open access

Vol 24, No 2 (2021)
Review paper
Submitted: 2020-11-19
Accepted: 2021-03-16
Published online: 2021-07-30
Get Citation

State-of-the-art modalities in cardio-oncology: insight from a nuclear medicine approach

Narges Jokar1, Abdullatif Amini2, Mohammadreza Ravanbod3, Maryam Barekat4, Hossein Shooli1, Ali Gholamrezanezhad5, Majid Assadi1
·
Pubmed: 34382672
·
Nucl. Med. Rev 2021;24(2):82-92.
Affiliations
  1. The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Radionuclide Therapy (MIRT), Bushehr Medical University Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
  2. Medical Heart Center, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
  3. Department of Internal Medicine, Persian Gulf Tropical Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran, Iran-bousher- boushehr university of medical science, 45654775 Bushehr, Iran, Islamic Republic Of
  4. Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
  5. Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, United States

open access

Vol 24, No 2 (2021)
Reviews
Submitted: 2020-11-19
Accepted: 2021-03-16
Published online: 2021-07-30

Abstract

Cancer and cardiovascular disease are the most significant causes of morbidity and mortality worldwide. Although the cancer survival rate has increased due to improved treatment approaches, especially targeted therapy, some side effects such as cardiotoxicity decrease the efficiency of the clinical outcome. Radiation therapy and chemotherapy have a long-established history of potential cardiotoxic effects. A new multi-disciplinary and translational field known as cardio-oncology has been developed for the identification, prevention, and treatment of cardiovascular dysfunctions associated with cancer treatment approaches. One of the important tools for detecting and monitoring cardiotoxic effects is non-invasive nuclear cardiac imaging techniques. Cardiac nuclear imaging modalities especially recent findings positron emission tomography (PET) tracers have a quintessential role in the early detection of cardiovascular disorders. Moreover, comprehensive studies are required to investigate novel nuclear medicine treatment approaches such as peptide receptor radionuclide therapy (PRRT), fibroblast activation protein (FAP), and chemokine receptor (CXCR) targeting probes for possible cardiac side effects that play important roles in the treatment of malignancies

Abstract

Cancer and cardiovascular disease are the most significant causes of morbidity and mortality worldwide. Although the cancer survival rate has increased due to improved treatment approaches, especially targeted therapy, some side effects such as cardiotoxicity decrease the efficiency of the clinical outcome. Radiation therapy and chemotherapy have a long-established history of potential cardiotoxic effects. A new multi-disciplinary and translational field known as cardio-oncology has been developed for the identification, prevention, and treatment of cardiovascular dysfunctions associated with cancer treatment approaches. One of the important tools for detecting and monitoring cardiotoxic effects is non-invasive nuclear cardiac imaging techniques. Cardiac nuclear imaging modalities especially recent findings positron emission tomography (PET) tracers have a quintessential role in the early detection of cardiovascular disorders. Moreover, comprehensive studies are required to investigate novel nuclear medicine treatment approaches such as peptide receptor radionuclide therapy (PRRT), fibroblast activation protein (FAP), and chemokine receptor (CXCR) targeting probes for possible cardiac side effects that play important roles in the treatment of malignancies

Get Citation

Keywords

cardiotoxicity; cardio-oncology; PRRT-related cardiotoxicity; nuclear medicine; FAPI; CXCR4; [99mTc]Sestamibi; [18F]MitoPhos; [68Ga]Galmydar

About this article
Title

State-of-the-art modalities in cardio-oncology: insight from a nuclear medicine approach

Journal

Nuclear Medicine Review

Issue

Vol 24, No 2 (2021)

Article type

Review paper

Pages

82-92

Published online

2021-07-30

Page views

7827

Article views/downloads

1237

DOI

10.5603/NMR.2021.0019

Pubmed

34382672

Bibliographic record

Nucl. Med. Rev 2021;24(2):82-92.

Keywords

cardiotoxicity
cardio-oncology
PRRT-related cardiotoxicity
nuclear medicine
FAPI
CXCR4
[99mTc]Sestamibi
[18F]MitoPhos
[68Ga]Galmydar

Authors

Narges Jokar
Abdullatif Amini
Mohammadreza Ravanbod
Maryam Barekat
Hossein Shooli
Ali Gholamrezanezhad
Majid Assadi

References (111)
  1. Fuster V, Voûte J. MDGs: chronic diseases are not on the agenda. The Lancet. 2005; 366(9496): 1512–1514.
  2. Hofmann L, Forschner A, Loquai C, et al. Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer. 2016; 60: 190–209.
  3. Sivapackiam J, Sharma M, Schindler TH, et al. PET radiopharmaceuticals for imaging chemotherapy-induced cardiotoxicity. Curr Cardiol Rep. 2020; 22(8): 62.
  4. Herrmann J, Lerman A, Sandhu NP, et al. Evaluation and management of patients with heart disease and cancer: cardio-oncology. Mayo Clin Proc. 2014; 89(9): 1287–1306.
  5. Lee CK, Aeppli D, Nierengarten ME. The need for long-term surveillance for patients treated with curative radiotherapy for Hodgkin's disease: University of Minnesota experience. Int J Radiat Oncol Biol Phys. 2000; 48(1): 169–179.
  6. Jaworski C, Mariani JA, Wheeler G, et al. Cardiac complications of thoracic irradiation. J Am Coll Cardiol. 2013; 61(23): 2319–2328.
  7. Aapro M, Bernard-Marty C, Brain EGC, et al. Anthracycline cardiotoxicity in the elderly cancer patient: a SIOG expert position paper. Ann Oncol. 2011; 22(2): 257–267.
  8. Serrano C, Cortés J, De Mattos-Arruda L, et al. Trastuzumab-related cardiotoxicity in the elderly: a role for cardiovascular risk factors. Ann Oncol. 2012; 23(4): 897–902.
  9. Jain D, Russell RR, Schwartz RG, et al. Cardiac complications of cancer therapy: pathophysiology, identification, prevention, treatment, and future directions. Curr Cardiol Rep. 2017; 19(5): 36.
  10. Russell RR, Alexander J, Jain D, et al. The role and clinical effectiveness of multimodality imaging in the management of cardiac complications of cancer and cancer therapy. J Nucl Cardiol. 2016; 23(4): 856–884.
  11. Sardaro A, Petruzzelli MF, D'Errico MP, et al. Radiation-induced cardiac damage in early left breast cancer patients: risk factors, biological mechanisms, radiobiology, and dosimetric constraints. Radiother Oncol. 2012; 103(2): 133–142.
  12. Hurtado-de-Mendoza D, Loaiza-Bonilla A, Bonilla-Reyes PA, et al. Cardio-oncology: cancer therapy-related cardiovascular complications in a molecular targeted era: new concepts and perspectives. Cureus. 2017; 9(5): e1258.
  13. Plana JC, Galderisi M, Barac A, et al. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J Am Soc Echocardiogr. 2014; 27(9): 911–939.
  14. Abdel-Qadir H, Austin PC, Lee DS, et al. A population-based study of cardiovascular mortality following early-stage breast cancer. JAMA Cardiol. 2017; 2(1): 88–93.
  15. Makavos G, Ikonomidis I, Palios J, et al. Cardiac imaging in cardiotoxicity: a focus on clinical practice. Heart Fail Rev. 2020 [Epub ahead of print].
  16. Pondé NF, Lambertini M, de Azambuja E. Twenty years of anti-HER2 therapy-associated cardiotoxicity. ESMO Open. 2016; 1(4): e000073.
  17. Bovelli D, Plataniotis G, Roila F, et al. ESMO Guidelines Working Group. Cardiotoxicity of chemotherapeutic agents and radiotherapy-related heart disease: ESMO Clinical Practice Guidelines. Ann Oncol. 2010; 21(Suppl 5): v277–v282.
  18. Donnellan E, Phelan D, McCarthy CP, et al. Radiation-induced heart disease: A practical guide to diagnosis and management. Cleve Clin J Med. 2016; 83(12): 914–922.
  19. Aleman BMP, van den Belt-Dusebout AW, Klokman WJ, et al. Long-term cause-specific mortality of patients treated for Hodgkin's disease. J Clin Oncol. 2003; 21(18): 3431–3439.
  20. Cutter DJ, Darby SC, Yusuf SW. Risks of heart disease after radiotherapy. Tex Heart Inst J. 2011; 38(3): 257–258.
  21. Early Breast Cancer Trialists' Collaborative Group (EBCTCG). Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. The Lancet. 2005; 366(9503): 2087–2106.
  22. Popescu CC, Olivotto IA, Beckham WA, et al. Volumetric modulated arc therapy improves dosimetry and reduces treatment time compared to conventional intensity-modulated radiotherapy for locoregional radiotherapy of left-sided breast cancer and internal mammary nodes. Int J Radiat Oncol Biol Phys. 2010; 76(1): 287–295.
  23. Osman SOS, Hol S, Poortmans PM, et al. Volumetric modulated arc therapy and breath-hold in image-guided locoregional left-sided breast irradiation. Radiother Oncol. 2014; 112(1): 17–22.
  24. Sakumi A, Shiraishi K, Onoe T, et al. Single-arc volumetric modulated arc therapy planning for left breast cancer and regional nodes. J Radiat Res. 2012; 53(1): 151–153.
  25. Lauche O, Kirova YM. Helical tomotherapy in breast cancer treatment. Breast Cancer Management. 2014; 3(5): 441–449.
  26. Lohr F, El-Haddad M, Dobler B, et al. Potential effect of robust and simple IMRT approach for left-sided breast cancer on cardiac mortality. Int J Radiat Oncol Biol Phys. 2009; 74(1): 73–80.
  27. Tan W, Wang X, Qiu D, et al. Dosimetric comparison of intensity-modulated radiotherapy plans, with or without anterior myocardial territory and left ventricle as organs at risk, in early-stage left-sided breast cancer patients. Int J Radiat Oncol Biol Phys. 2011; 81(5): 1544–1551.
  28. Cao J, Roeske JC, Chmura SJ, et al. Calculation and prediction of the effect of respiratory motion on whole breast radiation therapy dose distributions. Med Dosim. 2009; 34(2): 126–132.
  29. Borst GR, Sonke JJ, den Hollander S, et al. Clinical results of image-guided deep inspiration breath hold breast irradiation. Int J Radiat Oncol Biol Phys. 2010; 78(5): 1345–1351.
  30. Lymberis SC, deWyngaert JK, Parhar P, et al. Prospective assessment of optimal individual position (prone versus supine) for breast radiotherapy: volumetric and dosimetric correlations in 100 patients. Int J Radiat Oncol Biol Phys. 2012; 84(4): 902–909.
  31. Bradley JA, Dagan R, Ho MW, et al. Initial report of a prospective dosimetric and clinical feasibility trial demonstrates the potential of protons to increase the therapeutic ratio in breast cancer compared with photons. Int J Radiat Oncol Biol Phys. 2016; 95(1): 411–421.
  32. Coon AB, Dickler A, Kirk MC, et al. Tomotherapy and multifield intensity-modulated radiotherapy planning reduce cardiac doses in left-sided breast cancer patients with unfavorable cardiac anatomy. Int J Radiat Oncol Biol Phys. 2010; 78(1): 104–110.
  33. Kirova YM, Gambotti L, De Rycke Y, et al. Risk of second malignancies after adjuvant radiotherapy for breast cancer: a large-scale, single-institution review. Int J Radiat Oncol Biol Phys. 2007; 68(2): 359–363.
  34. Lally BE, Detterbeck FC, Geiger AM, et al. The risk of death from heart disease in patients with nonsmall cell lung cancer who receive postoperative radiotherapy: analysis of the surveillance, epidemiology, and end results database. Cancer. 2007; 110(4): 911–917.
  35. Wang K, Pearlstein KA, Patchett ND, et al. Heart dosimetric analysis of three types of cardiac toxicity in patients treated on dose-escalation trials for Stage III non-small-cell lung cancer. Radiother Oncol. 2017; 125(2): 293–300.
  36. Kimmick GG, Lenihan DJ, Sawyer DB, Mayer EL, Hershman DL. Cardio-oncology: the clinical overlap of cancer and heart disease. Springer, Cham 2017.
  37. Luminari S, Montanini A, Caballero D, et al. Nonpegylated liposomal doxorubicin (MyocetTM) combination (R-COMP) chemotherapy in elderly patients with diffuse large B-cell lymphoma (DLBCL): results from the phase II EUR018 trial. Ann Oncol. 2010; 21(7): 1492–1499.
  38. Cardinale D, Sandri MT, Martinoni A, et al. Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy. J Am Coll Cardiol. 2000; 36(2): 517–522.
  39. Drews J. Drug discovery: a historical perspective. Science. 2000; 287(5460): 1960–1964.
  40. Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 2004; 25(4): 581–611.
  41. Ferrara N. VEGF and the quest for tumour angiogenesis factors. Nat Rev Cancer. 2002; 2(10): 795–803.
  42. Shabalala S, Muller CJF, Louw J, et al. Polyphenols, autophagy and doxorubicin-induced cardiotoxicity. Life Sci. 2017; 180: 160–170.
  43. Novo G, Cadeddu C, Sucato V, et al. Role of biomarkers in monitoring antiblastic cardiotoxicity. J Cardiovasc Med (Hagerstown). 2016; 17(Suppl 1): S27–S34.
  44. Hall PS, Harshman LC, Srinivas S, et al. The frequency and severity of cardiovascular toxicity from targeted therapy in advanced renal cell carcinoma patients. JACC Heart Fail. 2013; 1(1): 72–78.
  45. Qi WX, Fu S, Zhang Q, et al. Bevacizumab increases the risk of severe congestive heart failure in cancer patients: an up-to-date meta-analysis with a focus on different subgroups. Clin Drug Investig. 2014; 34(10): 681–690.
  46. Groarke JD, Choueiri TK, Slosky D, et al. Recognizing and managing left ventricular dysfunction associated with therapeutic inhibition of the vascular endothelial growth factor signaling pathway. Curr Treat Options Cardiovasc Med. 2014; 16(9): 335.
  47. Choueiri TK, Mayer EL, Je Y, et al. Congestive heart failure risk in patients with breast cancer treated with bevacizumab. J Clin Oncol. 2011; 29(6): 632–638.
  48. Moja L, Tagliabue L, Balduzzi S, et al. Trastuzumab containing regimens for early breast cancer. Cochrane Database Syst Rev. 2012(4): CD006243.
  49. Blackwell KL, Pegram MD, Tan-Chiu E, et al. Single-agent lapatinib for HER2-overexpressing advanced or metastatic breast cancer that progressed on first- or second-line trastuzumab-containing regimens. Ann Oncol. 2009; 20(6): 1026–1031.
  50. Patnaik JL, Byers T, DiGuiseppi C, et al. Cardiovascular disease competes with breast cancer as the leading cause of death for older females diagnosed with breast cancer: a retrospective cohort study. Breast Cancer Res. 2011; 13(3): R64.
  51. Bellenger NG, Burgess MI, Ray SG, et al. Comparison of left ventricular ejection fraction and volumes in heart failure by echocardiography, radionuclide ventriculography and cardiovascular magnetic resonance; are they interchangeable? Eur Heart J. 2000; 21(16): 1387–1396.
  52. Schwartz RG, Jain D, Storozynsky E. Traditional and novel methods to assess and prevent chemotherapy-related cardiac dysfunction noninvasively. J Nucl Cardiol. 2013; 20(3): 443–464.
  53. Kolla BC, Roy SS, Duval S, et al. Cardiac imaging methods for chemotherapy-related cardiotoxicity screening and related radiation exposure: current practice and trends. Anticancer Res. 2017; 37(5): 2445–2449.
  54. Wexler O, Yoder SR, Elder JL, et al. Effect of gender on cardiovascular risk stratification with ECG gated SPECT left ventricular volume indices and ejection fraction. J Nucl Cardiol. 2009; 16(1): 28–37.
  55. Liu YH, Fazzone-Chettiar R, Sandoval V, et al. New approach for quantification of left ventricular function from low-dose gated bloodpool SPECT: Validation and comparison with conventional methods in patients. J Nucl Cardiol. 2019 [Epub ahead of print].
  56. Groch MW, DePuey EG, Belzberg AC, et al. Planar imaging versus gated blood-pool SPECT for the assessment of ventricular performance: a multicenter study. J Nucl Med. 2001; 42(12): 1773–1779.
  57. Lipshultz SE, Karnik R, Sambatakos P, et al. Anthracycline-related cardiotoxicity in childhood cancer survivors. Curr Opin Cardiol. 2014; 29(1): 103–112.
  58. Cutter DJ, Schaapveld M, Darby SC, et al. Risk of valvular heart disease after treatment for Hodgkin lymphoma. J Natl Cancer Inst. 2015; 107(4): djv008.
  59. Ichikawa Y, Ghanefar M, Bayeva M, et al. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J Clin Invest. 2014; 124(2): 617–630.
  60. Safee ZM, Baark F, Waters ECT, et al. Detection of anthracycline-induced cardiotoxicity using perfusion-corrected Tc sestamibi SPECT. Sci Rep. 2019; 9(1): 216.
  61. Carrió I, Estorch M, Berná L, et al. Indium-111-antimyosin and iodine-123-MIBG studies in early assessment of doxorubicin cardiotoxicity. J Nucl Med. 1995; 36(11): 2044–2049.
  62. Russell RR, Zaret BL. Nuclear cardiology: present and future. Curr Probl Cardiol. 2006; 31(9): 557–629.
  63. Rosa GM, Gigli L, Tagliasacchi MI, et al. Update on cardiotoxicity of anti-cancer treatments. Eur J Clin Invest. 2016; 46(3): 264–284.
  64. Wenningmann N, Knapp M, Ande A, et al. Insights into Doxorubicin-induced Cardiotoxicity: Molecular Mechanisms, Preventive Strategies, and Early Monitoring. Mol Pharmacol. 2019; 96(2): 219–232.
  65. Jones RL, Swanton C, Ewer MS. Anthracycline cardiotoxicity. Expert Opin Drug Saf. 2006; 5(6): 791–809.
  66. Pepe A, Pizzino F, Gargiulo P, et al. Cardiovascular imaging in the diagnosis and monitoring of cardiotoxicity: cardiovascular magnetic resonance and nuclear cardiology. J Cardiovasc Med (Hagerstown). 2016; 17(Suppl 1): S45–S54.
  67. Blankenberg FG. In vivo detection of apoptosis. J Nucl Med. 2008; 49(Suppl 2): 81S–95S.
  68. Wang X, Liu Y, Wang X, et al. The role of (99m)Tc-annexin V apoptosis scintigraphy in visualizing early stage glucocorticoid-induced femoral head osteonecrosis in the rabbit. Biomed Res Int. 2016: 7067259.
  69. Boersma HH, Kietselaer BL, Stolk LML, et al. Past, present, and future of annexin A5: from protein discovery to clinical applications. J Nucl Med. 2005; 46(12): 2035–2050.
  70. Kietselaer BL, Reutelingsperger CPM, Boersma HH, et al. Noninvasive detection of programmed cell loss with 99mTc-labeled annexin A5 in heart failure. J Nucl Med. 2007; 48(4): 562–567.
  71. Taki J, Higuchi T, Kawashima A, et al. Effect of postconditioning on myocardial 99mTc-annexin-V uptake: comparison with ischemic preconditioning and caspase inhibitor treatment. J Nucl Med. 2007; 48(8): 1301–1307.
  72. Bennink RJ, van den Hoff MJ, van Hemert FJ, et al. Annexin V imaging of acute doxorubicin cardiotoxicity (apoptosis) in rats. J Nucl Med. 2004; 45(5): 842–848.
  73. Knapp FF, Kropp J. Iodine-123-labelled fatty acids for myocardial single-photon emission tomography: current status and future perspectives. Eur J Nucl Med. 1995; 22(4): 361–381.
  74. Elmaleh DR, Fischman AJ, Shoup TM. Method for monitoring blood flow and metabolic uptake in tissue with radiolabeled alkanoic acid. Google Patents. 2009: US8268291B2.
  75. Hashimura H, Kiso K, Yamada N, et al. Myocardial impairment detected by late gadolinium enhancement in hypertrophic cardiomyopathy: comparison with 99mTc-MIBI/tetrofosmin and 123I-BMIPP SPECT. Kobe J Med Sci. 2013; 59(3): E81–E92.
  76. Nowsheen S, Viscuse PV, O'Sullivan CC, et al. Incidence, diagnosis, and treatment of cardiac toxicity from trastuzumab in patients with breast cancer. Curr Breast Cancer Rep. 2017; 9(3): 173–182.
  77. Martín M, Esteva FJ, Alba E, et al. Minimizing cardiotoxicity while optimizing treatment efficacy with trastuzumab: review and expert recommendations. Oncologist. 2009; 14(1): 1–11.
  78. McLarty K, Cornelissen B, Scollard DA, et al. Associations between the uptake of 111In-DTPA-trastuzumab, HER2 density and response to trastuzumab (Herceptin) in athymic mice bearing subcutaneous human tumour xenografts. Eur J Nucl Med Mol Imaging. 2009; 36(1): 81–93.
  79. Perik PJ, Lub-De Hooge MN, Gietema JA, et al. Indium-111-labeled trastuzumab scintigraphy in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer. J Clin Oncol. 2006; 24(15): 2276–2282.
  80. de Korte MA, de Vries EGE, Lub-de Hooge MN, et al. 111Indium-trastuzumab visualises myocardial human epidermal growth factor receptor 2 expression shortly after anthracycline treatment but not during heart failure: a clue to uncover the mechanisms of trastuzumab-related cardiotoxicity. Eur J Cancer. 2007; 43(14): 2046–2051.
  81. Bergmann SR, Fox KA, Geltman EM, et al. Positron emission tomography of the heart. Prog Cardiovasc Dis. 1985; 28(3): 165–194.
  82. Yoshinaga K, Klein R, Tamaki N. Generator-produced rubidium-82 positron emission tomography myocardial perfusion imaging-From basic aspects to clinical applications. J Cardiol. 2010; 55(2): 163–173.
  83. Sarocchi M, Bauckneht M, Arboscello E, et al. An increase in myocardial 18-fluorodeoxyglucose uptake is associated with left ventricular ejection fraction decline in Hodgkin lymphoma patients treated with anthracycline. J Transl Med. 2018; 16(1): 295.
  84. Zhang W, Cai Z, Li L, et al. Optimized and automated radiosynthesis of [F]DHMT for translational imaging of reactive oxygen species with positron emission tomography. Molecules. 2016; 21(12): 1696.
  85. Chu W, Chepetan A, Zhou D, et al. Development of a PET radiotracer for non-invasive imaging of the reactive oxygen species, superoxide, in vivo. Org Biomol Chem. 2014; 12(25): 4421–4431.
  86. Boutagy NE, Wu J, Cai Z, et al. In vivo reactive oxygen species detection with a novel positron emission tomography tracer, F-DHMT, allows for early detection of anthracycline-induced cardiotoxicity in rodents. JACC Basic Transl Sci. 2018; 3(3): 378–390.
  87. Sivapackiam J, Kabra S, Speidel S, et al. 68Ga-Galmydar: A PET imaging tracer for noninvasive detection of Doxorubicin-induced cardiotoxicity. PLoS One. 2019; 14(5): e0215579.
  88. McCluskey SP, Haslop A, Coello C, et al. Imaging of chemotherapy-induced acute cardiotoxicity with f-labeled lipophilic cations. J Nucl Med. 2019; 60(12): 1750–1756.
  89. Laursen AH, Elming MB, Ripa RS, et al. Rubidium-82 positron emission tomography for detection of acute doxorubicin-induced cardiac effects in lymphoma patients. J Nucl Cardiol. 2020; 27(5): 1698–1707.
  90. Ahmadzadehfar H, Eppard E, Kürpig S, et al. Therapeutic response and side effects of repeated radioligand therapy with 177Lu-PSMA-DKFZ-617 of castrate-resistant metastatic prostate cancer. Oncotarget. 2016; 7(11): 12477–12488.
  91. Yordanova A, Mayer K, Brossart P, et al. Safety of multiple repeated cycles of Lu-octreotate in patients with recurrent neuroendocrine tumour. Eur J Nucl Med Mol Imaging. 2017; 44(7): 1207–1214.
  92. Bodei L, Kidd M, Paganelli G, et al. Long-term tolerability of PRRT in 807 patients with neuroendocrine tumours: the value and limitations of clinical factors. Eur J Nucl Med Mol Imaging. 2015; 42(1): 5–19.
  93. Sabet A, Haslerud T, Pape UF, et al. Outcome and toxicity of salvage therapy with 177Lu-octreotate in patients with metastatic gastroenteropancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2014; 41(2): 205–210.
  94. Sabet A, Ezziddin K, Pape UF, et al. Long-term hematotoxicity after peptide receptor radionuclide therapy with 177Lu-octreotate. J Nucl Med. 2013; 54(11): 1857–1861.
  95. Sabet A, Ezziddin K, Pape UF, et al. Accurate assessment of long-term nephrotoxicity after peptide receptor radionuclide therapy with (177)Lu-octreotate. Eur J Nucl Med Mol Imaging. 2014; 41(3): 505–510.
  96. Ahmadzadehfar H, Rahbar K, Kürpig S, et al. Early side effects and first results of radioligand therapy with (177)Lu-DKFZ-617 PSMA of castrate-resistant metastatic prostate cancer: a two-centre study. EJNMMI Res. 2015; 5(1): 114.
  97. Skrabanek P, Cannon D, Kirrane J, et al. Substance P secretion by carcinoid tumours. Ir J Med Sci. 1978; 147(2): 47–49.
  98. van der Lely AJ, de Herder WW. Carcinoid syndrome: diagnosis and medical management. Arq Bras Endocrinol Metabol. 2005; 49(5): 850–860.
  99. Werner RA, Bundschuh RA, Bundschuh L, et al. Molecular imaging reporting and data systems (MI-RADS): a generalizable framework for targeted radiotracers with theranostic implications. Ann Nucl Med. 2018; 32(8): 512–522.
  100. Brabander T, van der Zwan WA, Teunissen JJM, et al. Long-term efficacy, survival, and safety of [177Lu, DOTA0, Tyr3] octreotate in patients with gastroenteropancreatic and bronchial neuroendocrine tumors. Clin Cancer Res. 2017; 23(16): 4617–4624.
  101. Bergsma H, van Lom K, Raaijmakers MH, et al. Persistent hematologic dysfunction after peptide receptor radionuclide therapy with 177Lu-DOTATATE: incidence, course, and predicting factors in patients with gastroenteropancreatic neuroendocrine tumors. J Nucl Med. 2018; 59(3): 452–458.
  102. Hall R. Neoplastic heart disease. The heart, arteries and veins. McGraw-Hill, New York 1990: 1382–1403.
  103. Smith WHT, Nair RU, Adamson D, et al. Somatostatin receptor subtype expression in the human heart: differential expression by myocytes and fibroblasts. J Endocrinol. 2005; 187(3): 379–386.
  104. Hendifar AE, Delpassand ES, Kittleson MM, et al. Cardiac toxicity in a patient receiving peptide receptor radionuclide therapy. Pancreas. 2018; 47(8): e55–e56.
  105. Chatterjee S, Azad BB, Nimmagadda S. The intricate role of CXCR4 in cancer. Adv Cancer Res. 2014; 124: 31–82.
  106. Kratochwil C, Flechsig P, Lindner T, et al. Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med. 2019; 60(6): 801–805.
  107. Lapa C, Lückerath K, Rudelius M, et al. [68Ga]Pentixafor-PET/CT for imaging of chemokine receptor 4 expression in small cell lung cancer--initial experience. Oncotarget. 2016; 7(8): 9288–9295.
  108. Kircher M, Herhaus P, Schottelius M, et al. CXCR4-directed theranostics in oncology and inflammation. Ann Nucl Med. 2018; 32(8): 503–511.
  109. Vag T, Gerngross C, Herhaus P, et al. First experience with chemokine receptor cxcr4-targeted PET imaging of patients with solid cancers. J Nucl Med. 2016; 57(5): 741–746.
  110. Schottelius M, Osl T, Poschenrieder A, et al. [Lu]pentixather: comprehensive preclinical characterization of a first cxcr4-directed endoradiotherapeutic agent. Theranostics. 2017; 7(9): 2350–2362.
  111. Giesel FL, Kratochwil C, Lindner T, et al. Ga-FAPI PET/CT: biodistribution and preliminary dosimetry estimate of 2 DOTA-containing FAP-targeting agents in patients with various cancers. J Nucl Med. 2019; 60(3): 386–392.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., Świętokrzyska 73 street, 80–180 Gdańsk, Poland

phone: +48 58 320 94 94, fax: +48 58 320 94 60, e-mail: viamedica@viamedica.pl