Search

Recent Articles

IJMPO—A Journey of a Thousand Miles

Author : Padmaj S. Kulkarni

Coronavirus Disease 2019 Treatment—T-Cells Hold the Key in Severe Cases

Author : Kunal Das, Nitika Agrawal, Mansi Kala, Rakhee Khanduri

Why Is China Importing COVID-19 Vaccine Now?

Author : Purvish M. Parikh

Imaging Recommendations for Theranostic PET-CT in Oncology

CC BY 4.0 · Indian J Med Paediatr Oncol 2023; 44(03): 314-321

DOI: DOI: 10.1055/s-0042-1760310

Abstract

We in this article have presented a review of the guideline recommendations on theranostic positron emission tomography-computed tomography (PET-CT) imaging which will be helpful to assist practitioners in providing appropriate patient care. Multiple guidelines by different societies and medical associations provide standards for diagnosis, imaging, and treatment of cancer patients. They have generated a number of recommendations related to 68Ga-DOTATATE and 68Ga-PSMA-11 PET-CT, which are the classical examples of theranostic PET-CT imaging in current practice.

Authors' Contributions

Rahul V. Parghane was involved in conceptualization, designing, definition of intellectual content, literature search, manuscript preparation, manuscript editing, and manuscript review. Abhishek Mahajan contributed to conceptualization, designing, manuscript editing, and manuscript review. Nivedita Chakrabarty edited and reviewed the manuscript. Sandip Basu contributed to conceptualization, designing, definition of intellectual content, manuscript editing, and manuscript review.

Ethical Committee Clearance

Not required as patient data not revealed.

The manuscript has been read and approved by all the authors and the requirements for authorship have been met, and each author believes that the manuscript represents honest work.

Supplementary Material

Publication History

Article published online:
01 March 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

We recommend

  1. Emerging clinical applications of PET based molecular imaging in oncology: the promising future potential for evolving personalized cancer care
    Vandana K Dhingra et al., Indian J Radiol Imaging, 2015
  2. Whole-Body [18F]-FDG-PET/MRI for Oncology: A Consensus Recommendation
    Lale Umutlu et al., Zentralblatt für Chirurgie - Zeitschrift für Allgemeine, Viszeral-, Thorax- und Gefäßchirurgie, 2019
  3. Whole-Body [18F]-FDG-PET/MRI for Oncology: A Consensus Recommendation
    Lale Umutlu et al., Zentralblatt für Chirurgie - Zeitschrift für Allgemeine, Viszeral-, Thorax- und Gefäßchirurgie, 2019
  4. Imaging Recommendations for Diagnosis, Staging, and Management of Carcinoma of Unknown Origin (Lymph Node, Pulmonary, Liver, Skeletal, and Brain) with Emphasis on the Current Position of PET-CT in Carcinoma of Unknown Origin (CUP)
    Sarvesh Loharkar et al., Indian Journal of Medical and Paediatric Oncology
  5. FDG-PET/CT in oncology
    B. J. Krause et al., Nuklearmedizin, 2007
  1. Recent progress of nanotechnology-based theranostic systems in cancer treatments
    Ying Xue et al., Cancer Biology & Medicine, 2021
  2. Imaging in translational cancer research
    Felix T. Kurz et al., Cancer Biology & Medicine, 2022
  3. Evaluation of glial cell proliferation with non-invasive molecular imaging methods after stroke
    Ana Joya et al., Neural Regeneration Research, 2021
  4. 11C-PK11195 plasma metabolization has the same rate in multiple sclerosis patients and healthy controls: a cross-sectional study
    Aline Morais de Souza et al., Neural Regeneration Research, 2021
  5. Occult diaphramatic injury in penetrating trauma: a diagnostic challenge
    Sohil Pothiawala, Emergency and Critical Care Medicine, 2023

Abstract

We in this article have presented a review of the guideline recommendations on theranostic positron emission tomography-computed tomography (PET-CT) imaging which will be helpful to assist practitioners in providing appropriate patient care. Multiple guidelines by different societies and medical associations provide standards for diagnosis, imaging, and treatment of cancer patients. They have generated a number of recommendations related to 68Ga-DOTATATE and 68Ga-PSMA-11 PET-CT, which are the classical examples of theranostic PET-CT imaging in current practice.

Introduction

The term “theranostics” is fusion of two words diagnostics and therapeutics in which diagnostic and therapeutic tools related to the same specific molecular targets are coupled. In essence, theranostics integrates diagnostic modality for the detection of a molecular target for which a specific therapy is intended. Although the term theranostics is reportedly new and probably first used by John Funkhouser in 1998 for the development of a test for monitoring the efficacy of a new anticoagulant drug, the concept behind “theranostics” is not and had been applied to imaging and treatment of thyroid diseases for more than 80 years and revisited over the years.[1] [2]

In nuclear medicine, the theranostic system includes use of two identical or very closely related radiopharmaceuticals for diagnosis and therapeutic purpose. The tumor-specific substrates, receptor ligands, transporter, or cell surface proteins can serve as target for development of theranostic couples when labeled with specific radionuclides for imaging or therapy purpose as mentioned in [Table 1]. Theranostics has been used in nuclear medicine over past eight decades. Radioiodine is a prime example of a classic theranostic agent with use of same radioisotope 131I for same molecular target of sodium iodide symporter for imaging and therapeutics in patients with differentiated thyroid carcinoma.

Table 1

Theranostic pairs commonly used in clinical practice

Abbreviations: FAPI, fibroblast activation protein inhibitor; PSMA, prostate-specific membrane antigen; SSTR, somatostatin receptor.

Recent advancement in molecular biology and radiochemistry has led to introduction of newer theranostic agents for neuroendocrine tumors (NET) and prostate cancer (PC) in nuclear medicine. These agents used combination of two different radioisotopes (each one emitting different types of radiation: electromagnetic radiation for imaging and particulate irradiations for therapy) lined to the specific molecules that utilized same cellular structure, biologic process and also same target for imaging and therapy intent are called theranostic pairs as shown in [Table 1]. The main concept behind theranostics is “We treat what we see and we see what we treated.”[3] For this purpose, molecular imaging plays a cornerstone role for determining target lesions, quantification, and prognostication of disease process in individual patients. Over the past two decades, imaging technology evolved tremendously with emergence of hybrid imaging in nuclear medicine such as positron emission tomography-computed tomography (PET-CT) examinations. The hybrid PET-CT imaging is a double-edged sword with improved cutting-edge imaging technology on one side and cost and radiation exposure on another side. Hence, there is requirement for recommendation and appropriate use criteria on clinical use of hybrid PET-CT imaging and these are formulated by multidisciplinary panels and presented in various guidelines. We reviewed the guidelines for clinical use of theranostic PET-CT imaging in oncological conditions and compiled and presented in this article.

68Ga-labeled Somatostatin Analogs for PET-CT Imaging

Somatostatin is a naturally occurring hormone that acts by binding to somatostatin receptors (SSTR) and these receptors are over-expressed in most of NETs. SSTR is a target for therapy in NET over last three decades. In initial years of peptide receptor radionuclide therapy (PRRT) procedures, 111In-pentetreotide (imaging by using gamma camera based scintigraphy and therapy with help of Auger electrons) was used as theranostic agent in metastatic NET cases.[4] [5] [6] The emergence of hybrid PET-CT imaging and development of newer radiopharmaceuticals based on 68Ga-labeled somatostatin analog (68Ga-DOTATOC, 68Ga-DOTATATE, and 68Ga- DOTANOC) was a game changer and led to upgradation of imaging in NET,[7] as PET-CT imaging with 68Ga-labeled somatostatin analog has several advantages, including improved radiation dosimetry, measurement of lesional activity by using semiquantitative PET-based analysis, higher spatial resolution with better sensitivity and specificity compared to scintigraphy, and conventional imaging.[8] [9]

On June 1, 2016, U.S. Food and Drug Administration approved 68Ga-DOTATATE PET-CT imaging as a diagnostic tool for the detection of location and extent of tumor in NET patients.[10] This led to inclusion of 68Ga-labeled somatostatin analog PET-CT scans in many guidelines.[11] [12] [13]

European Association of Nuclear Medicine (EANM) Recommendations

Recently, the procedure guideline given by EANM on PET-CT study with 68Ga-labeled somatostatin analogs has been revised and updated.[14] They provided the clinical indication of 68Ga-labeled somatostatin analogue PET-CT imaging in NET as follows:

  • To detect and localize primary site (diagnosis of NET).

  • To determine extent of local and metastatic disease (staging of NET).

  • To detect residual, recurrent, or progressive disease (restaging of NET).

  • To determine SSTR status and select patients with metastatic disease for PRRT based upon SSTR status (management and prognosis of NET).

  • To determine therapy response (surgery, radiotherapy, chemotherapy or PRRT).

They recommended discontinuation of somatostatin analogue therapy prior to 68Ga-labeled somatostatin analog PET-CT scan for 1 day for short-lived molecules and 3 to 4 weeks for long-acting somatostatin analogue therapy to avoid possible SSTR blockade. They also recommended use of 68Ga-labeled somatostatin analog PET-CT scan for determining primary site in metastatic NET case of an unknown primary with no evidence of a primary disease on conventional imaging and cited use of 68Ga-labeled somatostatin analogue PET-CT scan for characterization of a bronchial mass suspicious of bronchial NET, when other diagnostic modalities are inconclusive.[15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29]

ESMO Recommendations

The ESMO provided guidelines for diagnosis, treatment, and follow-up of patients with gastroenteropancreatic NET. They recommended use of 68Ga-labeled somatostatin analog PET-CT scan for tumor staging, preoperative imaging, and restaging in NET patients. They also mentioned if PET not available, somatostatin receptor scintigraphy (SRS) may be used as less sensitive imaging modality as compared to PET-CT examination. They recommended the use of PRRT as second-line therapy in patients with midgut and pancreatic NET who fulfil general requirements for PRRT. One important prerequisite for PRRT is high-grade SSTR [removed]Krenning 3/4) in lesions, which is assessed by using 68Ga-labeled somatostatin analogue PET-CT scan before PRRT in these NET patients. They cited lifelong follow-up in treated NET patients, which included clinical symptom evaluation, biochemical parameters analysis, conventional, and SSTR imaging. Hence, they recommended use of 68Ga-labeled somatostatin analog PET-CT scan in follow-up of SSTR expressing NET patients at the interval of 12 to 36 months.[30]

European Neuroendocrine Tumor Society (ENETS) Recommendations

The European Neuroendocrine Tumor Society (ENETS) provided consensus guidelines on radiological, nuclear medicine, and hybrid imaging with standards of care in NET patients. Similar to ESMO guidelines, they also recommended use of 68Ga-labeled somatostatin analog PET-CT scan for tumor staging, preoperative imaging, and restaging in NET patients. They also mentioned high sensitivity of 68Ga-labeled somatostatin analog PET-CT imaging for the detection of lymph node metastases, bone metastases, liver metastases, small peritoneal lesions, and primary site of small-intestinal NET as compared with conventional imaging modalities.[31]

National Comprehensive Cancer Network

The National Comprehensive Cancer Network (NCCN) version 4.2021 to January 2022 mentioned appropriateness of SSTR imaging by using 68Ga-labeled somatostatin analogue PET-CT or PET- magnetic resonance imaging (MRI) for assessment of distant disease and SSTR status in NET patients. They mentioned that SSTR imaging is particularly important for evaluation of benefit from SSTR-directed therapies. They also mentioned that whenever possible PET-CT or PET-MRI should be performed in combination of contrast-enhanced CT or MRI imaging in order to minimize total numbers of imaging studies.[32]

North American Neuroendocrine Tumor Society and Other Societies

Representatives from various societies assembled under auspices of an autonomous workgroup to develop appropriate use criteria for SSTR-PET imaging (68Ga-DOTATOC and 68Ga-DOTATATE) in patients with well-differentiated NET (grade 1 and grade 2). They evaluated 12 clinical scenarios. Out of these, nine were recommended as appropriate use criteria:

  • Initial staging of NET after the histological confirmation

  • Detection of primary site in known metastatic NET

  • Selection NET patients for PRRT

  • Staging of NET prior to plan surgery

  • Evaluation of a mass suggestive of NET not amenable to endoscopic or percutaneous biopsy

  • Monitoring of NET

  • Evaluation of patients with biochemical evidence and symptoms of a NET without evidence of it on conventional imaging and or prior histological diagnosis

  • Restaging of NET patients with symptomatic or biochemical progressive disease but without progressive disease on conventional imaging

  • Conventional imaging showing new indeterminate lesion but having unclear progressive disease in NETs

They mentioned that SSTR-based PET demonstrated better sensitivity and specificity than conventional imaging and 111In-pentetreotide. They also cited that, SSTR-based PET is clearly preferred in initial diagnosis, selecting patients for PRRT, and localizing of unknown primaries in known metastatic NETs.[13]

Prostate-Specific Membrane Antigen PET-CT Imaging

The PC over-expresses the prostate-specific membrane antigen (PSMA), a transmembrane glycoprotein, present on cell membrane. Over-expression of PSMA is associated with higher grade tumor, metastatic castration-resistant tumor, and tumor aggressiveness in PC. The PSMA is an excellent target for theranostics in PC, as it is internalized after binding to the ligand, leading to high detection rate and better lesion to background uptake ratio for imaging purpose and inducing direct DNA damage with less risk of nonspecific radiation for therapeutic purpose. The 68Ga-PSMA-11, 68Ga-PSMA-617, and 68Ga-PSMA-I&T are developed as PET tracer in PC imaging. There is no data available for direct comparison of these different ligands. Most of published clinical work and clinical practices in nuclear medicine used 68Ga-PSMA-11 PET-CT imaging in PC and translated into 177Lu-PSMA-617 and 225Ac-PSMA-617 agents for therapeutic purposes.[33] [34] [35] [36] [37] [38] [39] [40] [41] [42] Various clinical guidelines given by the medical societies (namely urological, oncological, nuclear medicine, and radiation oncology societies) have recommended use of 68Ga-PSMA PET-CT imaging in PC.

Society of Nuclear Medicine and Molecular Imaging and European Association of Nuclear Medicine

The Society of Nuclear Medicine and Molecular Imaging and EANM jointly provided procedure guidelines[43] for the recommendation, performance, interpretation, and reporting of PSMA PET-CT imaging in PC as follows:

  • To detect tumor lesions in the presence of biochemical recurrence of PC (especially low serum prostate-specific antigen [PSA] values between 0.2 and 10 ng/mL).

  • For primary staging of PC with high-risk disease before surgical or external beam radiation planning.

  • For staging prior and during PSMA-based radionuclide therapy.

  • To do molecular targeted biopsy in high suspicion case of PC with prior negative biopsy.

  • To monitor response of systemic therapy in metastatic PC.

American Society of Clinical Oncology (ASCO) Recommendations:

The ASCO provided evidence and expert recommendations for optimal use of imaging in advanced PC. They mentioned use of either conventional imaging (defined as CT, bone scan, and prostate MRI) and/or next generation imaging (PET, PET-CT, PET-MRI, whole-body MRI) in advanced PC according to the clinical scenario. They recommended use of PET-CT for added clinical benefits and clarification in high-risk/very high-risk localized PC with negative or equivocal finding on conventional imaging. Another clinical scenario in which PSMA-based PET-CT imaging is recommended by ASCO is rising serum PSA level after prostatectomy or radiotherapy with negative conventional imaging in men for whom salvage local or regional therapy is contemplated. ASCO also recommended use of PET-CT imaging in hormone-sensitive metastatic PC at initial diagnosis or after initial treatment. For metastatic castration-resistant prostate cancer (CRPC) with serum PSA progression, they mentioned that use of next-generation imaging (PET, PET-CT, PET-MRI, whole-body MRI) for this cohort is unclear in view of a paucity of prospective data. However, they mentioned that use of next-generation imaging (PET, PET-CT, PET-MRI, whole-body MRI) could be contemplated, especially in the setting of a clinical trial.[44]

European Association of Urology, European Society of Urogenital Radiology, and Other Societies (The EAU-EANM-ESTRO-ESUR-ISUP-SIOG), in their guidelines on PC, recommended use of PSMA-based PET-CT imaging for staging in PC with high-risk localized disease/locally advanced disease and for the management of PC with persistent/recurrence serum PSA level after radical prostatectomy (serum PSA level > 0.2 ng/mL) or radiotherapy in which the result of PSMA PET-CT imaging will influence subsequent treatment decisions. This guideline also recommended use of 177Lu-PSMA-617 therapy in metastatic CRPC patients, which are showing high expression of PSMA (exceeding uptake in lesion over liver) on PSMA PET-CT scan.[45] [46]

NCCN Recommendations

The NCCN version 3.2022-January 2022 mentioned that 68Ga-PSMA-11 PET-CT imaging can be considered as an alternate to standard imaging of bone and soft tissue for initial staging, detection of lesions in biochemically recurrent disease, and as workup for progressive disease in bone and soft tissue. They cited that 68Ga-PSMA-11 PET-CT imaging has increased sensitivity and specificity for detecting micrometastatic disease compared to conventional imaging (CT, MRI) at initial staging and biochemical recurrence. Therefore, the panel of NCCN does not feel that conventional imaging is a necessary prerequisite to PSMA PET-CT. To reduce false-positive rate of PSMA PET-CT in PC, use of radiographic or histological confirmation is recommended by NCCN.[47]

68Ga-Fibroblast Activation Protein Inhibitors PET-CT Imaging

The fibroblast activation protein (FAP) is a serine proteinase and highly expressed on cell surface of activated fibroblasts but absent in resting fibroblasts. Over-expression of FAP is related in wound healing, fibrotic processes, and stroma of many malignancies. The FAP-associated fibroblasts are found in more than 90%-of epithelial tumors on histopathological studies. The cancer-associated fibroblasts with extracellular fibrosis contributed up to 90%-of gross tumor mass, leaving original tumor cells in minority. Hence, FAP is a potential target for theranostic in a large variety of cancers.[48]

The University Hospital Heidelberg group recently developed a series of quinoline-based FAP inhibitors (FAPI) based on clinical and preclinical research.[49] They labeled these FAPI compounds with diagnostic and therapeutic radioisotopes via chelator DOTA. For diagnostic purposes, they labeled FAPI-01, FAPI-02, FAPI-04, FAPI-21, and FAPI-46 with 68Ga. Kratochwil et al demonstrated that 68Ga-FAPI-04 PET-CT imaging is capable to visualize tumor lesions in 28 different types of cancer with high sensitivity and image quality. They cited that 68Ga-FAPI-04 PET-CT imaging showed overall intense tracer uptake with high-contrast images in various types of cancers, such as sarcomas, cholangio-carcinoma, esophageal, breast, lung, hepatocellular, colorectal, head-neck, ovarian, pancreatic, and PCs, etc., and use of 68Ga-FAPI-04 PET-CT imaging led to non-invasive tumor characterization, tumor staging, and radio-ligand therapy pre-evaluation for theranostic purpose.[50]

The 90Y, 177Lu, or 153Sm-labeled FAPI-derivatives (FAPI-46, DOTA.SA.FAPI, and FAPI-04) were used as therapeutic agents on compassionate ground in treating breast cancer, sarcoma, and pancreatic cancer patients in various case reports and a few studies. Case report and studies cited that FAPI-derivatives therapies were well tolerated and showed signs of clinical responses, such as stable disease or tumor lesion shrinkage as well as reduction in clinical symptoms.[51] [52] [53]

Guideline for FAPI PET-CT Imaging

There is paucity of FAPI PET-CT imaging data in literature. Hence, role of FAPI PET-CT scan in cancer imaging and management required further exploration in larger prospective trials before coming to any conclusion on clinical use of these imaging agents. Radiolabeled FAPI as theranostic agents is under development and still in its infancy but showed promising results in preliminary reports and expected to enter clinical trials in near future. Therefore, at present no guidelines on FAPI PET-CT imaging exist in the literature.

Advantages of Theranostic PET over Conventional Imaging in PC and NET

[Supplementary Table S1] (in supplement) shows the comparison of CECT, FDG-PET-CT, MRI, and PSMA PET-CT for PC.[54] [55] [56] [57] [58] [59] [60] As shown in [Table 1], PSMA PET-CT is superior to the conventional imaging modalities for the localization of primary lesion, staging, and detection of relapse. Besides, PSMA PET-CT also gives lower radiation exposure when compared to CECT and FDG-PET-CT. [Supplementary Table S2] (in supplement) shows the comparison of CECT, FDG-PET-CT, MRI, and 68Ga- DOTATOC PET-CT for NET.[61] [62] [63] [64] [65] Similarly, 68Ga-DOTATOC PET-CT appears to be better for the localization of primary lesion, staging, and response evaluation. Also, radiation exposure with 68Ga-DOTATOC PET-CT is lower than CECT and FDG-PET-CT.

Indian Experience on 68Ga-DOTA-NOC/TATE and 68Ga-PSMA PET-CT

There has been substantial use of both 68Ga-labeled somatostatin analogs and 68Ga-PSMA PET-CT imaging for varied applications in a wide variety of tumors, though no Indian guideline exists on the clinical use of these diagnostic modalities. Naswa et al conducted a prospective study on comparison of 68Ga-DOTANOC PET-CT and conventional imaging in NETs. They found superior sensitivity and specificity of 68Ga-DOTANOC PET-CT imaging over conventional imaging in detecting primary site and metastatic disease, with significant change in the management of NETs after use of 68Ga-DOTANOC PET-CT imaging.[66] In our experience, 68Ga-labeled somatostatin analogs PET-CT have outperformed over conventional imaging both for staging and restaging of NETs.[67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] Sampathirao and Basu studied 51 patients with CUP-NETs with 68Ga-DOTATATE PET-CT: unknown primary was detected in 31 of 51 patients (resulting in sensitivity of 60.78%), while overall lesion detection sensitivity was 96.87%. Tumor heterogeneity exists in NET, hence 68Ga-labeled somatostatin analogs PET-CT in combination of 18F-FDG-PET-CT (dual tracer PET-CT) have evolved as an important functional imaging approach over Ki-67 labelling index for deciding treatment strategies in metastatic and inoperable NETs (PRRT vs. chemo-PRRT). In neoadjuvant setting, cross-sectional imaging modality, such as triphasic CECT, is superior for identifying involvement of major abdominal blood vessels by tumor over noncontrast PET-CT imaging. The whole body 68Ga-labeled somatostatin receptor-based PET-CT is more valuable imaging modality over conventional imaging for response evaluation and surveillance in NETs. Theranostic 68Ga-labeled somatostatin analogs PET-CT is an excellent imaging modality for deciding PRRT in clinical conditions beyond NETs such as metastatic/inoperable medullary thyroid carcinoma, metastatic/inoperable paraganglioma/pheochromocytoma, noniodine concentrating metastatic differentiated thyroid carcinoma, Merkel cell carcinoma, meningioma, and recurrent/inoperable phosphaturic mesenchymal tumor.[67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] Jain et al prospectively evaluated diagnostic accuracy of 68Ga-PSMA PET-CT imaging in PC. They found that 68Ga-PSMA PET-CT imaging significantly improved detection rate of PC by using a SUVmax cutoff value in patients with raised PSA between 4 and 20 ng/mL or abnormal digital rectal examination findings.[78] Kallur et al evaluated the clinical utility of 68Ga-PSMA PET-CT imaging in 262 patients of PC.[79] In our experience, 68Ga-PSMA PET-CT imaging is better imaging modality in detection of primary, staging, restaging, response evaluation, and prognostication of PC patients.[79] [80]

Theranostic 68Ga-Labeled SSTR-Based PET-CT imaging in Pediatric Population

At present no guideline exists for 68Ga-labeled somatostatin analogs PET-CT imaging in pediatric group. As such, available literature for clinical use of theranostic 68Ga-labeled somatostatin analogs PET-CT imaging in pediatric group is limited in view of low prevalence and rare incidence of NETs in this group. Most studies suggest 68Ga-labeled somatostatin analogs PET-CT imaging should be considered as first-line imaging modality in pediatric NETs. Goel et al evaluated role of 68Ga-DOTATATE PET-CT in 30 pediatric NET patients and they found that 68Ga-DOTATATE PET-CT was superior imaging technique over conventional imaging for detecting bone metastases.[81] Jha et al found that better lesion detection rate of 68Ga-DOTATATE PET-CT in pediatric group of pheochromocytomas.[82] Well-differentiated neuroblastoma has high expression of SSTR2, resulting in better sensitivity of 68Ga-labeled somatostatin analogs for lesion detection as compared to 123I-MIBG imaging. But still there are no current guidelines on 68Ga-labeled somatostatin-based PET-CT imaging in view of scarcity of data in pediatric population. Overall, 68Ga-labeled somatostatin analogue PET-CT imaging in pediatric group showed number of advantages over conventional imaging such as low radiation exposure, fast clearance time, simple preparation, few pharmacological interactions, higher prognostic value, and identifying individuals for PRRT.[83]

Conclusion

Theranostic approach in oncological condition requires integration of therapeutic and diagnostic modality for targeting specific molecule, which can be labeled with radiopharmaceuticals for imaging or therapeutic purpose. Theranostic PET-CT imaging is helpful for diagnosis, finding status of target molecule in the tumor cells, and determining extension of disease that helps in clinical decision-making and design of effective treatment plan. Other advantages of theranostic PET-CT imaging include assessment of biologic behavior and tumoral heterogeneity, prognostification of disease, and predicting response and toxicities of therapies. This guideline review on theranostic PET-CT imaging thus summarizes the perspectives expressed in multiple guidelines on various receptor-based PET-CT imaging by different societies, discussing upon their clinical use, in order to achieve the goal of best management of cancer patients with reduced expenditure and avoiding potentially unnecessary treatments and interventions.

Conflict of Interest

None declared.

Authors' Contributions

Rahul V. Parghane was involved in conceptualization, designing, definition of intellectual content, literature search, manuscript preparation, manuscript editing, and manuscript review. Abhishek Mahajan contributed to conceptualization, designing, manuscript editing, and manuscript review. Nivedita Chakrabarty edited and reviewed the manuscript. Sandip Basu contributed to conceptualization, designing, definition of intellectual content, manuscript editing, and manuscript review.

Ethical Committee Clearance

Not required as patient data not revealed.

The manuscript has been read and approved by all the authors and the requirements for authorship have been met, and each author believes that the manuscript represents honest work.

Diagnostic agents

Therapeutic agents

Molecular/Cellular target

Function of target

Oncological conditions

68Ga-DOTATOC

68Ga-DOTATATE

68Ga- DOTANOC

177Lu-DOTATATE

90Y-DOTATATE

225Ac-DOTATATE

SSTR

Cell-surface receptor

Well-differentiated neuroendocrine tumors

68Ga-PSMA-11

68Ga-PSMA-617 68Ga-PSMA-I&T

177Lu-PSMA-617

225Ac-PSMA-617

PSMA

Cell-surface protein

Metastatic castration-resistant prostate cancer

68Ga-FAPI-04 and FAPI-derivatives

90Y, 177Lu or

153Sm-labeled

FAPI-derivatives

FAP

Stroma of cancer-cell surface of activated fibroblasts

Various types of cancers-sarcomas, breast esophageal, lung, pancreatic, prostate cancers, etc.

Supplementary Material

Reference

  1.  Kelkar SS, Reineke TM. Theranostics: combining imaging and therapy. Bioconjug Chem 2011; 22 (10) 1879-1903
  2.  DeNardo GL, DeNardo SJ. Concepts, consequences, and implications of theranosis. Semin Nucl Med 2012; 42 (03) 147-150
  3.  Turner JH. Recent advances in theranostics and challenges for the future. Br J Radiol 2018; 91 (1091): 20170893
  4.  Modlin IM, Oberg K, Chung DC. et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol 2008; 9 (01) 61-72
  5.  Krenning EP, Kwekkeboom DJ, Bakker WH. et al. Somatostatin receptor scintigraphy with [111In-DTPA-D-Phe1]- and [123I-Tyr3]-octreotide: the Rotterdam experience with more than 1000 patients. Eur J Nucl Med 1993; 20 (08) 716-731
  6.  Krenning EP, Bakker WH, Breeman WA. et al. Localisation of endocrine-related tumours with radioiodinated analogue of somatostatin. Lancet 1989; 1 (8632): 242-244
  7.  Hofmann M, Maecke H, Börner R. et al. Biokinetics and imaging with the somatostatin receptor PET radioligand (68)Ga-DOTATOC: preliminary data. Eur J Nucl Med 2001; 28 (12) 1751-1757
  8.  Singh S, Poon R, Wong R, Metser U. 68Ga PET imaging in patients with neuroendocrine tumors: a systematic review and meta-analysis. Clin Nucl Med 2018; 43 (11) 802-810
  9.  Levine R, Krenning EP. Clinical history of the theranostic radionuclide approach to neuroendocrine tumors and other types of cancer: historical review based on an interview of Eric P. Krenning by Rachel Levine. J Nucl Med 2017; 58 (Suppl 2): 3S-9S
  10.  Raj N, Reidy-Lagunes D. The Role of 68Ga-DOTATATE positron emission tomography/computed tomography in well-differentiated neuroendocrine tumors: a case-based approach illustrates potential benefits and challenges. Pancreas 2018; 47 (01) 1-5
  11.  Öberg K, Knigge U, Kwekkeboom D, Perren A. ESMO Guidelines Working Group. Neuroendocrine gastro-enteropancreatic tumors: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2012; 23 (suppl7): 124-130
  12.  Shah MH, Goldner WS, Halfdanarson TR. et al. NCCN guidelines insights: neuroendocrine and adrenal tumors, Version 2.2018. J Natl Compr Canc Netw 2018; 16 (06) 693-702
  13.  Hope TA, Bergsland EK, Bozkurt MF. et al. Appropriate use criteria for somatostatin receptor PET imaging in neuroendocrine tumors. J Nucl Med 2018; 59 (01) 66-74
  14.  Bozkurt MF, Virgolini I, Balogova S. et al. Guideline for PET/CT imaging of neuroendocrine neoplasms with 68Ga-DOTA-conjugated somatostatin receptor targeting peptides and 18F-DOPA. Eur J Nucl Med Mol Imaging 2017; 44 (09) 1588-1601
  15.  Campana D, Ambrosini V, Pezzilli R. et al. Standardized uptake values of (68)Ga-DOTANOC PET: a promising prognostic tool in neuroendocrine tumors. J Nucl Med 2010; 51 (03) 353-359
  16.  Conry BG, Papathanasiou ND, Prakash V. et al. Comparison of (68)Ga-DOTATATE and (18)F-fluorodeoxyglucose PET/CT in the detection of recurrent medullary thyroid carcinoma. Eur J Nucl Med Mol Imaging 2010; 37 (01) 49-57
  17.  Kayani I, Bomanji JB, Groves A. et al. Functional imaging of neuroendocrine tumors with combined PET/CT using 68Ga-DOTATATE (DOTA-DPhe1,Tyr3-octreotate) and 18F-FDG. Cancer 2008; 112 (11) 2447-2455
  18.  Ambrosini V, Tomassetti P, Castellucci P. et al. Comparison between 68Ga-DOTA-NOC and 18F-DOPA PET for the detection of gastro-entero-pancreatic and lung neuro-endocrine tumours. Eur J Nucl Med Mol Imaging 2008; 35 (08) 1431-1438
  19.  Fanti S, Ambrosini V, Tomassetti P. et al. Evaluation of unusual neuroendocrine tumours by means of 68Ga-DOTA-NOC PET. Biomed Pharmacother 2008; 62 (10) 667-671
  20.  Kayani I, Conry BG, Groves AM. et al. A comparison of 68Ga-DOTATATE and 18F-FDG PET/CT in pulmonary neuroendocrine tumors. J Nucl Med 2009; 50 (12) 1927-1932
  21.  Ambrosini V, Castellucci P, Rubello D. et al. 68Ga-DOTA-NOC: a new PET tracer for evaluating patients with bronchial carcinoid. Nucl Med Commun 2009; 30 (04) 281-286
  22.  Schartinger VH, Dudás J, Decristoforo C. et al. 68Ga-DOTA0-Tyr3-octreotide positron emission tomography in head and neck squamous cell carcinoma. Eur J Nucl Med Mol Imaging 2013; 40 (09) 1365-1372
  23.  Kauhanen S, Seppänen M, Minn H. et al. Fluorine-18-L-dihydroxyphenylalanine (18F-DOPA) positron emission tomography as a tool to localize an insulinoma or beta-cell hyperplasia in adult patients. J Clin Endocrinol Metab 2007; 92 (04) 1237-1244
  24.  Traub-Weidinger T, Putzer D, von Guggenberg E. et al. Multiparametric PET imaging in thyroid malignancy characterizing tumour heterogeneity: somatostatin receptors and glucose metabolism. Eur J Nucl Med Mol Imaging 2015; 42 (13) 1995-2001
  25.  Prasad V, Ambrosini V, Hommann M, Hoersch D, Fanti S, Baum RP. Detection of unknown primary neuroendocrine tumours (CUP-NET) using (68)Ga-DOTA-NOC receptor PET/CT. Eur J Nucl Med Mol Imaging 2010; 37 (01) 67-77
  26.  Putzer D, Gabriel M, Henninger B. et al. Bone metastases in patients with neuroendocrine tumor: 68Ga-DOTA-Tyr3-octreotide PET in comparison to CT and bone scintigraphy. J Nucl Med 2009; 50 (08) 1214-1221
  27.  Ambrosini V, Nanni C, Zompatori M. et al. (68)Ga-DOTA-NOC PET/CT in comparison with CT for the detection of bone metastasis in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2010; 37 (04) 722-727
  28.  Ugur O, Kothari PJ, Finn RD. et al. Ga-66 labeled somatostatin analogue DOTA-DPhe1-Tyr3-octreotide as a potential agent for positron emission tomography imaging and receptor mediated internal radiotherapy of somatostatin receptor positive tumors. Nucl Med Biol 2002; 29 (02) 147-157
  29.  Gabriel M, Oberauer A, Dobrozemsky G. et al. 68Ga-DOTA-Tyr3-octreotide PET for assessing response to somatostatin-receptor-mediated radionuclide therapy. J Nucl Med 2009; 50 (09) 1427-1434
  30.  Pavel M, Öberg K, Falconi M. et al; ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Gastroenteropancreatic neuroendocrine neoplasms: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020; 31 (07) 844-860
  31.  Sundin A, Arnold R, Baudin E. et al; Antibes Consensus Conference participants. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: radiological, nuclear medicine & hybrid imaging. Neuroendocrinology 2017; 105 (03) 212-244
  32.  Shah MH, Goldner WS, Benson AB. et al. Neuroendocrine and adrenal tumors, Version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2021; 19 (07) 839-868
  33.  Barbosa FG, Queiroz MA, Nunes RF. et al. Revisiting prostate cancer recurrence with PSMA PET: atlas of typical and atypical patterns of spread. Radiographics 2019; 39 (01) 186-212
  34.  Scher HI, Fizazi K, Saad F. et al; AFFIRM Investigators. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012; 367 (13) 1187-1197
  35.  Kratochwil C, Giesel FL, Eder M. et al. [177Lu]Lutetium-labelled PSMA ligand-induced remission in a patient with metastatic prostate cancer. Eur J Nucl Med Mol Imaging 2015; 42 (06) 987-988
  36.  Afshar-Oromieh A, Avtzi E, Giesel FL. et al. The diagnostic value of PET/CT imaging with the (68)Ga-labelled PSMA ligand HBED-CC in the diagnosis of recurrent prostate cancer. Eur J Nucl Med Mol Imaging 2015; 42 (02) 197-209
  37.  Hofman MS, Hicks RJ, Maurer T, Eiber M. Prostate-specific membrane antigen PET: clinical utility in prostate cancer, normal patterns, pearls, and pitfalls. Radiographics 2018; 38 (01) 200-217
  38.  Eiber M, Maurer T, Souvatzoglou M. et al. Evaluation of hybrid 68Ga-PSMA ligand PET/CT in 248 patients with biochemical recurrence after radical prostatectomy. J Nucl Med 2015; 56 (05) 668-674
  39.  Rauscher I, Maurer T, Fendler WP, Sommer WH, Schwaiger M, Eiber M. (68)Ga-PSMA ligand PET/CT in patients with prostate cancer: how we review and report. Cancer Imaging 2016; 16 (01) 14
  40.  Ballas LK, de Castro Abreu AL, Quinn DI. What medical, urologic, and radiation oncologists want from molecular imaging of prostate cancer. J Nucl Med 2016; 57 (Suppl 3): 6S-12S
  41.  Fendler WP, Rahbar K, Herrmann K, Kratochwil C, Eiber M. 177Lu-PSMA radioligand therapy for prostate cancer. J Nucl Med 2017; 58 (08) 1196-1200
  42.  Emmett L, Willowson K, Violet J, Shin J, Blanksby A, Lee J. Lutetium 177 PSMA radionuclide therapy for men with prostate cancer: a review of the current literature and discussion of practical aspects of therapy. J Med Radiat Sci 2017; 64 (01) 52-60
  43.  Fendler WP, Eiber M, Beheshti M. et al. 68Ga-PSMA PET/CT: joint EANM and SNMMI procedure guideline for prostate cancer imaging: version 1.0. Eur J Nucl Med Mol Imaging 2017; 44 (06) 1014-1024
  44.  Trabulsi EJ, Rumble RB, Jadvar H. et al. Optimum imaging strategies for advanced prostate cancer: ASCO guideline. J Clin Oncol 2020; 38 (17) 1963-1996
  45.  Mottet N, van den Bergh RCN, Briers E. et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer-2020 Update. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol 2021; 79 (02) 243-262
  46.  Cornford P, van den Bergh RCN, Briers E. et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer. Part II-2020 update: treatment of relapsing and metastatic prostate cancer. Eur Urol 2021; 79 (02) 263-282
  47.  NCCN CLINICAL PRACTICE GUIDELINES IN ONCOLOGY. Prostate cancer version 3.2022-January 2022. Accessed December 12, 2022, at: https://www.nccn.org/login?ReturnURL=https://www.nccn.org/professionals/physician_gls/pdf/prostate.pdf
  48.  Jiang GM, Xu W, Du J. et al. The application of the fibroblast activation protein α-targeted immunotherapy strategy. Oncotarget 2016; 7 (22) 33472-33482
  49.  Lindner T, Loktev A, Altmann A. et al. Development of quinoline-based theranostic ligands for the targeting of fibroblast activation protein. J Nucl Med 2018; 59 (09) 1415-1422
  50.  Kratochwil C, Flechsig P, Lindner T. et al. 68Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med 2019; 60 (06) 801-805
  51.  Ballal S, Yadav MP, Kramer V. et al. A theranostic approach of [68Ga]Ga-DOTA.SA.FAPi PET/CT-guided [177Lu]Lu-DOTA.SA.FAPi radionuclide therapy in an end-stage breast cancer patient: new frontier in targeted radionuclide therapy. Eur J Nucl Med Mol Imaging 2021; 48 (03) 942-944
  52.  Ferdinandus J, Costa PF, Kessler L. et al. Initial clinical experience with 90Y-FAPI-46 radioligand therapy for advanced stage solid tumors: a case series of nine patients. J Nucl Med 2022; 63 (05) 727-734
  53.  Kratochwil C, Giesel FL, Rathke H. et al. [153Sm]Samarium-labeled FAPI-46 radioligand therapy in a patient with lung metastases of a sarcoma. Eur J Nucl Med Mol Imaging 2021; 48 (09) 3011-3013
  54.  Regmi SK, Sathianathen N, Stout TE, Konety BR. MRI/PET Imaging in elevated PSA and localized prostate cancer: a narrative review. Transl Androl Urol 2021; 10 (07) 3117-3129
  55.  Liu F, Dong J, Shen Y. et al. Comparison of PET/CT and MRI in the diagnosis of bone metastasis in prostate cancer patients: a network analysis of diagnostic studies. Front Oncol 2021; 11: 736654
  56.  Kichloo A, Amir R, Aljadah M. et al. FDG-PET versus PSMA-PET: a patient with prostate cancer. J Investig Med High Impact Case Rep 2020; 8: 2324709620941313
  57.  Tsechelidis I, Vrachimis A. PSMA PET in Imaging Prostate Cancer. Front Oncol 2022; 12: 831429
  58.  Smith-Bindman R, Lipson J, Marcus R. et al. Radiation dose associated with common computed tomography examinations and the associated lifetime attributable risk of cancer. Arch Intern Med 2009; 169 (22) 2078-2086
  59.  Kwon HW, Kim JP, Lee HJ. et al. Radiation dose from whole-body F-18 fluorodeoxyglucose positron emission tomography/computed tomography: Nationwide Survey in Korea. J Korean Med Sci 2016; 31 Suppl 1 (Suppl 1): S69-S74
  60.  Waller J, Flavell R, Heath CL. High accuracy of PSMA PET in initial staging of high-risk prostate cancer. Radiol Imaging Cancer 2020; 2 (04) e204025
  61.  Maxwell JE, Howe JR. Imaging in neuroendocrine tumors: an update for the clinician. Int J Endocr Oncol 2015; 2 (02) 159-168
  62.  Gamal GH. The utility of 18F-FDG PET/CT in the diagnosis, staging of non-functioning pancreatic neuroendocrine tumors. Egypt J Radiol Nucl Med 2021; 52 (01) 234
  63.  Schraml C, Schwenzer NF, Sperling O. et al. Staging of neuroendocrine tumours: comparison of [68Ga]DOTATOC multiphase PET/CT and whole-body MRI. Cancer Imaging 2013; 13 (01) 63-72
  64.  Garcia-Carbonero R, Garcia-Figueiras R, Carmona-Bayonas A. et al; Spanish Cooperative Group of Neuroendocrine Tumors (GETNE). Imaging approaches to assess the therapeutic response of gastroenteropancreatic neuroendocrine tumors (GEP-NETs): current perspectives and future trends of an exciting field in development. Cancer Metastasis Rev 2015; 34 (04) 823-842
  65.  Accessed December 12, 2022, at: https://clinicaltrials.gov/ProvidedDocs/28/NCT03136328/Prot_SAP_000.pdf
  66.  Naswa N, Sharma P, Kumar A. et al. Gallium-68-DOTA-NOC PET/CT of patients with gastroenteropancreatic neuroendocrine tumors: a prospective single-center study. AJR Am J Roentgenol 2011; 197 (05) 1221-1228
  67.  Thapa P, Ranade R, Ostwal V, Shrikhande SV, Goel M, Basu S. Performance of 177Lu-DOTATATE-based peptide receptor radionuclide therapy in metastatic gastroenteropancreatic neuroendocrine tumor: a multiparametric response evaluation correlating with primary tumor site, tumor proliferation index, and dual tracer imaging characteristics. Nucl Med Commun 2016; 37 (10) 1030-1037
  68.  Sampathirao N, Basu S. MIB-1 index-stratified assessment of dual-tracer PET/CT with 68Ga-DOTATATE and 18F-FDG and multimodality anatomic imaging in metastatic neuroendocrine tumors of unknown primary in a PRRT workup setting. J Nucl Med Technol 2017; 45 (01) 34-41
  69.  Basu S, Sirohi B, Shrikhande SV. Dual tracer imaging approach in assessing tumor biology and heterogeneity in neuroendocrine tumors: its correlation with tumor proliferation index and possible multifaceted implications for personalized clinical management decisions, with focus on PRRT. Eur J Nucl Med Mol Imaging 2014; 41 (08) 1492-1496
  70.  Basu S, Chakraborty S, Parghane RV. et al. One decade of ‘Bench-to-Bedside’ peptide receptor radionuclide therapy with indigenous [177Lu]Lu-DOTATATE obtained through ‘Direct’ neutron activation route: lessons learnt including practice evolution in an Indian setting. Am J Nucl Med Mol Imaging 2020; 10 (04) 178-211
  71.  Basu S, Parghane RV, Kamaldeep, Chakrabarty S. Peptide receptor radionuclide therapy of neuroendocrine tumors. Semin Nucl Med 2020; 50 (05) 447-464
  72.  Parghane RV, Talole S, Basu S. Prevalence of hitherto unknown brain meningioma detected on 68Ga-DOTATATE positron-emission tomography/computed tomography in patients with metastatic neuroendocrine tumor and exploring potential of 177Lu-DOTATATE peptide receptor radionuclide therapy as single-shot treatment approach targeting both tumors. World J Nucl Med 2019; 18 (02) 160-170
  73.  Parghane RV, Talole S, Basu S. 131I-MIBG negative progressive symptomatic metastatic paraganglioma: response and outcome with 177Lu-DOTATATE peptide receptor radionuclide therapy. Ann Nucl Med 2021; 35 (01) 92-101
  74.  Parghane RV, Ostwal V, Ramaswamy A. et al. Long-term outcome of “Sandwich” chemo-PRRT: a novel treatment strategy for metastatic neuroendocrine tumors with both FDG- and SSTR-avid aggressive disease. Eur J Nucl Med Mol Imaging 2021; 48 (03) 913-923
  75.  Parghane RV, Naik C, Talole S. et al. Clinical utility of 177 Lu-DOTATATE PRRT in somatostatin receptor-positive metastatic medullary carcinoma of thyroid patients with assessment of efficacy, survival analysis, prognostic variables, and toxicity. Head Neck 2020; 42 (03) 401-416
  76.  Basu S, Parghane RV, Naik C. Clinical efficacy of 177Lu-DOTATATE peptide receptor radionuclide therapy in thyroglobulin-elevated negative iodine scintigraphy: a “not-so-promising” result compared to GEP-NETs. World J Nucl Med 2020; 19 (03) 205-210
  77.  Parghane RV, Bhandare M, Chaudhari V. et al. Surgical feasibility, determinants, and overall efficacy of neoadjuvant 177Lu-DOTATATE PRRT for locally advanced unresectable gastroenteropancreatic neuroendocrine tumors. J Nucl Med 2021; 62 (11) 1558-1563
  78.  Jain H, Sood R, Faridi MS, Goel H, Sharma U. Role of 68Ga-PSMA-PET/CT for the detection of primary prostate cancer prior to biopsy: a prospective study. Cent European J Urol 2021; 74 (03) 315-320
  79.  Kallur KG, Ramachandra PG, Rajkumar K. et al. Clinical utility of Gallium-68 PSMA PET/CT scan for prostate cancer. Indian J Nucl Med 2017; 32 (02) 110-117
  80.  Suman S, Parghane RV, Joshi A. et al. Therapeutic efficacy, prognostic variables and clinical outcome of 177Lu-PSMA-617 PRLT in progressive mCRPC following multiple lines of treatment: prognostic implications of high FDG uptake on dual tracer PET-CT vis-à-vis Gleason score in such cohort. Br J Radiol 2019; 92 (1104): 20190380
  81.  Goel R, Shukla J, Bansal D. et al. (68)Ga-DOTATATE positron emission tomography/computed tomography scan in the detection of bone metastases in pediatric neuroendocrine tumors. Indian J Nucl Med 2014; 29 (01) 13-17
  82.  Jha A, Ling A, Millo C. et al. Superiority of 68Ga-DOTATATE over 18F-FDG and anatomic imaging in the detection of succinate dehydrogenase mutation (SDHx )-related pheochromocytoma and paraganglioma in the pediatric population. Eur J Nucl Med Mol Imaging 2018; 45 (05) 787-797
  83.  Maaz AUR, O'Doherty J, Djekidel M. 68Ga-DOTATATE PET/CT for neuroblastoma staging: utility for clinical use. J Nucl Med Technol 2021; 49 (03) 265-268

Address for correspondence

Sandip Basu, MBBS, DRM, DNB, MNAMS
Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe
Jerbai Wadia Road, Parel, Mumbai, Maharashtra, 400 012
India   

Publication History

Article published online:
01 March 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

We recommend

  1. Emerging clinical applications of PET based molecular imaging in oncology: the promising future potential for evolving personalized cancer care
    Vandana K Dhingra et al., Indian J Radiol Imaging, 2015
  2. Whole-Body [18F]-FDG-PET/MRI for Oncology: A Consensus Recommendation
    Lale Umutlu et al., Zentralblatt für Chirurgie - Zeitschrift für Allgemeine, Viszeral-, Thorax- und Gefäßchirurgie, 2019
  3. Whole-Body [18F]-FDG-PET/MRI for Oncology: A Consensus Recommendation
    Lale Umutlu et al., Zentralblatt für Chirurgie - Zeitschrift für Allgemeine, Viszeral-, Thorax- und Gefäßchirurgie, 2019
  4. Imaging Recommendations for Diagnosis, Staging, and Management of Carcinoma of Unknown Origin (Lymph Node, Pulmonary, Liver, Skeletal, and Brain) with Emphasis on the Current Position of PET-CT in Carcinoma of Unknown Origin (CUP)
    Sarvesh Loharkar et al., Indian Journal of Medical and Paediatric Oncology
  5. FDG-PET/CT in oncology
    B. J. Krause et al., Nuklearmedizin, 2007

Reference

  1.  Kelkar SS, Reineke TM. Theranostics: combining imaging and therapy. Bioconjug Chem 2011; 22 (10) 1879-1903
  2.  DeNardo GL, DeNardo SJ. Concepts, consequences, and implications of theranosis. Semin Nucl Med 2012; 42 (03) 147-150
  3.  Turner JH. Recent advances in theranostics and challenges for the future. Br J Radiol 2018; 91 (1091): 20170893
  4.  Modlin IM, Oberg K, Chung DC. et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol 2008; 9 (01) 61-72
  5.  Krenning EP, Kwekkeboom DJ, Bakker WH. et al. Somatostatin receptor scintigraphy with [111In-DTPA-D-Phe1]- and [123I-Tyr3]-octreotide: the Rotterdam experience with more than 1000 patients. Eur J Nucl Med 1993; 20 (08) 716-731
  6.  Krenning EP, Bakker WH, Breeman WA. et al. Localisation of endocrine-related tumours with radioiodinated analogue of somatostatin. Lancet 1989; 1 (8632): 242-244
  7.  Hofmann M, Maecke H, Börner R. et al. Biokinetics and imaging with the somatostatin receptor PET radioligand (68)Ga-DOTATOC: preliminary data. Eur J Nucl Med 2001; 28 (12) 1751-1757
  8.  Singh S, Poon R, Wong R, Metser U. 68Ga PET imaging in patients with neuroendocrine tumors: a systematic review and meta-analysis. Clin Nucl Med 2018; 43 (11) 802-810
  9.  Levine R, Krenning EP. Clinical history of the theranostic radionuclide approach to neuroendocrine tumors and other types of cancer: historical review based on an interview of Eric P. Krenning by Rachel Levine. J Nucl Med 2017; 58 (Suppl 2): 3S-9S
  10.  Raj N, Reidy-Lagunes D. The Role of 68Ga-DOTATATE positron emission tomography/computed tomography in well-differentiated neuroendocrine tumors: a case-based approach illustrates potential benefits and challenges. Pancreas 2018; 47 (01) 1-5
  11.  Öberg K, Knigge U, Kwekkeboom D, Perren A. ESMO Guidelines Working Group. Neuroendocrine gastro-enteropancreatic tumors: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2012; 23 (suppl7): 124-130
  12.  Shah MH, Goldner WS, Halfdanarson TR. et al. NCCN guidelines insights: neuroendocrine and adrenal tumors, Version 2.2018. J Natl Compr Canc Netw 2018; 16 (06) 693-702
  13.  Hope TA, Bergsland EK, Bozkurt MF. et al. Appropriate use criteria for somatostatin receptor PET imaging in neuroendocrine tumors. J Nucl Med 2018; 59 (01) 66-74
  14.  Bozkurt MF, Virgolini I, Balogova S. et al. Guideline for PET/CT imaging of neuroendocrine neoplasms with 68Ga-DOTA-conjugated somatostatin receptor targeting peptides and 18F-DOPA. Eur J Nucl Med Mol Imaging 2017; 44 (09) 1588-1601
  15.  Campana D, Ambrosini V, Pezzilli R. et al. Standardized uptake values of (68)Ga-DOTANOC PET: a promising prognostic tool in neuroendocrine tumors. J Nucl Med 2010; 51 (03) 353-359
  16.  Conry BG, Papathanasiou ND, Prakash V. et al. Comparison of (68)Ga-DOTATATE and (18)F-fluorodeoxyglucose PET/CT in the detection of recurrent medullary thyroid carcinoma. Eur J Nucl Med Mol Imaging 2010; 37 (01) 49-57
  17.  Kayani I, Bomanji JB, Groves A. et al. Functional imaging of neuroendocrine tumors with combined PET/CT using 68Ga-DOTATATE (DOTA-DPhe1,Tyr3-octreotate) and 18F-FDG. Cancer 2008; 112 (11) 2447-2455
  18.  Ambrosini V, Tomassetti P, Castellucci P. et al. Comparison between 68Ga-DOTA-NOC and 18F-DOPA PET for the detection of gastro-entero-pancreatic and lung neuro-endocrine tumours. Eur J Nucl Med Mol Imaging 2008; 35 (08) 1431-1438
  19.  Fanti S, Ambrosini V, Tomassetti P. et al. Evaluation of unusual neuroendocrine tumours by means of 68Ga-DOTA-NOC PET. Biomed Pharmacother 2008; 62 (10) 667-671
  20.  Kayani I, Conry BG, Groves AM. et al. A comparison of 68Ga-DOTATATE and 18F-FDG PET/CT in pulmonary neuroendocrine tumors. J Nucl Med 2009; 50 (12) 1927-1932
  21.  Ambrosini V, Castellucci P, Rubello D. et al. 68Ga-DOTA-NOC: a new PET tracer for evaluating patients with bronchial carcinoid. Nucl Med Commun 2009; 30 (04) 281-286
  22.  Schartinger VH, Dudás J, Decristoforo C. et al. 68Ga-DOTA0-Tyr3-octreotide positron emission tomography in head and neck squamous cell carcinoma. Eur J Nucl Med Mol Imaging 2013; 40 (09) 1365-1372
  23.  Kauhanen S, Seppänen M, Minn H. et al. Fluorine-18-L-dihydroxyphenylalanine (18F-DOPA) positron emission tomography as a tool to localize an insulinoma or beta-cell hyperplasia in adult patients. J Clin Endocrinol Metab 2007; 92 (04) 1237-1244
  24.  Traub-Weidinger T, Putzer D, von Guggenberg E. et al. Multiparametric PET imaging in thyroid malignancy characterizing tumour heterogeneity: somatostatin receptors and glucose metabolism. Eur J Nucl Med Mol Imaging 2015; 42 (13) 1995-2001
  25.  Prasad V, Ambrosini V, Hommann M, Hoersch D, Fanti S, Baum RP. Detection of unknown primary neuroendocrine tumours (CUP-NET) using (68)Ga-DOTA-NOC receptor PET/CT. Eur J Nucl Med Mol Imaging 2010; 37 (01) 67-77
  26.  Putzer D, Gabriel M, Henninger B. et al. Bone metastases in patients with neuroendocrine tumor: 68Ga-DOTA-Tyr3-octreotide PET in comparison to CT and bone scintigraphy. J Nucl Med 2009; 50 (08) 1214-1221
  27.  Ambrosini V, Nanni C, Zompatori M. et al. (68)Ga-DOTA-NOC PET/CT in comparison with CT for the detection of bone metastasis in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2010; 37 (04) 722-727
  28.  Ugur O, Kothari PJ, Finn RD. et al. Ga-66 labeled somatostatin analogue DOTA-DPhe1-Tyr3-octreotide as a potential agent for positron emission tomography imaging and receptor mediated internal radiotherapy of somatostatin receptor positive tumors. Nucl Med Biol 2002; 29 (02) 147-157
  29.  Gabriel M, Oberauer A, Dobrozemsky G. et al. 68Ga-DOTA-Tyr3-octreotide PET for assessing response to somatostatin-receptor-mediated radionuclide therapy. J Nucl Med 2009; 50 (09) 1427-1434
  30.  Pavel M, Öberg K, Falconi M. et al; ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Gastroenteropancreatic neuroendocrine neoplasms: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020; 31 (07) 844-860
  31.  Sundin A, Arnold R, Baudin E. et al; Antibes Consensus Conference participants. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: radiological, nuclear medicine & hybrid imaging. Neuroendocrinology 2017; 105 (03) 212-244
  32.  Shah MH, Goldner WS, Benson AB. et al. Neuroendocrine and adrenal tumors, Version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2021; 19 (07) 839-868
  33.  Barbosa FG, Queiroz MA, Nunes RF. et al. Revisiting prostate cancer recurrence with PSMA PET: atlas of typical and atypical patterns of spread. Radiographics 2019; 39 (01) 186-212
  34.  Scher HI, Fizazi K, Saad F. et al; AFFIRM Investigators. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012; 367 (13) 1187-1197
  35.  Kratochwil C, Giesel FL, Eder M. et al. [177Lu]Lutetium-labelled PSMA ligand-induced remission in a patient with metastatic prostate cancer. Eur J Nucl Med Mol Imaging 2015; 42 (06) 987-988
  36.  Afshar-Oromieh A, Avtzi E, Giesel FL. et al. The diagnostic value of PET/CT imaging with the (68)Ga-labelled PSMA ligand HBED-CC in the diagnosis of recurrent prostate cancer. Eur J Nucl Med Mol Imaging 2015; 42 (02) 197-209
  37.  Hofman MS, Hicks RJ, Maurer T, Eiber M. Prostate-specific membrane antigen PET: clinical utility in prostate cancer, normal patterns, pearls, and pitfalls. Radiographics 2018; 38 (01) 200-217
  38.  Eiber M, Maurer T, Souvatzoglou M. et al. Evaluation of hybrid 68Ga-PSMA ligand PET/CT in 248 patients with biochemical recurrence after radical prostatectomy. J Nucl Med 2015; 56 (05) 668-674
  39.  Rauscher I, Maurer T, Fendler WP, Sommer WH, Schwaiger M, Eiber M. (68)Ga-PSMA ligand PET/CT in patients with prostate cancer: how we review and report. Cancer Imaging 2016; 16 (01) 14
  40.  Ballas LK, de Castro Abreu AL, Quinn DI. What medical, urologic, and radiation oncologists want from molecular imaging of prostate cancer. J Nucl Med 2016; 57 (Suppl 3): 6S-12S
  41.  Fendler WP, Rahbar K, Herrmann K, Kratochwil C, Eiber M. 177Lu-PSMA radioligand therapy for prostate cancer. J Nucl Med 2017; 58 (08) 1196-1200
  42.  Emmett L, Willowson K, Violet J, Shin J, Blanksby A, Lee J. Lutetium 177 PSMA radionuclide therapy for men with prostate cancer: a review of the current literature and discussion of practical aspects of therapy. J Med Radiat Sci 2017; 64 (01) 52-60
  43.  Fendler WP, Eiber M, Beheshti M. et al. 68Ga-PSMA PET/CT: joint EANM and SNMMI procedure guideline for prostate cancer imaging: version 1.0. Eur J Nucl Med Mol Imaging 2017; 44 (06) 1014-1024
  44.  Trabulsi EJ, Rumble RB, Jadvar H. et al. Optimum imaging strategies for advanced prostate cancer: ASCO guideline. J Clin Oncol 2020; 38 (17) 1963-1996
  45.  Mottet N, van den Bergh RCN, Briers E. et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer-2020 Update. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol 2021; 79 (02) 243-262
  46.  Cornford P, van den Bergh RCN, Briers E. et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer. Part II-2020 update: treatment of relapsing and metastatic prostate cancer. Eur Urol 2021; 79 (02) 263-282
  47.  NCCN CLINICAL PRACTICE GUIDELINES IN ONCOLOGY. Prostate cancer version 3.2022-January 2022. Accessed December 12, 2022, at: https://www.nccn.org/login?ReturnURL=https://www.nccn.org/professionals/physician_gls/pdf/prostate.pdf
  48.  Jiang GM, Xu W, Du J. et al. The application of the fibroblast activation protein α-targeted immunotherapy strategy. Oncotarget 2016; 7 (22) 33472-33482
  49.  Lindner T, Loktev A, Altmann A. et al. Development of quinoline-based theranostic ligands for the targeting of fibroblast activation protein. J Nucl Med 2018; 59 (09) 1415-1422
  50.  Kratochwil C, Flechsig P, Lindner T. et al. 68Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med 2019; 60 (06) 801-805
  51.  Ballal S, Yadav MP, Kramer V. et al. A theranostic approach of [68Ga]Ga-DOTA.SA.FAPi PET/CT-guided [177Lu]Lu-DOTA.SA.FAPi radionuclide therapy in an end-stage breast cancer patient: new frontier in targeted radionuclide therapy. Eur J Nucl Med Mol Imaging 2021; 48 (03) 942-944
  52.  Ferdinandus J, Costa PF, Kessler L. et al. Initial clinical experience with 90Y-FAPI-46 radioligand therapy for advanced stage solid tumors: a case series of nine patients. J Nucl Med 2022; 63 (05) 727-734
  53.  Kratochwil C, Giesel FL, Rathke H. et al. [153Sm]Samarium-labeled FAPI-46 radioligand therapy in a patient with lung metastases of a sarcoma. Eur J Nucl Med Mol Imaging 2021; 48 (09) 3011-3013
  54.  Regmi SK, Sathianathen N, Stout TE, Konety BR. MRI/PET Imaging in elevated PSA and localized prostate cancer: a narrative review. Transl Androl Urol 2021; 10 (07) 3117-3129
  55.  Liu F, Dong J, Shen Y. et al. Comparison of PET/CT and MRI in the diagnosis of bone metastasis in prostate cancer patients: a network analysis of diagnostic studies. Front Oncol 2021; 11: 736654
  56.  Kichloo A, Amir R, Aljadah M. et al. FDG-PET versus PSMA-PET: a patient with prostate cancer. J Investig Med High Impact Case Rep 2020; 8: 2324709620941313
  57.  Tsechelidis I, Vrachimis A. PSMA PET in Imaging Prostate Cancer. Front Oncol 2022; 12: 831429
  58.  Smith-Bindman R, Lipson J, Marcus R. et al. Radiation dose associated with common computed tomography examinations and the associated lifetime attributable risk of cancer. Arch Intern Med 2009; 169 (22) 2078-2086
  59.  Kwon HW, Kim JP, Lee HJ. et al. Radiation dose from whole-body F-18 fluorodeoxyglucose positron emission tomography/computed tomography: Nationwide Survey in Korea. J Korean Med Sci 2016; 31 Suppl 1 (Suppl 1): S69-S74
  60.  Waller J, Flavell R, Heath CL. High accuracy of PSMA PET in initial staging of high-risk prostate cancer. Radiol Imaging Cancer 2020; 2 (04) e204025
  61.  Maxwell JE, Howe JR. Imaging in neuroendocrine tumors: an update for the clinician. Int J Endocr Oncol 2015; 2 (02) 159-168
  62.  Gamal GH. The utility of 18F-FDG PET/CT in the diagnosis, staging of non-functioning pancreatic neuroendocrine tumors. Egypt J Radiol Nucl Med 2021; 52 (01) 234
  63.  Schraml C, Schwenzer NF, Sperling O. et al. Staging of neuroendocrine tumours: comparison of [68Ga]DOTATOC multiphase PET/CT and whole-body MRI. Cancer Imaging 2013; 13 (01) 63-72
  64.  Garcia-Carbonero R, Garcia-Figueiras R, Carmona-Bayonas A. et al; Spanish Cooperative Group of Neuroendocrine Tumors (GETNE). Imaging approaches to assess the therapeutic response of gastroenteropancreatic neuroendocrine tumors (GEP-NETs): current perspectives and future trends of an exciting field in development. Cancer Metastasis Rev 2015; 34 (04) 823-842
  65.  Accessed December 12, 2022, at: https://clinicaltrials.gov/ProvidedDocs/28/NCT03136328/Prot_SAP_000.pdf
  66.  Naswa N, Sharma P, Kumar A. et al. Gallium-68-DOTA-NOC PET/CT of patients with gastroenteropancreatic neuroendocrine tumors: a prospective single-center study. AJR Am J Roentgenol 2011; 197 (05) 1221-1228
  67.  Thapa P, Ranade R, Ostwal V, Shrikhande SV, Goel M, Basu S. Performance of 177Lu-DOTATATE-based peptide receptor radionuclide therapy in metastatic gastroenteropancreatic neuroendocrine tumor: a multiparametric response evaluation correlating with primary tumor site, tumor proliferation index, and dual tracer imaging characteristics. Nucl Med Commun 2016; 37 (10) 1030-1037
  68.  Sampathirao N, Basu S. MIB-1 index-stratified assessment of dual-tracer PET/CT with 68Ga-DOTATATE and 18F-FDG and multimodality anatomic imaging in metastatic neuroendocrine tumors of unknown primary in a PRRT workup setting. J Nucl Med Technol 2017; 45 (01) 34-41
  69.  Basu S, Sirohi B, Shrikhande SV. Dual tracer imaging approach in assessing tumor biology and heterogeneity in neuroendocrine tumors: its correlation with tumor proliferation index and possible multifaceted implications for personalized clinical management decisions, with focus on PRRT. Eur J Nucl Med Mol Imaging 2014; 41 (08) 1492-1496
  70.  Basu S, Chakraborty S, Parghane RV. et al. One decade of ‘Bench-to-Bedside’ peptide receptor radionuclide therapy with indigenous [177Lu]Lu-DOTATATE obtained through ‘Direct’ neutron activation route: lessons learnt including practice evolution in an Indian setting. Am J Nucl Med Mol Imaging 2020; 10 (04) 178-211
  71.  Basu S, Parghane RV, Kamaldeep, Chakrabarty S. Peptide receptor radionuclide therapy of neuroendocrine tumors. Semin Nucl Med 2020; 50 (05) 447-464
  72.  Parghane RV, Talole S, Basu S. Prevalence of hitherto unknown brain meningioma detected on 68Ga-DOTATATE positron-emission tomography/computed tomography in patients with metastatic neuroendocrine tumor and exploring potential of 177Lu-DOTATATE peptide receptor radionuclide therapy as single-shot treatment approach targeting both tumors. World J Nucl Med 2019; 18 (02) 160-170
  73.  Parghane RV, Talole S, Basu S. 131I-MIBG negative progressive symptomatic metastatic paraganglioma: response and outcome with 177Lu-DOTATATE peptide receptor radionuclide therapy. Ann Nucl Med 2021; 35 (01) 92-101
  74.  Parghane RV, Ostwal V, Ramaswamy A. et al. Long-term outcome of “Sandwich” chemo-PRRT: a novel treatment strategy for metastatic neuroendocrine tumors with both FDG- and SSTR-avid aggressive disease. Eur J Nucl Med Mol Imaging 2021; 48 (03) 913-923
  75.  Parghane RV, Naik C, Talole S. et al. Clinical utility of 177 Lu-DOTATATE PRRT in somatostatin receptor-positive metastatic medullary carcinoma of thyroid patients with assessment of efficacy, survival analysis, prognostic variables, and toxicity. Head Neck 2020; 42 (03) 401-416
  76.  Basu S, Parghane RV, Naik C. Clinical efficacy of 177Lu-DOTATATE peptide receptor radionuclide therapy in thyroglobulin-elevated negative iodine scintigraphy: a “not-so-promising” result compared to GEP-NETs. World J Nucl Med 2020; 19 (03) 205-210
  77.  Parghane RV, Bhandare M, Chaudhari V. et al. Surgical feasibility, determinants, and overall efficacy of neoadjuvant 177Lu-DOTATATE PRRT for locally advanced unresectable gastroenteropancreatic neuroendocrine tumors. J Nucl Med 2021; 62 (11) 1558-1563
  78.  Jain H, Sood R, Faridi MS, Goel H, Sharma U. Role of 68Ga-PSMA-PET/CT for the detection of primary prostate cancer prior to biopsy: a prospective study. Cent European J Urol 2021; 74 (03) 315-320
  79.  Kallur KG, Ramachandra PG, Rajkumar K. et al. Clinical utility of Gallium-68 PSMA PET/CT scan for prostate cancer. Indian J Nucl Med 2017; 32 (02) 110-117
  80.  Suman S, Parghane RV, Joshi A. et al. Therapeutic efficacy, prognostic variables and clinical outcome of 177Lu-PSMA-617 PRLT in progressive mCRPC following multiple lines of treatment: prognostic implications of high FDG uptake on dual tracer PET-CT vis-à-vis Gleason score in such cohort. Br J Radiol 2019; 92 (1104): 20190380
  81.  Goel R, Shukla J, Bansal D. et al. (68)Ga-DOTATATE positron emission tomography/computed tomography scan in the detection of bone metastases in pediatric neuroendocrine tumors. Indian J Nucl Med 2014; 29 (01) 13-17
  82.  Jha A, Ling A, Millo C. et al. Superiority of 68Ga-DOTATATE over 18F-FDG and anatomic imaging in the detection of succinate dehydrogenase mutation (SDHx )-related pheochromocytoma and paraganglioma in the pediatric population. Eur J Nucl Med Mol Imaging 2018; 45 (05) 787-797
  83.  Maaz AUR, O'Doherty J, Djekidel M. 68Ga-DOTATATE PET/CT for neuroblastoma staging: utility for clinical use. J Nucl Med Technol 2021; 49 (03) 265-268