic review of positron emission tomography (PET) and positron emission tomography/computed tomography (PET/CT) for the diagnosis of breast cancer recurrence. Health Technol Assess 2010; 14: 1103.
  11. 11. Rosen EL, Eubank WB, Mankoff DA et al. FDG PET, PET/CT, and breast cancer imaging. Radiographics 2007; 27 (Suppl 1): S215S229.
  12. 12. Veronesi U, De Cicco C, Galimberti VE et al. A comparative study on the value of FDG-PET and sentinel node. Ann Oncol 2007; 18: 473478.

VIII. Uro-genital tract cancer

  1. 1. Albers P, Bender H, Yilmaz H et al. Positron emission tomography in the clinical staging of patients with Stage I and II testicular germ cell tumors. Urology 1999; 53: 808811.
  2. 2. Albrecht S, Buchegger F, Soloviev D et al. 11C-acetate PET in the early evaluation of prostate cancer recurrence. Eur J Nucl Med Mol Imaging 2007; 34: 185196.
  3. 3. Basu S, Rubello D. PET imaging in the management of tumors of testis and ovary: current thinking and future directions. Minerva Endocrinol 2008; 33: 229256.
  4. 4. Becherer A, De Santis M, Karanikas G et al. FDG PET is superior to CT in the prediction of viable tumour in post-chemotherapy seminoma residuals. Eur J Radiol 2005; 54: 284288.
  5. 5. Bokemeyer C, Kollmannsberger C, Oechsle K et al. Early prediction of treatment response to high dose salvage chemotherapy in patients with relapsed germ cell cancer using [18F]FDG PET. Br J Cancer 2002; 86: 506511.
  6. 6. Bokemeyer C, Kollmannsberger C, Oechsle K et al. Early prediction of treatment response to high dose salvage chemotherapy in patients with relapsed germ cell cancer using [18F]FDG PET. Br J Cancer 2002; 86: 506511.
  7. 7. Brogsitter C, Zöphel K, Kotzerke J. 18F-Choline, 11C-choline and 11C-acetate PET/CT: comparative analysis for imaging prostate cancer patients. Eur J Nucl Med Mol Imaging 2013; 40 (Suppl 1): S18S27.
  8. 8. de Jong JS, van Ginkel RJ, Slart RH et al. FDG-PET probe-guided surgery for recurrent retroperitoneal testicular tumor recurrences. Eur J Surg Oncol 2010; 36: 10921095.
  9. 9. De Santis M, Becherer A, Bokemeyer C et al. 2-18fluoro-deoxy-D-glucose positron emission tomography is a reliable predictor for viable tumor in postchemotherapy seminoma: an update of the prospective multicentric SEMPET trial. J Clin Oncol 2004; 22: 10341039.
  10. 10. De Santis M, Bokemeyer C, Becherer A et al. Predictive impact of 2-18fluoro-2-deoxy-D-glucosepositron emission tomography for residual post chemotherapy masses in patients with bulky seminoma. J Clin Oncol 2001; 19: 37403744.
  11. 11. Ehrlich Y, Brames MJ, Beck SD et al. Long-term follow-up of Cisplatin combination chemotherapy in patients with disseminated nonseminomatous germ cell tumors: is a postchemotherapy retroperitoneal lymph node dissection needed after complete remission? J Clin Oncol 2010; 28: 531536.
  12. 12. Even-Sapir E, Mishani E, Flusser G et al. 18F-fluoride positron emission tomography and positron emission tomography/computed tomography. Semin Nucl Med 2007; 37: 462469.
  13. 13. Fanti S, Nanni C, Ambrosini V et al. PET in genitourinary tract cancers. Q J Nucl Med Mol Imaging 2007; 51: 260271.
  14. 14. Gadducci A, Cosio S, Zola P et al. Surveillance procedures for patients treated for epithelial ovarian cancer: a review of the literature. Int J Gynecol Cancer 2007; 17: 2131.
  15. 15. Graafland NM, Leijte JA, Valdés Olmos RA et al. Scanning with 18F-FDG-PET/CT for detection of pelvic nodal involvement in inguinal node-positive penile carcinoma. Eur Urol 2009; 56: 339345.
  16. 16. Hain SF, O’Doherty MJ, Timothy AR et al. Fluorodeoxyglucose PET in the initial staging of germ cell tumours. Eur J Nucl Med 2000; 27: 590594.
  17. 17. Karapetis CS, Strickland AH, Yip D et al. Use of fluorodeoxyglucose positron emission tomography scans in patients with advanced germ cell tumour following chemotherapy: single-centre experience with long-term follow up. Intern Med J 2003; 33: 427435.
  18. 18. Kochhar R, Taylor B, Sangar V. Imaging in primary penile cancer: current status and future directions. Eur Radiol 2010; 20: 3647.
  19. 19. Kollmannsberger C, Oechsle K, Dohmen BM et al. Prospective comparison of [18F]fluorodeoxyglucose positron emission tomography with conventional assessment by computed tomography scans and serum tumor markers for the evaluation of residual masses in patients with nonseminomatous germ cell carcinoma. Cancer 2002; 94: 23532362.
  20. 20. Lai CH, Yen TC, Chang TC. Positron emission tomography imaging for gynecologic malignancy. Curr Opin Obstet Gynecol 2007; 19: 3741.
  21. 21. Lassen U, Daugaard G, Eigtved A et al. Whole body FDG-PET in patients with stage I nonseminomatous germ cell tumours. Eur J Nucl Med Mol Imaging 2003; 30: 396402.
  22. 22. Leskinen S, Elomaa I, Minn H et al. Detection of residual tumours in post chemotherapy testicular cancer by FDG-PET. Eur J Cancer 1997; 33: 12341241.
  23. 23. Lumbroso J, Theodore C, Foehrenbach H et al. Fluorodeoxyglucose (FDO) positron emission tomography (PET) in testicular germ cell tumors in adults: preliminary French clinical evaluation, development of the technique and its clinical applications. Prog Urol 2000; 10: 11901199.
  24. 24. O’Doherty MJ, Timothy AR, Leslie MD et al. Fluorodeoxyglucose positron emission tomography in the evaluation of germ cell tumours at relapse. Br J Cancer 2000; 83: 863869.
  25. 25. Oyama N, Okazawa H, Kusukawa N et al. 11C-Acetate PET imaging for renal cell carcinoma. Eur J Nucl Med Mol Imaging 2009; 36: 422427.
  26. 26. Park JW, Jo MK, Lee HM et al. Significance of 18F-fluorodeoxyglucose positron-emission tomography/computed tomography for the postoperative surveillance of advanced renal cell carcinoma. BJU Int 2009; 103: 615619.
  27. 27. Picchio M, Treiber U, Beer AJ et al. Value of 11C-Choline PET and contrast-enhanced CT for staging of bladder cancer: correlation with histopathologic findings. J Nucl Med 2006; 47: 938944.
  28. 28. Reske SN, Blumstein NM, Neumaier B et al. Imaging Prostate Cancer with 11C-Choline PET/CT. J Nucl Med 2006; 47: 12491254.
  29. 29. Rufini V, Calcagni ML, Baum RP. Imaging of neuroendocrine tumors. Semin Nucl Med. 2006; 36: 228247.
  30. 30. Sephens AW, Gonin R, Hutchins GD et al. Positron emission tomography evaluation of residual radiographic abnormalities in post chemotherapy germ cell tumor patients. J Clin Oncol 1996; 14: 16371641.
  31. 31. Spermon JR, De Geus-Oei LF, Kiemeney LALM et al. The role of 18fluoro-2-deoxyglucose positron emission tomography in initial staging and re-staging after chemotherapy for testicular germ cell tumours. BJU Int 2002; 89: 549556.
  32. 32. Wachter S, Tomek S, Kurtaran A et al. 11C-Acetate positron emission tomography imaging and image fusion with computed tomography and magnetic resonance imaging in patients with recurrent prostate cancer. J Clin Oncol 2006; 24: 25132519.
  33. 33. Wildberger JE, Borchers H, Zimny M et al. Does positron emission tomography using 18-fluoro-2-deoxyglucoseimprove clinical staging of testicular cancer? Results of a study in 50 patients. Urology 1999; 54: 900904.
  34. 34. Yang Z, Cheng J, Pan L, Hu S et al. Is whole-body fluorine-18 fluorodeoxyglucose PET/CT plus additional pelvic images (oral hydration-voiding-refilling) useful for detecting recurrent bladder cancer? Ann Nucl Med 2012; 26: 571577.

IX. Melanoma

  1. 1. Barsky M, Cherkassky L, Vezeridis M, Miner TJ. The role of preoperative positron emission tomography/computed tomography (PET/CT) in patients with high-risk melanoma. J Surg Oncol 2014; 109: 726729.
  2. 2. Bastiaannet E, Oyen WJ, Meijer S et al. Impact of [18F]fluorodeoxyglucose positron emission tomography on surgical management of melanoma patients. Br J Surg 2006; 93: 243249.
  3. 3. Bastiaannet E, Uyl-de Groot CA, Brouwers AH et al. Cost-effectiveness of adding FDG-PET or CT to the diagnostic work-up of patients with stage III melanoma. Ann Surg 2012; 255: 771776.
  4. 4. Bourgeois AC, Chang TT, Fish LM, Bradley YC. Positron emission tomography/computed tomography in melanoma. Radiol Clin North Am 2013; 51: 865879.
  5. 5. El-Maraghi RH, Kielar AZ. PET vs sentinel lymph node biopsy for staging melanoma: a patient intervention, comparison, outcome analysis. J Am Coll Radiol 2008; 5: 924931.
  6. 6. Gulec SA, Faries MB, Lee CC et al. The role of fluorine-18 deoxyglucose positron emission tomography in the management of patients with metastatic melanoma: impact on surgical decision making. Clin Nucl Med 2003; 28: 961965.
  7. 7. Krug B, Crott R, Lonneux M et al. Role of PET in the initial staging of cutaneous malignant melanoma: systematic review. Radiology 2008; 249: 836844.
  8. 8. Oberholzer PA, Urosevic M, Steinert HC et al. Baseline staging of melanoma with unknown primary site: the value of serum s100 protein and positron emission tomography. Dermatology 2008; 217: 351355.
  9. 9. Römer W, Nömayr A, Greess H et al. Retrospective interactive rigid fusion of (18)F-FDG-PET and CT. Additional diagnostic information in melanoma patients. Nuklearmedizin 2006; 45: 8895.
  10. 10. Singh B, Ezziddin S, Palmedo H et al. Preoperative 18F-FDG-PET/CT imaging and sentinel node biopsy in the detection of regional lymph node metastases in malignant melanoma. Melanoma Res 2008; 18: 346352.
  11. 11. Veit-Haibach P, Vogt FM, Jablonka R et al. Diagnostic accuracy of contrast-enhanced FDG-PET/CT in primary staging of cutaneous malignant melanoma. Eur J Nucl Med Mol Imaging 2009; 36: 910918.
  12. 12. Wagner T, Meyer N, Zerdoud S et al. Fluorodeoxyglucose positron emission tomography fails to detect distant metastases at initial staging of melanoma patients with metastatic involvement of sentinel lymph node. Br J Derm 2011; 164: 12351240.

X. Bones and soft tissues sarcomas

  1. 1. Antoch G, Kanja J, Bauer S et al. Comparison of PET, CT, and dual-modality PET/CT imaging for monitoring of imatinib (STI571) therapy in patients with gastrointestinal stromal tumors. J Nucl Med 2004; 45: 357365.
  2. 2. Bastiaannet E, Groen H, Jager PL et al. The value of FDG PET in the detection, grading and response to therapy of soft tissue and bone sarcomas; a systematic review and meta-analysis. Cancer Treat Rev 2004; 30: 83101.
  3. 3. Choi H, Charnsangavej Ch, Faria SC et al. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinibmesylate: proposal of new computed tomography response criteria. J Clin Oncol 2007; 25: 17531759.
  4. 4. Costelloe CM, Chuang HH, Madewell JE. FDG PET/CT of primary bone tumors. Am J Roentgenol 2014; 202: 521531.
  5. 5. Franzius C, Sciuk J, Brinkschmidt C et al. Evaluation of chemotherapy response in primary bone tumors with F-18 FDG positron emission tomography compared with histologically assessed tumor necrosis. Clin Nucl Med 2000; 25: 874881.
  6. 6. Gayed I, Vu T, Iyer R et al. The role of 18F-FDG PET in staging and early prediction of response to therapy of recurrent gastrointestinal stromal tumors. J Nucl Med 2004; 45: 1721.
  7. 7. Goerres GW, Stupp R, Barghouth G et al. The value of PET, CT and in-line PET/CT in patients with gastrointestinal stromal tumours: long-term outcome of treatment with imatinibmesylate. Eur J Nucl Med Mol Imaging 2005; 32: 153162.
  8. 8. Hawkins DS, Schuetze SM, Butrynski JE et al. [18F]Fluorodeoxyglucose positron emission tomography predicts outcome for Ewing sarcoma family of tumors. J Clin Oncol 2005; 23: 88288834.
  9. 9. Holdsworth CH, Badawi RD, Manola JB et al. CT and PET: Early prognostic indicators of response to imatinibmesylate in patients with gastrointestinal stromal tumor. Am J Roentgenol 2007; 189: 324330.
  10. 10. Jager PL, Gietema J, Avan der Graaf WT. Imatinibmesylate for the treatment of gastrointestinal stromal tumours: best monitored with FDG PET. Nucl Med Commun 2004; 25: 433438.
  11. 11. Khamly KK, Hicks RJ, McArthur GA et al. The promise of PET in clinical management and as a sensitive test for drug cytotoxicity in sarcomas. Expert Rev Mol Diagn 2008; 8: 105119.
  12. 12. Lisle JW, Eary JF, O’Sullivan J et al. Risk assessment based on FDG-PET imaging in patients with synovial sarcoma. Clin Orthop Relat Res 2009; 467: 16051611.
  13. 13. McAuliffe JC, Hunt KK, Lazar AJF et al. A randomized, phase II study of preoperative plus postoperative imatinib in GIST: evidence of rapid radiographic response and temporal induction of tumor cell apoptosis. Ann Surg Oncol 2009; 16: 910919.
  14. 14. Prior JO, Montemurro M, Orcurto M-V et al. Early prediction of response to sunitinib after imatinib failure by 18F-Fluorodeoxyglucose positron emission tomography in patients with gastrointestinal stromal tumor. J Clin Oncol 2008; 27: 439445.
  15. 15. Schuetze SM. Utility of positron emission tomography in sarcomas. Curr Opin Oncol 2006; 18: 369373.
  16. 16. Schwarzbach MH, Hinz U, Dimitrakopoulou-Strauss A et al. Prognostic significance of preoperative [18-F] fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging in patients with resectable soft tissue sarcomas. Ann Surg 2005; 241: 286294.
  17. 17. Stroobants S, Goeminne J, Seegers M et al. 18F- FDG-Positron emission tomography for the early prediction of response in advanced soft tissue sarcoma treated with imatinibmesylate. Eur J Cancer 2003; 39: 20122020.
  18. 18. Toner GC, Hicks RJ. PET for sarcomas other than gastrointestinal stromal tumors. Oncologist 2008; 13 (Suppl 2): S22S26.

XI. Head and neck cancer

  1. 1. Bussink J, van Herpen CM, Kaanders JH et al. PET-CT for response assessment and treatment adaptation in head and neck cancer. Lancet Oncol 2010; 11: 661669.
  2. 2. Graham MM, Hoffman HT, Smith RB et al. The role of post-radiatiotherapy FDG PET in prediction of necessity for post-radiation therapy neck dissection in locally advanced head-and-neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys 2004; 59: 10011010.
  3. 3. Kwee RM. Prediction of tumor response to neoadjuvant therapy in patients with esophageal cancer with use of 18F FDG PET: a systematic review. Radiology 2010; 254: 707717.
  4. 4. Mehanna HM, Wong WL, McConkey CC et al. PET-NECK: a multicentre, randomized, phase III trial comparing PET-CT guided active surveillance with planned neck dissection for locally advanced (N2/N3) nodal metastases in patients with head and neck squamous cell cancer treated with primary radical chemoradiotherapy. J Clin Oncol 2015; 33 (Suppl): a6009.
  5. 5. Schöder H, Fury M, Lee N. PET monitoring of therapy response in head and neck squamous cell carcinoma. J Nucl Med 2009; 50 (Suppl 1): S74S88.
  6. 6. Troost EG, Schinagl DA, Bussink J et al. Clinical evidence on PET-CT for radiation therapy planning in head and neck tumours. Radiother Oncol 2010; 96: 328334.
  7. 7. Troost EG, Schinagl DA, Bussink J et al. Innovations in radiotherapy planning of head and neck cancers: role of PET. J Nucl Med 2010; 51: 6676.
  8. 8. Wong RJ. Current status of FDG-PET for head and neck cancer. J Surg Oncol 2008; 97: 649652.

XII. Neoplasms of the central nervous system

  1. 1. Benard F, Romsa J, Hustinx R. Imaging gliomas with positron emission tomography and single-photon emission computed tomography. Semin Nucl Med 2003; 33: 148162.
  2. 2. Ceyssens S, van Laere K, de Groot T et al. [11C]Methionine PET, histopathology and survival in primary brain tumors and recurrence. AJNR 2006; 27: 14321437.
  3. 3. Chen W, Silverman DHS. Advances in evaluation of primary brain tumors. Semin Nucl Med 2008; 38: 240250.
  4. 4. Feldman SC, Chu D, Schulder M et al. The blood oxygen level-dependent functional MR imaging signal can be used to identify brain tumors and distinguish them from normal tissue. AJNR 2009; 30: 389395.
  5. 5. Gómez-Río M, Rodríguez-Fernández A, Ramos-Font C et al. Diagnostic accuracy of 201Thallium-SPECT and 18F-FDG-PET in the clinical assessment of glioma recurrence. Eur J Nucl Med Mol Imaging 2008; 35: 966975.
  6. 6. Hatakeyama T, Kawai N, Nishiyama Y et al. 11C-methionine (MET) and 18F-fluorothymidine (FLT) PET in patients with newly diagnosed glioma. Eur J Nucl Med Mol Imaging 2008; 11: 20092017.
  7. 7. Kato T, Shinoda J, Nakayama N et al. Metabolic assessment of gliomas using 11C-Methionine, [18F] Fluorodeoxyglucose, and 11C-Choline positron-emission tomography. AJNR 2008; 29: 11761182.
  8. 8. Kato T, Shinoda J, Oka N et al. Analysis of 11C-methionine uptake in low-grade gliomas and correlation with proliferative activity. AJNR 2008; 29: 18671871.
  9. 9. Kosaka N, Tsuchida T, Uematsu H et al. 18F-FDG PET of common enhancing malignant brain tumors. Am J Roentgenol 2008; 190: 365369.
  10. 10. Majos C, Aguilera C, Alonso J et al. Proton MR spectroscopy improves discrimination between tumor and pseudotumoral lesion in solid brain masses. AJNR 2009; 30: 544551.
  11. 11. Noguchi T, Yoshiura T, Hiwatashi A et al. Perfusion imaging of brain tumors using arterial spin labeling: correlation with histopathologic vascular density. AJNR 2008; 29: 689693.
  12. 12. Pinker K, Noebauer-Huhmann IM, Stavrou I et al. High-resolution contrast-enhanced, susceptibility-weighted MR imaging at 3T in patients with brain tumors: Correlation with positron-emission tomography and histopathologic findings. AJNR 2007; 28: 12801286.
  13. 13. Sadeghi N, Salmon I, Decaestecker C et al. Stereotactic comparison among cerebral blood volume, methionine uptake, and histopathology in brain glioma. AJNR 2007; 28: 455461.
  14. 14. Schiepers Ch, Chen W, Dahlbom M et al. 18F-fluorothymidine kinetics of malignant brain tumors. Eur J Nucl Med Mol Imaging 2007; 7: 10031011.
  15. 15. Suchorska B, Tonn JC, Jansen NL. PET imaging for brain tumor diagnostisc. Curr Opin Neurol 2014; 27: 683688.
  16. 16. Wong TZ, van der Westhuizen GJ, Coleman RE. Positron emission tomography imaging of brain tumors. Neuroimaging Clin N Am 2002; 12: 615626.

XIII. Thyroid cancer

  1. 1. Brandt-Mainz K, Muller SP, Gorges R et al. The value of 18F-FDG in patients with medullary thyroid cancer. Eur J Nucl Med 2000; 27:490496.
  2. 2. Conti PS, Durski JM, Bacqai F et al. Imaging of locally recurrent and metastatic thyroid cancer with PET. Thyroid 1999; 9: 797804.
  3. 3. Dietlein M, Scheidhauer K, Voth E et al. Fluorine-18fluorodeoxyglucose positron emission tomography and iodine-131 Whole-body scintigraphy in the follow up of differentiated thyroid cancer. Eur J Nucl Med 2007; 24: 13421348.
  4. 4. Giammarile F, Hafdi Z, Bournaud C et al. Is [18F]-2-fluoro-2-deoxy-d-glucose (FDG)scintigraphy with non-dedicated positron emission tomography useful in the diagnostic management of suspected metastatic thyroid carcinoma in patients with no detectable radioiodine uptake? Eur J Endocrinol 2003; 149: 293300.
  5. 5. Grunwald F, Menzel C, Bender H et al. Comparison of 18F-FDG-PET. Thyroid 1997; 7: 327335.
  6. 6. Helal BO, Merlet P, Toubert ME et al. Clinical impact of 18F-FDG PET in thyroid carcinoma patients with elevated thyroglobulin levels and negative 131I scanning results after therapy. J Nucl Med 2001; 42: 14641469.
  7. 7. Muros MA, Llamas-Elvira JM, Ramirez-Navarro A et al. Utility of 18F- FDG in differentiated thyroid carcinoma with negative radioiodine scans andelevated serum thyroglobulin levels. Am J Surg 2000; 179: 457461.
  8. 8. Wu HS, Huang WS, Liu YC et al. Comparison of FDGPET and technetium-99m MIBI SPECT to detect metastatic cervical lymph nodes in well-differentiated thyroid carcinoma with elevated serum HTG but negative I-131 whole body scan. Anticancer Res 2003; 23: 42354238.

XIV. Metastasis of the unknown primary site

  1. 1. Alberini JL, Belhocine T, Hustinx R et al. Whole-body positron emission tomography using fluorodeoxyglucose in patients with metastases of unknown primary tumours (CUP syndrome). Nucl Medi Commun 2003; 24: 10811086.
  2. 2. Fogarty GB, Peters LJ, Stewart J et al. The usefulness of fluorine 18-labelled deoxyglucose positron emission tomography in the investigation of patients with cervical lymphadenopathy from an unknown primary tumor. Head Neck 2003; 25: 138145.
  3. 3. Johansen J, Eigtved A, Buchwald C et al. Implication of 18F-fluoro-2-deoxy-D-glucose positron emission tomography on management of carcinoma of unknown primary in the head and neck: a Danish cohort study. Laryngoscope 2002; 112: 20092014.
  4. 4. Klee B, Law I, Højgaard L et al. Detection of unknown primary tumours in patients with cerebral metastases using whole-body 18Fflouorodeoxyglucose positron emission tomography. Eur J Neurology 2002; 9: 657662.
  5. 5. Stoeckli SJ, Mosna-Firlejczyk K, Goerres GW. Lymph node metastasis of squamous cell carcinoma from an unknown primary: impact of positron emission tomography. Eur J Nucl Med Mol Imaging 2000; 30: 411416.
  6. 6. Wong WL, Saunders M. The Impact of FDG PET on the management of occult primary head and neck tumours. Clin Oncol 2003; 15: 461466.

Address for correspondence:

Prof. Maciej Krzakowski

Centrum Onkologii — Instytut

im. Marii Skłodowskiej-Curie

Klinika Nowotworów Płuca

i Klatki Piersiowej

ul. Roentgena 5, 02–781 Warszawa

e-mail: maciekk@coi.waw.pl

EXPERTS` OPINION