1
|
Gaidzik VI, Mayer-Steinacker R, Wittau M, Schultheiß M, v. Baer A, Oehl-Huber K, Dahlum S, Fischer A, Gerstenmaier U, Seufferlein T, Buck A, Beer A, Thaiss W, Möller P, Döhner H, Siebert R, Marienfeld R, Barth TFE. Desmoplastic Small Round Cell Tumors: Clinical Presentation, Molecular Characterization, and Therapeutic Approach of Seven Patients. Sarcoma 2024; 2024:5036102. [PMID: 39411551 PMCID: PMC11479773 DOI: 10.1155/2024/5036102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Desmoplastic small round blue cell tumor (DSRCT) is a highly aggressive fatal sarcoma without evidence-based therapeutic guidelines. We present here seven patients with DSRCT including immunohistochemistry combined with fluorescence in situ hybridization (FISH), next generation sequencing (NGS, n = 6) as well as OncoScan array (n = 3) analyses and show consecutive therapeutic approaches. All seven DSRCT patients presented with an extended abdominal mass; median age at diagnosis was 24.8 years. NGS analyses revealed five class 4 or 5 sequence variants. Remarkably, OncoScan and targeted analyses by FISH identified genomic gains of CCND1 in two cases. Cyclin D1 expression was present in all seven tumors as shown by immunohistochemical staining. Multimodal therapeutic concepts included systemic therapies, resection, and radiation. Six patients were treated as first-line therapy with conventional chemotherapy. All except one patient had a dismal therapy response. Subsequent therapy lines consisted of chemotherapeutic combinations followed by targeted therapies. Due to Cyclin D1 expression, the CDK4/6 inhibitor palbociclib was applied to four patients. The median therapy duration until disease progression in these patients was 4.5 months (range, 1.5-5 months). So, CCND1 genomic gain and Cyclin D1 expression are common features pointing to cell-cycle deregulation as a possible therapeutic target.
Collapse
Affiliation(s)
- Verena I. Gaidzik
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
- Center of Personalised Medicine, University Hospital of Ulm, Ulm, Germany
| | | | - Mathias Wittau
- Department of General and Visceral Surgery, University Hospital of Ulm, Ulm, Germany
| | - Markus Schultheiß
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital of Ulm, Ulm, Germany
| | - Alexandra v. Baer
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital of Ulm, Ulm, Germany
| | - Kathrin Oehl-Huber
- Institute of Human Genetics, University Hospital of Ulm and University of Ulm, Ulm, Germany
| | - Sonja Dahlum
- Institute of Human Genetics, University Hospital of Ulm and University of Ulm, Ulm, Germany
| | - Anja Fischer
- Institute of Human Genetics, University Hospital of Ulm and University of Ulm, Ulm, Germany
| | | | - Thomas Seufferlein
- Center of Personalised Medicine, University Hospital of Ulm, Ulm, Germany
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, University Hospital of Ulm, Ulm, Germany
| | - Wolfgang Thaiss
- Department of Nuclear Medicine, University Hospital of Ulm, Ulm, Germany
| | - Peter Möller
- Institute of Pathology, University of Ulm, Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University Hospital of Ulm and University of Ulm, Ulm, Germany
| | | | | |
Collapse
|
2
|
Oldan JD, Pomper MG, Werner RA, Higuchi T, Rowe SP. The cutting edge: Promising oncology radiotracers in clinical development. Diagn Interv Imaging 2024; 105:400-406. [PMID: 38744576 DOI: 10.1016/j.diii.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/16/2024]
Abstract
Molecular imaging moves forward with the development of new imaging agents, and among these are new radiotracers for nuclear medicine applications, particularly positron emission tomography (PET). A number of new targets are becoming accessible for use in oncologic applications. In this review, major new radiotracers in clinical development are discussed. Prominent among these is the family of fibroblast-activation protein-targeted agents that interact with the tumor microenvironment and may show superiority to 2-deoxy-2-[18F]fluoro-d-glucose in a subset of different tumor histologies. Additionally, carbonic anhydrase IX (CAIX) inhibitors are directed at clear cell renal cell carcinoma, which has long lacked an effective PET imaging agent. Those CAIX agents may also have utility in hypoxic tumors. Pentixafor, which binds to a transmembrane receptor, may similarly allow for visualization by PET of low-grade lymphomas, as well as being a second agent for multiple myeloma that opens theranostic possibilities. There are new adrenergic agents aimed at providing a PET-visible replacement to the single-photon-emitting radiotracer meta-[123I]iodobenzylguanidine (MIBG). Finally, in response to a major development in oncologic chemotherapy, there are new radiotracers targeted at assessing the suitability or use of immunotherapeutic agents. All of these and the existing evidence for their utility are discussed.
Collapse
Affiliation(s)
- Jorge D Oldan
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC 27516, USA
| | - Martin G Pomper
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rudolf A Werner
- Goethe University Frankfurt, University Hospital, Department of Diagnostic and Interventional Radiology and Nuclear Medicine, Division of Nuclear Medicine, 60590 Frankfurt, Germany
| | - Takahiro Higuchi
- Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Steven P Rowe
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC 27516, USA.
| |
Collapse
|
3
|
Crabbé M, Opsomer T, Vermeulen K, Ooms M, Segers C. Targeted radiopharmaceuticals: an underexplored strategy for ovarian cancer. Theranostics 2024; 14:6281-6300. [PMID: 39431018 PMCID: PMC11488094 DOI: 10.7150/thno.99782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/19/2024] [Indexed: 10/22/2024] Open
Abstract
Ovarian cancer is the most common gynecological malignancy worldwide with the highest mortality. This low survival rate can be attributed to the fact that symptoms arise only at an advanced disease stage, characterized by a (micro)metastatic spread across the peritoneal cavity. Radiopharmaceuticals, composed of a targeting moiety coupled with either a diagnostic or therapeutic radionuclide, constitute a relatively underexplored theranostic approach that may improve the current standard of care. Efficient patient stratification, follow-up and treatment are several caveats that could be addressed with theranostics to improve patient outcomes. So far, the bulk of research is situated and often halted at the preclinical level, employing murine models of primary and metastatic peritoneal disease that do not necessarily provide an accurate representation of the disease heterogeneity, (intrinsic) drug resistance or the complex physiological interactions with the tumor microenvironment. Radioimmunoconjugates with therapeutic α- and electron-emitting radionuclides have been the prevailing standard, targeting a myriad of cell-membrane markers that are expressed in the various heterogeneous histological subtypes of ovarian cancer. Evidently, several hurdles exist within preclinical research that are potentially withholding these agents from advancing into clinical practice. On the other hand, the field of nuclear medicine has also seen significant innovation to address shortcomings related to target/ligand identification, preclinical research models, radiochemistry, radiopharmacy and dosimetry, as outlined in this review. Altogether, theranostics hold great promise to answer an unmet medical need for ovarian cancer.
Collapse
Affiliation(s)
| | | | | | | | - Charlotte Segers
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| |
Collapse
|
4
|
Liu M, Chen X, Ding H, Shu Q, Zheng Y, Chen Y, Cai L. Comparison of [ 18F]FDG and [ 68 Ga]pentixafor PET/CT in Nasopharyngeal Carcinoma. Mol Imaging Biol 2024; 26:658-667. [PMID: 38627276 DOI: 10.1007/s11307-024-01913-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/11/2024] [Accepted: 03/30/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE This study aimed to explore the feasibility of [68 Ga]pentixafor positron emission tomography/computed tomography (PET/CT) in patients with nasopharyngeal carcinoma (NPC). PROCEDURES This prospective study included patients with NPC who underwent [68 Ga]pentixafor PET/CT and 2-[18F]fuoro-2-deoxy-D-glucose ([18F]FDG) PET/CT within one week between November 2022 and March 2023. The [68 Ga]pentixafor and [18F]FDG uptakes in primary and metastatic lesions were measured and compared. RESULTS Twenty-five participants (21 patients for initial stage and four patients for recurrence detection) were enrolled in our study. The participants underwent [18F]FDG PET/CT and [68 Ga]pentixafor PET/CT. [68 Ga]pentixafor PET/CT had the same detection rate as [18F]FDG for primary tumor (96% vs. 96%). The [68 Ga]pentixafor maximum standard uptake value (SUVmax) and target-to-background ratio (TBR) of primary tumors were lower than those of [18F]FDG (SUVmax: 8.13 ± 2.78 vs. 14.25 ± 6.45; P < 0.01; TBR: 5.17 ± 2.14 vs. 9.81 ± 5.30, P < 0.01). The difference between tumor volume of [68 Ga]pentixafor (TVpentixafor) and tumor volume of [18F]FDG (TVFDG) showed no significance (median: 16.01 vs. 9.56, P = 0.332). In the detection of suspected metastatic cervical lymph nodes (CLNs), [68 Ga]pentixafor PET possessed a lower SUVmax than [18F]FDG PET/CT (SUVmax: 6.86 ± 2.63 vs. 10.39 ± 5.28, P < 0.01), but there was no significant difference in the detection rate between [68 Ga]pentixafor and [18F]FDG PET/CT (96 vs. 98, P = 0.613). CONCLUSIONS [68 Ga]pentixafor is a promising imaging tracer for detecting primary and metastatic NPC. [68 Ga]pentixafor PET/CT is comparable to [18F]FDG PET/CT in the detection rate of primary tumors and metastatic cervical lymph nodes in nasopharyngeal carcinoma, but [68 Ga]pentixafor uptake was heterogeneous. [68 Ga]pentixafor PET/CT may help select patients most likely to benefit from CXCR4-directed endoradiotherapy. CLINICAL TRIAL REGISTRATION NO ChiCTR2200065902.
Collapse
Affiliation(s)
- Mengna Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xi Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Haoyuan Ding
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Qiaoqiao Shu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Yun Zheng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Liang Cai
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China.
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
5
|
Gäble A, Enke JS, Hügle MJ, Grochowski P, Trepel M, Dierks A, Pfob CH, Bundschuh RA, Lapa C, Kircher M. Disclosing tumor biology by means of molecular imaging in a patient with malignant melanoma and chronic lymphocytic leukemia. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06834-3. [PMID: 39008062 DOI: 10.1007/s00259-024-06834-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024]
Affiliation(s)
- Alexander Gäble
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Johanna S Enke
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Martin J Hügle
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Przemyslaw Grochowski
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Martin Trepel
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Alexander Dierks
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Christian H Pfob
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Ralph A Bundschuh
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Malte Kircher
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| |
Collapse
|
6
|
Hadebe B, Harry L, Gabela L, Masikane S, Patel M, Zwane S, Pillay V, Bipath P, Cebekhulu N, Nyakale N, Ramdass P, Msimang M, Aldous C, Sathekge M, Vorster M. Chemokine Receptor-4 Targeted PET/CT Imaging with 68Ga-Pentixafor in Head and Neck Cancer-A Comparison with 18F-FDG and CXCR4 Immunohistochemistry. Diagnostics (Basel) 2024; 14:1375. [PMID: 39001265 PMCID: PMC11240717 DOI: 10.3390/diagnostics14131375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is common, and its incidence is increasing, particularly in HIV-infected individuals who present with more aggressive disease. Despite aggressive treatment, the prognosis remains poor because of resistance to chemoradiation therapy. So far, studies report very low [68Ga]Ga-Pentixafor avidity in HNSCC. This study investigated the diagnostic performance of CXCR4-directed imaging of carcinoma of the oral cavity, oropharynx, and nasopharynx with positron emission tomography/computed tomography (PET/CT) using the radiolabelled chemokine ligand [68Ga]Ga-Pentixafor and explored its ability to quantify CXCR4 expression in vivo. MATERIALS AND METHODS In this prospective cross-sectional study, twenty-three (23) patients aged 52.9 ± 10.4 (19.6), 17 males and 6 females with primarily diagnosed (n = 17) or pre-treated (n = 6) SCC of the oral cavity (OCSCC, n = 11), oropharynx (OPSCC, n = 9), nasopharynx (NPSCC, n = 2) and unknown primary (n = 1) underwent imaging with [68Ga]Ga-Pentixafor-PET/CT. In 16/23 patients 2-[18F]fluoro-2-deoxy-D-glucose ([18F]F-FDG) served as a standard reference. All lesions were visually rated using a 5-point Likert scale. For both tracers, maximum standardized uptake values (SUVmax) and the total lesion uptake (TLU) were recorded and compared using the Wilcox-signed rank test. In addition, the tumor-to-background ratios were derived using the liver (TLR), spleen (TSR), and posterior cervical muscles (TMR) as background. The relationships between the SUVs of the two tracers were assessed using the Spearman correlation. CXCR4 immunohistochemistry (IHC) staining was correlated with 68Ga-Pentixafor-PET/CT in 21/23 patients. RESULTS Ninety-one percent (21/23) of tumors were visually detected on [68Ga]Ga-Pentixafor; however, [68Ga]Ga-Pentixafor was less intense compared with [18F]F-FDG-PET. Quantitative analysis showed higher [18F]F-FDG SUVmax in comparison with [68Ga]Ga-Pentixafor (16 ± 6.7 vs. 5.8 ± 2.6 g/mL, p = 0.011) and SUVmean (9.3 ± 4.1 vs. 3± 1.6 g/mL, p < 0.001) and TBR 4.9 ± 2.3 vs. 2.36 ± 1.4 p = 0.014. Nasopharyngeal cancer demonstrated more intense tracer accumulation than oropharyngeal and oral cavity malignancies. CXCR4 IHC staining was positive in 15/21 patients, and there was a statistically significant correlation between IHC staining and [68Ga]Ga-Pentixafor SUVmean r = 0.5 p = 0.027, and performance status r = 0.83 p = 0.0104. CONCLUSIONS In conclusion, although [68Ga]Ga-Pentixafor cannot replace [18F]F-FDG as a diagnostic tool because of its lower avidity, the correlation between CXCR4 targeted 68Ga-Pentixafor PET imaging and CXCR4 IHC staining indicates the potential of 68Ga-Pentixafor as an effective tool for selecting patients who may benefit from therapies targeting CXCR4. In addition, [68Ga]Ga-Pentixafor has no physiological brown fat uptake, which often obscures cervical lesions on [18F]F-FDG PET/CT imaging.
Collapse
Affiliation(s)
- Bawinile Hadebe
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Lerwine Harry
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Lerato Gabela
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Siphelele Masikane
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Maryam Patel
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Sizwe Zwane
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Venesen Pillay
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Presha Bipath
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
- Department of Radiation Oncology, College of Health Sciences, University of KwaZulu Natal, Private Bag X03, Durban 4001, South Africa
| | - Nonhlanhla Cebekhulu
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
- Department of Radiation Oncology, College of Health Sciences, University of KwaZulu Natal, Private Bag X03, Durban 4001, South Africa
| | - Nozipho Nyakale
- Department of Nuclear Medicine, Sefako Makgatho Health Science University, Pretoria 0208, South Africa
| | - Prathima Ramdass
- Department of Nuclear Medicine, Jawaharlal Nehru Hospital, Rose Belle 51829, Mauritius
| | - Mpumelelo Msimang
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
- Department of Anatomical Pathology, National Health Laboratory Service, Durban 4000, South Africa
| | - Colleen Aldous
- Department of Genetics, College of Health Sciences, University of KwaZulu Natal, Durban 4001, South Africa
| | - Mike Sathekge
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Mariza Vorster
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| |
Collapse
|
7
|
Sun S, Yang Q, Jiang D, Zhang Y. Nanobiotechnology augmented cancer stem cell guided management of cancer: liquid-biopsy, imaging, and treatment. J Nanobiotechnology 2024; 22:176. [PMID: 38609981 PMCID: PMC11015566 DOI: 10.1186/s12951-024-02432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent both a key driving force and therapeutic target of tumoral carcinogenesis, tumor evolution, progression, and recurrence. CSC-guided tumor diagnosis, treatment, and surveillance are strategically significant in improving cancer patients' overall survival. Due to the heterogeneity and plasticity of CSCs, high sensitivity, specificity, and outstanding targeting are demanded for CSC detection and targeting. Nanobiotechnologies, including biosensors, nano-probes, contrast enhancers, and drug delivery systems, share identical features required. Implementing these techniques may facilitate the overall performance of CSC detection and targeting. In this review, we focus on some of the most recent advances in how nanobiotechnologies leverage the characteristics of CSC to optimize cancer diagnosis and treatment in liquid biopsy, clinical imaging, and CSC-guided nano-treatment. Specifically, how nanobiotechnologies leverage the attributes of CSC to maximize the detection of circulating tumor DNA, circulating tumor cells, and exosomes, to improve positron emission computed tomography and magnetic resonance imaging, and to enhance the therapeutic effects of cytotoxic therapy, photodynamic therapy, immunotherapy therapy, and radioimmunotherapy are reviewed.
Collapse
Affiliation(s)
- Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiang Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
8
|
Michalski K, Schlötelburg W, Hartrampf P, Heinrich M, Serfling S, Buck AK, Werner RA, Kosmala A, Weich A. Volumetric Parameters Derived from CXCR4-Directed PET/CT Predict Outcome in Patients with Gastrointestinal Neuroendocrine Carcinomas. Mol Imaging Biol 2024; 26:344-350. [PMID: 38332341 DOI: 10.1007/s11307-024-01899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Gastro-entero-pancreatic neuroendocrine carcinomas (GEP-NECs) are an aggressive subgroup of neuroendocrine neoplasms (NENs). In patients affected with NEN, there is a growing body of evidence that increased C-X-C motif chemokine receptor (CXCR4) expression is linked to decreasing overall survival (OS) in an ex-vivo setting. Thus, we aimed to determine whether the in-vivo-derived CXCR4-directed whole-body PET signal can also determine GEP-NEC patients with shorter OS. METHODS We retrospectively included 16 patients with histologically proven GEP-NEC, who underwent CXCR4-directed PET/CT for staging and therapy planning. We assessed maximum, peak, and mean standardized uptake values as well as whole-body tumor volume (TV) and total-lesion uptake (TLU = SUVmean × TV) using a semi-automatic segmentation tool with a 50% threshold. Association of PET-based biomarkers and OS or radiographic progression-free survival (rPFS; according to RECIST 1.1 criteria) was analyzed using univariable and multivariable cox regression. RESULTS Median OS and rPFS was 7.5 and 7 months, respectively. A significant correlation between TV and TLU was found for OS (TV: hazard ratio (HR) 1.007 95% confidence interval (CI) 1.000-1.014, p = 0.0309; TLU: HR 1.002 95% CI 1.000-1.003, p = 0.0350) and rPFS (TV: HR 1.010 95% CI 1.002-1.021; p = 0.0275; TLU: HR 1.002 95% CI 1.000-1.004, p = 0.0329), respectively. No significant correlation with OS or rPFS was found for non-volumetric parameters (p > 0.4). TV remained a significant predictive marker for OS and rPFS in multivariable analysis (OS: HR 1.012 95%, CI 1.003-1.022, p = 0.0084; rPFS: HR 1.009, 95% CI 0.9999-1.019, p = 0.0491), whereas TLU remained only prognostic for OS (HR 1.009, 95% CI 0.9999-1.019, p = 0.0194) but narrowly failed significance for rPFS (p = 0.0559). CONCLUSION In-vivo assessment of CXCR4 PET-derived volumetric parameters is predictive for outcome of patients with GEP-NEC and could be used as a risk stratification tool, which detects patients prone to early progression.
Collapse
Affiliation(s)
- Kerstin Michalski
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany.
| | - Wiebke Schlötelburg
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Philipp Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Marieke Heinrich
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Sebastian Serfling
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080, Würzburg, Germany
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, 21218, USA
- Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, University Hospital, Goethe University Frankfurt, 60596, Frankfurt, Germany
| | - Aleksander Kosmala
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Alexander Weich
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080, Würzburg, Germany
- Department of Internal Medicine II, Gastroenterology, University Hospital Würzburg, 97080, Würzburg, Germany
| |
Collapse
|
9
|
Dreher N, Hahner S, Fuß CT, Schlötelburg W, Hartrampf PE, Serfling SE, Schirbel A, Samnick S, Higuchi T, Weich A, Lapa C, Rosenwald A, Buck AK, Kircher S, Werner RA. CXCR4-directed PET/CT with [ 68 Ga]Ga-pentixafor in solid tumors-a comprehensive analysis of imaging findings and comparison with histopathology. Eur J Nucl Med Mol Imaging 2024; 51:1383-1394. [PMID: 38082196 PMCID: PMC10957681 DOI: 10.1007/s00259-023-06547-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 11/26/2023] [Indexed: 03/22/2024]
Abstract
BACKGROUND C-X-C motif chemokine receptor 4 (CXCR4) is overexpressed in various solid cancers and can be targeted by CXCR4-directed molecular imaging. We aimed to characterize the in-vivo CXCR4 expression in patients affected with solid tumors, along with a comparison to ex-vivo findings. METHODS A total 142 patients with 23 different histologically proven solid tumors were imaged with CXCR4-directed PET/CT using [68 Ga]Ga-pentixafor (total number of scans, 152). A semi-quantitative analysis of the CXCR4-positive tumor burden including maximum standardized uptake values (SUVmax) and target-to-background ratios (TBR) using blood pool was conducted. In addition, we performed histopathological staining to determine the immuno-reactive score (IRS) from patients' tumor tissue and investigated possible correlations with SUVmax (by providing Spearman's rho ρ). Based on imaging, we also assessed the eligibility for CXCR4-targeted radioligand therapy or non-radioactive CXCR4 inhibitory treatment (defined as more than five CXCR4-avid target lesions [TL] with SUVmax above 10). RESULTS One hundred three of 152 (67.8%) scans showed discernible uptake above blood pool (TBR > 1) in 462 lesions (52 primary tumors and 410 metastases). Median TBR was 4.4 (1.05-24.98), thereby indicating high image contrast. The highest SUVmax was observed in ovarian cancer, followed by small cell lung cancer, desmoplastic small round cell tumor, and adrenocortical carcinoma. When comparing radiotracer accumulation between primary tumors and metastases for the entire cohort, comparable SUVmax was recorded (P > 0.999), except for pulmonal findings (P = 0.013), indicative for uniform CXCR4 expression among TL. For higher IRS, a weak, but statistically significant correlation with increased SUVmax was observed (ρ = 0.328; P = 0.018). In 42/103 (40.8%) scans, more than five TL were recorded, with 12/42 (28.6%) exhibiting SUVmax above 10, suggesting eligibility for CXCR4-targeted treatment in this subcohort. CONCLUSIONS In a whole-body tumor read-out, a substantial portion of prevalent solid tumors demonstrated increased and uniform [68 Ga]Ga-pentixafor uptake, along with high image contrast. We also observed a respective link between in- and ex-vivo CXCR4 expression, suggesting high specificity of the PET agent. Last, a fraction of patients with [68 Ga]Ga-pentixafor-positive tumor burden were rendered potentially suitable for CXCR4-directed therapy.
Collapse
Affiliation(s)
- Niklas Dreher
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.
| | - Stefanie Hahner
- Department of Internal Medicine I, Endocrinology, University Hospital Würzburg, Würzburg, Germany
| | - Carmina T Fuß
- Department of Internal Medicine I, Endocrinology, University Hospital Würzburg, Würzburg, Germany
| | - Wiebke Schlötelburg
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Sebastian E Serfling
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Alexander Weich
- Department of Internal Medicine II, Gastroenterology, University Hospital Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Stefan Kircher
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, Division of Nuclear Medicine, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| |
Collapse
|
10
|
Manafi-Farid R, Mahini M, Mirshahvalad SA, Fallahi B, Fard-Esfahani A, Emami-Ardekani A, Eftekhari M, Mousavi SA, Beiki D. Diagnostic value of [ 68 Ga]Ga-Pentixafor PET/CT in malignant melanoma: a pilot study. Nucl Med Commun 2024; 45:221-228. [PMID: 38214076 DOI: 10.1097/mnm.0000000000001806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
OBJECTIVE To evaluate the diagnostic value of [ 68 Ga] Ga-Pentixafor in malignant melanoma patients. METHODS In this prospective study, patients with histology-proven melanoma were included and underwent [ 18 F]fluoro-D-glucose ([ 18 F]FDG) and [ 68 Ga] Ga-Pentixafor PET/computed tomography (CT) within a week. Suspicious lesions were interpreted as benign vs. malignant, and the corresponding semi-quantitative PET/CT parameters were recorded and compared. RESULTS Twelve consecutive melanoma patients (mean age: 60 ± 6) were included. Two patients were referred for initial staging, two for detecting recurrence and eight for evaluating the extent of metastases. Overall, [ 18 F]FDG PET/CT showed 236 tumoral lesions, including two primary tumors, two recurrent lesions, 29 locoregional metastases and 203 distant metastases. In [ 68 Ga]Ga-Pentixafor PET/CT, 101 tumoral lesions were detected, including two primary tumors, one recurrence, 16 locoregional metastases and 82 distant metastases. Notably, a documented brain metastasis was only visualized on [ 68 Ga]Ga-Pentixafor PET/CT images. Compared with [ 18 F]FDG, [ 68 Ga]Ga-Pentixafor PET/CT provided a 42% detection rate. Regarding semi-quantitative measures, the intensity of uptake and tumor-to-background ratios were significantly lower on [ 68 Ga]Ga-Pentixafor PET/CT [average maximum standard uptake value (SUV max ) of 2.72 ± 1.33 vs. 11.41 ± 14.79; P value <0.001 and 1.17 ± 0.53 vs. 5.32 ± 7.34; P value <0.001, respectively]. CONCLUSION When comparing [ 68 Ga]Ga-Pentixafor PET/CT with [ 18 F]FDG PET/CT, not only did [ 68 Ga]Ga-Pentixafor PET/CT detect fewer lesions, but the intensity of uptake and the TBRs were also lower on [ 68 Ga]Ga-Pentixafor PET/CT. Thus, our results may indicate a limited potential of this novel tracer in cutaneous melanoma patients compared to [ 18 F]FDG PET/CT. Given the lower TBRs, applying this radiotracer in radioligand therapies is also questionable.
Collapse
Affiliation(s)
| | - Marjan Mahini
- Research Center for Nuclear Medicine, Shariati Hospital
| | | | - Babak Fallahi
- Research Center for Nuclear Medicine, Shariati Hospital
| | | | | | | | - Seied Asadollah Mousavi
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Beiki
- Research Center for Nuclear Medicine, Shariati Hospital
| |
Collapse
|
11
|
Zuo R, Xu L, Pang H. 68Ga-DOTATATE and 68Ga-Pentixafor PET/CT in a Patient with Castleman Disease of the Retroperitoneum. Diagnostics (Basel) 2024; 14:372. [PMID: 38396411 PMCID: PMC10888364 DOI: 10.3390/diagnostics14040372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
This is a case of a 42-year-old man with recurrent symptoms of dizziness and a newly found retroperitoneal mass with no 131I-MIBG uptake who was referred for restaging with 68Ga-DOTATATE PET/CT and local 68Ga-pentixafor PET/CT. The examinations both showed intense radioactivity uptake in the retroperitoneal mass and no abnormal uptake in the right adrenal nodule. Two lesions showed distinct properties of radioactivity uptake, which suggested the possibility of different sources. A postoperative pathological test revealed that the morphology and immunohistochemistry of the retroperitoneal mass was found to be consistent with Castleman disease, and the right adrenal gland was normal tissue.
Collapse
Affiliation(s)
| | | | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Chongqing 400016, China; (R.Z.); (L.X.)
| |
Collapse
|
12
|
Zirakchian Zadeh M. The role of conventional and novel PET radiotracers in assessment of myeloma bone disease. Bone 2024; 179:116957. [PMID: 37972747 DOI: 10.1016/j.bone.2023.116957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Over 80 % of patients with multiple myeloma (MM) experience osteolytic bone lesions, primarily due to an imbalanced interaction between osteoclasts and osteoblasts. This imbalance can lead to several adverse outcomes such as pain, fractures, limited mobility, and neurological impairments. Myeloma bone disease (MBD) raises the expense of management in addition to being a major source of disability and morbidity in myeloma patients. Whole-body x-ray radiography was the gold standard imaging modality for detecting lytic lesions. Osteolytic lesions are difficult to identify at an earlier stage on X-ray since the lesions do not manifest themselves on conventional radiographs until at least 30 % to 50 % of the bone mass has been destroyed. Hence, early diagnosis of osteolytic lesions necessitates the utilization of more complex and advanced imaging modalities, such as PET. One of the PET radiotracers that has been frequently investigated in MM is 18F-FDG, which has demonstrated a high level of sensitivity and specificity in detecting myeloma lesions. However, 18F-FDG PET/CT has several restrictions, and therefore the novel PET tracers that can overcome the limitations of 18F-FDG PET/CT should be further examined in assessment of MBD. The objective of this review article is to thoroughly examine the significance of both conventional and novel PET radiotracers in the assessment of MBD. The intention is to present the information in a manner that would be easily understood by healthcare professionals from diverse backgrounds, while minimizing the use of complex nuclear medicine terminology.
Collapse
Affiliation(s)
- Mahdi Zirakchian Zadeh
- Molecular Imaging and Therapy and Interventional Radiology Services, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
13
|
Lindenberg L, Ahlman M, Lin F, Mena E, Choyke P. Advances in PET Imaging of the CXCR4 Receptor: [ 68Ga]Ga-PentixaFor. Semin Nucl Med 2024; 54:163-170. [PMID: 37923671 PMCID: PMC10792730 DOI: 10.1053/j.semnuclmed.2023.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
[68Ga]Ga-PentixaFor, a PET agent targeting CXCR4 is emerging as a versatile radiotracer with promising applications in oncology, cardiology and inflammatory disease. Preclinical work in various cancer cell lines have demonstrated high specificity and selectivity. In human investigations of several tumors, the most promising applications may be in multiple myeloma, certain lymphomas and myeloproliferative neoplasms. In the nononcologic setting, [68Ga]Ga-PentixaFor could greatly improve detection for primary aldosteronism and other endocrine abnormalities. Similarly, atherosclerotic disease and other inflammatory conditions could also benefit from enhanced identification by CXCR4 targeting. Rapidly cleared from the body with a favorable imaging and radiation dosimetry profile that has been already studied in over 1000 patients, [68Ga]Ga-PentixaFor is a worthy agent for further clinical exploration with potential for theranostic applications in hematologic malignancies.
Collapse
Affiliation(s)
- Liza Lindenberg
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD; Uniformed Services University of the Health Sciences, Bethesda, MD.
| | - Mark Ahlman
- Department of Radiology and Imaging, Medical College of Georgia, Augusta, GA
| | - Frank Lin
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Esther Mena
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Peter Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
14
|
Mirshahvalad SA, Manafi-Farid R, Fallahi B, Seifi S, Geramifar P, Emami-Ardekani A, Eftekhari M, Beiki D. Diagnostic value of [ 68 Ga]Ga-Pentixafor versus [ 18 F]FDG PET/CTs in non-small cell lung cancer: a head-to-head comparative study. Nucl Med Commun 2023; 44:803-809. [PMID: 37334548 DOI: 10.1097/mnm.0000000000001719] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
OBJECTIVE In this study, we aimed to compare the diagnostic value of [ 68 Ga]Ga-Pentixafor and [ 18 F]FDG PET/CT in the evaluation of non-small cell lung cancer (NSCLC) patients. METHODS Patients with pathology-proven NSCLC were prospectively included. Patients underwent [ 18 F]FDG and [ 68 Ga]Ga-Pentixafor PET/CT within 1 week. All suspicious lesions were interpreted as benign or malignant, and the corresponding PET/CT semi-quantitative parameters were recorded. A two-sided P -value <0.05 was considered significant. RESULTS Twelve consecutive NSCLC patients (mean age: 60 ± 7) were included. All patients underwent both [ 18 F]FDG and [ 68 Ga]Ga-Pentixafor PET/CT scans with a median interval of 2 days. Overall, 73 abnormal lesions were detected, from which 58 (79%) were concordant between [ 18 F]FDG and [ 68 Ga]Ga-Pentixafor PET/CT. All primary tumors were clearly detectable in both scans visually. Also, [ 68 Ga]Ga-Pentixafor PET/CT demonstrated rather comparable results with [ 18 F]FDG PET/CT scan in detecting metastatic lesions. However, malignant lesions demonstrated significantly higher SUVmax and SUVmean in [ 18 F]FDG PET/CT ( P -values <0.05). Regarding the advantages, [ 68 Ga]Ga-Pentixafor depicted two brain metastases that were missed by [ 18 F]FDG PET/CT. Also, a highly suspicious lesion for recurrence on [ 18 F]FDG PET/CT scan was correctly classified as benign by subsequent [ 68 Ga]Ga-Pentixafor PET/CT. CONCLUSION [ 68 Ga]Ga-Pentixafor PET/CT was concordant with [ 18 F]FDG PET/CT in detecting primary NSCLC tumors and could visualize the majority of metastatic lesions. Moreover, this modality was found to be potentially helpful in excluding tumoural lesions when the [ 18 F]FDG PET/CT was equivocal, as well as in detecting brain metastasis where [ 18 F]FDG PET/CT suffers from poor sensitivity. However, the count statistics were significantly lower.
Collapse
Affiliation(s)
- Seyed Ali Mirshahvalad
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences
| | - Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences
| | - Babak Fallahi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences
| | - Sharareh Seifi
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences
| | - Alireza Emami-Ardekani
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences
| | - Mohammad Eftekhari
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences
| | - Davood Beiki
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences
| |
Collapse
|
15
|
de Jong D, Desperito E, Al Feghali KA, Dercle L, Seban RD, Das JP, Ma H, Sajan A, Braumuller B, Prendergast C, Liou C, Deng A, Roa T, Yeh R, Girard A, Salvatore MM, Capaccione KM. Advances in PET/CT Imaging for Breast Cancer. J Clin Med 2023; 12:4537. [PMID: 37445572 PMCID: PMC10342839 DOI: 10.3390/jcm12134537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
One out of eight women will be affected by breast cancer during her lifetime. Imaging plays a key role in breast cancer detection and management, providing physicians with information about tumor location, heterogeneity, and dissemination. In this review, we describe the latest advances in PET/CT imaging of breast cancer, including novel applications of 18F-FDG PET/CT and the development and testing of new agents for primary and metastatic breast tumor imaging and therapy. Ultimately, these radiopharmaceuticals may guide personalized approaches to optimize treatment based on the patient's specific tumor profile, and may become a new standard of care. In addition, they may enhance the assessment of treatment efficacy and lead to improved outcomes for patients with a breast cancer diagnosis.
Collapse
Affiliation(s)
- Dorine de Jong
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | | | - Laurent Dercle
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Romain-David Seban
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210 Saint-Cloud, France;
- Laboratory of Translational Imaging in Oncology, Paris Sciences et Lettres (PSL) Research University, Institut Curie, 91401 Orsay, France
| | - Jeeban P. Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (J.P.D.); (R.Y.)
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Abin Sajan
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Brian Braumuller
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Conor Prendergast
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Connie Liou
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Aileen Deng
- Department of Hematology and Oncology, Novant Health, 170 Medical Park Road, Mooresville, NC 28117, USA;
| | - Tina Roa
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (J.P.D.); (R.Y.)
| | - Antoine Girard
- Department of Nuclear Medicine, Centre Eugène Marquis, Université Rennes 1, 35000 Rennes, France;
| | - Mary M. Salvatore
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Kathleen M. Capaccione
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| |
Collapse
|
16
|
Weich A, Serfling SE, Schlötelburg W, Higuchi T, Hartrampf PE, Schirbel A, Heinrich M, Buck AK, Rowe SP, Kosmala A, Werner RA. Impact of CXCR4-Directed PET/CT on Staging and Proposed Oncologic Management in Patients With Digestive System Tumors. Clin Nucl Med 2023; 48:586-593. [PMID: 37167408 DOI: 10.1097/rlu.0000000000004674] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
PURPOSE To elucidate the influence of CXC motif chemokine receptor 4 (CXCR4)-directed imaging on staging and proposed oncologic management in patients with digestive system tumors compared with guideline-appropriate imaging (GAI). METHODS From our PET/CT database, we retrospectively identified 37 patients with advanced digestive system tumors, which had been scheduled for CXCR4-targeted [ 68 Ga]Ga-pentixafor PET/CT for potential theranostic considerations. In all subjects, concurrent GAI was also available. Patients were afflicted with gastroenteropancreatic neuroendocrine neoplasms (21/37 [56.8%]), pancreatic duct adenocarcinoma (6/37 [16.2%]), cholangiocarcinoma (5/37 [13.5%]), hepatocellular carcinoma (4/37 [10.8%]), and colorectal carcinoma (1/37 [2.7%]). Staging results and impact on proposed oncologic management by a board-certified gastroenterologist were compared between GAI and [ 68 Ga]Ga-pentixafor PET/CT. RESULTS Relative to GAI, CXCR4-directed PET/CT resulted in staging changes in 14 of 37 patients (37.8%). Upstaging was seen in 1 of 14 patients (7.1%), whereas downstaging was recorded in the remaining 13 of 14 patients (92.9%). Among those, staging changes would not have triggered any changes in oncological management in 4 of 14 (28.6%). For the remaining 10 of 14 patients (71.4%), however, findings on [ 68 Ga]Ga-pentixafor PET/CT would have impacted subsequent clinical algorithm, including the necessity for further diagnostic steps or failure to initiate antitumor therapy. CONCLUSION [ 68 Ga]Ga-pentixafor PET/CT missed tumor lesions in 13 patients with digestive system tumors, which would have led to inappropriate downstaging and clinical treatment of 10 patients. As such, our results do not support a more widespread use of [ 68 Ga]Ga-pentixafor PET/CT for clinical staging in those tumor entities.
Collapse
Affiliation(s)
| | | | - Wiebke Schlötelburg
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | | | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | | | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Steven P Rowe
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Sciences, Baltimore, MD
| | - Aleksander Kosmala
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | | |
Collapse
|
17
|
Iurchenko NP, Nesina IP, Glushchenko NМ, Buchynska LG. ROLE OF STROMAL MICROENVIRONMENT IN THE FORMATION OF INVASIVE, ANGIOGENIC, AND METASTATIC POTENTIAL OF ENDOMETRIOID CARCINOMA OF ENDOMETRIUM. Exp Oncol 2023; 45:51-61. [PMID: 37417282 DOI: 10.15407/exp-oncology.2023.01.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Indexed: 07/08/2023]
Abstract
The aim of the study was to determine the association of indicators of the progression of endometrioid carcinoma of the endometrium (ECE) with the type of stromal microenvironment, the counts of CXCL12+ fibroblasts and CD163+ macrophages, and the expression of the chemokine CXCL12 and its receptor CXCR4 in tumor cells. MATERIALS AND METHODS Histological preparations of ECE samples (n = 51) were analyzed. Expression of CXCL2 and CXCR4 antigens in tumor cells, the content of CXCL12+ fibroblasts and CD163+ macrophages, and the density of microvessels were determined by the immunohistochemical method. RESULTS Groups of ECE with desmoplastic and inflammatory stromal reactions were delineated. The majority (80.0%) of tumors with desmoplasia were of low differentiation grade, deeply invading the myometrium; 65.0% of patients with these tumors were at stage III of the disease. In ECE cases of stages I-II, 77.4% of ECE showed an inflammatory type of stroma. The high angiogenic and invasive potential of EC of stages I-II was associated with an inflammatory stromal type, high counts of CD163+ macrophages and CXCL12+ fibroblasts in the tumor microenvironment, high expression of the chemokine receptor CXCR4, and reduced expression of its ligand CXCL12 in tumor cells. In the majority of EC of stage III, the increase in angiogenic, invasive, and metastatic potential was accompanied by the presence of desmoplastic stroma, increased expression of CXCR4 in tumor cells, and a high count of CXCL12+ fibroblasts. CONCLUSIONS The obtained results showed that the morphological architecture of the stromal ECE component is related to the molecular features of its constituents and tumor cells. Their interaction modulates the phenotypic characteristics of ECE associated with the degree of malignancy.
Collapse
Affiliation(s)
- N P Iurchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| | - I P Nesina
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| | - N М Glushchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| | - L G Buchynska
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of the National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| |
Collapse
|
18
|
Lepareur N, Ramée B, Mougin-Degraef M, Bourgeois M. Clinical Advances and Perspectives in Targeted Radionuclide Therapy. Pharmaceutics 2023; 15:1733. [PMID: 37376181 DOI: 10.3390/pharmaceutics15061733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted radionuclide therapy has become increasingly prominent as a nuclear medicine subspecialty. For many decades, treatment with radionuclides has been mainly restricted to the use of iodine-131 in thyroid disorders. Currently, radiopharmaceuticals, consisting of a radionuclide coupled to a vector that binds to a desired biological target with high specificity, are being developed. The objective is to be as selective as possible at the tumor level, while limiting the dose received at the healthy tissue level. In recent years, a better understanding of molecular mechanisms of cancer, as well as the appearance of innovative targeting agents (antibodies, peptides, and small molecules) and the availability of new radioisotopes, have enabled considerable advances in the field of vectorized internal radiotherapy with a better therapeutic efficacy, radiation safety and personalized treatments. For instance, targeting the tumor microenvironment, instead of the cancer cells, now appears particularly attractive. Several radiopharmaceuticals for therapeutic targeting have shown clinical value in several types of tumors and have been or will soon be approved and authorized for clinical use. Following their clinical and commercial success, research in that domain is particularly growing, with the clinical pipeline appearing as a promising target. This review aims to provide an overview of current research on targeting radionuclide therapy.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, 35000 Rennes, France
- Inserm, INRAE, Institut NUMECAN (Nutrition, Métabolismes et Cancer)-UMR 1317, Univ Rennes, 35000 Rennes, France
| | - Barthélémy Ramée
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
| | - Marie Mougin-Degraef
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
- Groupement d'Intérêt Public ARRONAX, 1 Rue Aronnax, 44817 Saint Herblain, France
| |
Collapse
|
19
|
Hartrampf PE, Kosmala A, Serfling SE, Bundschuh L, Higuchi T, Lapa C, Rowe SP, Matsusaka Y, Weich A, Buck AK, Bundschuh RA, Werner RA. Interobserver Agreement Rates on C-X-C Motif Chemokine Receptor 4-Directed Molecular Imaging and Therapy. Clin Nucl Med 2023; 48:483-488. [PMID: 36947793 PMCID: PMC10184817 DOI: 10.1097/rlu.0000000000004629] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/06/2023] [Indexed: 03/24/2023]
Abstract
BACKGROUND We aimed to evaluate the interobserver agreement rates in patients scanned with C-X-C motif chemokine receptor 4 (CXCR4)-directed PET/CT, including the rate of patients eligible for CXCR4-targeted radioligand therapy (RLT) based on scan results. METHODS Four independent observers reviewed 50 CXCR4-targeted [ 68 Ga]pentixafor PET/CT of patients with various solid cancers. On a visual level, the following items were assessed by each reader: overall scan impression, number of organ and lymph node (LN) metastases and number of affected organs and LN regions. For a quantitative investigation, readers had to choose a maximum of 3 target lesions, defined as largest in size and/or most intense uptake per organ compartment. Reference tissues were also quantified, including unaffected hepatic parenchyma and blood pool. Last, all observers had to decide whether patients were eligible for CXCR4-targeted RLT. Concordance rates were tested using intraclass correlation coefficients (ICCs). For interpretation, we applied the definition of Cicchetti (with 0.4-0.59 indicating fair; 0.6-0.74, good; 0.75-1, excellent agreement). RESULTS On a visual level, fair agreement was achieved for an overall scan impression (ICC, 0.58; 95% confidence interval, 0.45-0.71). Organ and LN involvement (ICC, ≥0.4) demonstrated fair, whereas CXCR4 density and number of LN and organ metastases showed good agreement rates (ICC, ≥0.65). Number of affected organs and affected LN areas, however, showed excellent concordance (ICC, ≥0.76). Quantification in LN and organ lesions also provided excellent agreement rates (ICC, ≥0.92), whereas quantified uptake in reference organs provided fair concordance (ICC, ≥0.54). Again, excellent agreement rates were observed when deciding on patients eligible for CXCR4-RLT (ICC, 0.91; 95% confidence interval, 0.85-0.95). CONCLUSIONS In patients scanned with CXCR4-targeted PET/CT, we observed fair to excellent agreement rates for both molecular imaging and therapy parameters, thereby favoring a more widespread adoption of [ 68 Ga]pentixafor in the clinic.
Collapse
Affiliation(s)
| | - Aleksander Kosmala
- From the Department of Nuclear Medicine, University Hospital Würzburg, Würzburg
| | | | - Lena Bundschuh
- Nuclear Medicine, Medical Faculty, University Hospital Augsburg, Augsburg, Germany
| | - Takahiro Higuchi
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University Hospital Augsburg, Augsburg, Germany
| | - Steven P. Rowe
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Sciences, Baltimore, MD
| | - Yohji Matsusaka
- From the Department of Nuclear Medicine, University Hospital Würzburg, Würzburg
| | - Alexander Weich
- Internal Medicine II, Gastroenterology, University Hospital Würzburg
- NET-Zentrum Würzburg, European Neuroendocrine Tumor Society Center of Excellence (ENETS CoE), University Hospital Würzburg, Würzburg, Germany
| | - Andreas K. Buck
- From the Department of Nuclear Medicine, University Hospital Würzburg, Würzburg
| | - Ralph A. Bundschuh
- Nuclear Medicine, Medical Faculty, University Hospital Augsburg, Augsburg, Germany
| | - Rudolf A. Werner
- From the Department of Nuclear Medicine, University Hospital Würzburg, Würzburg
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Sciences, Baltimore, MD
- NET-Zentrum Würzburg, European Neuroendocrine Tumor Society Center of Excellence (ENETS CoE), University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
20
|
Maas M, Kurcz A, Hennenlotter J, Scharpf M, Fend F, Walz S, Stühler V, Todenhöfer T, Stenzl A, Bedke J, Rausch S. Differential Expression and Clinical Relevance of C-X-C Motif Chemokine Receptor 4 (CXCR4) in Renal Cell Carcinomas, Benign Renal Tumors, and Metastases. Int J Mol Sci 2023; 24:ijms24065227. [PMID: 36982302 PMCID: PMC10048828 DOI: 10.3390/ijms24065227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/13/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
C-X-C Motif Chemokine Receptor 4 (CXCR4) is part of the human chemokine system and involved in progression and metastasis in renal cell carcinoma (RCC). However, the role of CXCR4 protein expression in RCC remains controversial. In particular, data regarding the subcellular distribution of CXCR4 in RCC and RCC metastasis as well as CXCR4 expression in renal tumors of variant histology are limited. The aim of the present study was the evaluation of the differential CXCR4 expression in RCC primary tumor and metastatic tissue as well as in variant renal histologies. In addition, the prognostic capacity of CXCR4 expression in organ-confined clear cell RCC (ccRCC) was evaluated. Three independent renal tumor cohorts (primary ccRCC cohort n1 = 64; cohort of various histological entities n2 = 146; metastatic RCC tissue cohort n3 = 92) were evaluated using tissue microarrays (TMA). After immunohistochemical staining for CXCR4, nuclear and cytoplasmic expression patterns were evaluated. CXCR4 expression was correlated with validated pathologic prognosticators, clinical data, and overall and cancer-specific survival. Positive cytoplasmic staining was observed in 98% of the benign and 38.9% of the malignant samples. Nuclear staining was positive for 94.1% of the benign samples and 83% of the malignant samples. The median cytoplasmic expression score was found to be higher in benign tissue than in ccRCC (130.00 vs. 0.00); median nuclear expression score analysis indicated the opposite (56.0 vs. 71.0). Within malignant subtypes, the highest expression score was seen in papillary renal cell carcinomas (cytoplasmic: 117.50, nuclear: 41.50). Within benign renal tumors, high cytoplasmic and nuclear CXCR4 expression scores were seen for oncocytomas (cytoplasmic: 100.00, nuclear: 31.00). Expression scores in RCC metastasis ranked between benign renal tissue and ccRCC in cytoplasmic and nuclear expression. Cytoplasmic CXCR4 expression was identified as a prognostic factor for OS and CSS (p = 0.042; p = 0.019). Multivariate analysis including clinicopathological parameters did not reveal an independent prognostic character of CXCR4 expression. CXCR4 expression differs significantly within benign lesions and renal neoplasms. Cytoplasmic and nuclear expression of CXCR4 could be detected across all RCC subtypes. The prognostic value of CXCR4 in ccRCC was confirmed in univariate analysis.
Collapse
Affiliation(s)
- Moritz Maas
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Aymone Kurcz
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Marcus Scharpf
- Department of Pathology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Falko Fend
- Department of Pathology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Simon Walz
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Viktoria Stühler
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Tilman Todenhöfer
- Clinical Trial Unit Studienpraxis Urologie, D-72622 Nürtingen, Germany
- Medical School, Eberhard-Karls-University, D-72076 Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Jens Bedke
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
| | - Steffen Rausch
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler Strasse 3, D-72076 Tübingen, Germany
- Correspondence: ; Tel.: +49-7071-29-86000; Fax: +49-7071-29-5880
| |
Collapse
|
21
|
Watts A, Singh B, Singh H, Bal A, Kaur H, Dhanota N, Arora SK, Mittal BR, Behera D. [ 68Ga]Ga-Pentixafor PET/CT imaging for in vivo CXCR4 receptor mapping in different lung cancer histologic sub-types: correlation with quantitative receptors' density by immunochemistry techniques. Eur J Nucl Med Mol Imaging 2023; 50:1216-1227. [PMID: 36482077 DOI: 10.1007/s00259-022-06059-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE In vivo CXCR4 receptor quantification in different lung cancer (LC) sub-types using [68Ga]Ga-Pentixafor PET/CT and to study correlation with quantitative CXCR4 receptors' tissue density by immunochemistry analyses. METHODS [68Ga]Ga-Pentixafor PET/CT imaging was performed prospectively in 94 (77 M: 17F, mean age 60.1 ± 10.1 years) LC patients. CXCR4 receptors' expression on lung mass in all the patients was estimated by immunohistochemistry (IHC) and fluorescence-activated cell sorting (FACS) analyses. SUVmax on PET, intensity score on IHC, and mean fluorescence index (MFI) on FACS analyses were measured. RESULTS A total of 75/94 (79.8%) cases had non-small cell lung cancer (NSCLC), 14 (14.9%) had small cell lung cancer (SCLC), and 5 (5.3%) had lung neuroendocrine neoplasm (NEN). All LC types showed increased CXCR4 expression on PET (SUVmax) and FACS (MFI). However, both these parameters (mean SUVmax = 10.3 ± 5.0; mean MFI = 349.0 ± 99.0) were significantly (p = 0.005) higher in SCLC as compared to those in NSCLC and lung NEN. The mean SUVmax in adenocarcinoma (n = 16) was 8.0 ± 1.9 which was significantly (p = 0.003) higher than in squamous cell carcinoma (n = 54; 6.2 ± 2.1) and in not-otherwise specified (NOS) sub-types (n = 5; 5.8 ± 1.5) of NSCLC. A significant correlation (r = 0.697; p = 001) was seen between SUVmax and MFI values in squamous cell NSCLC as well as in NSCLC adenocarcinoma (r = 0.538, p = 0.031) which supports the specific in vivo uptake of [68Ga]Ga-Pentixafor by CXCR4 receptors. However, this correlation was not significant in SCLC (r = 0.435, p = 0.121) and NEN (r = 0.747, p = 0.147) which may be due to the small sample size. [68Ga]Ga-Pentixafor PET/CT provided good sensitivity (85.7%) and specificity (78.1%) for differentiating SCLC from NSCLC (ROC cutoff SUVmax = 7.2). This technique presented similar sensitivity (87.5%) and specificity (71.4%) (ROC cutoff SUVmax = 6.7) for differentiating adenocarcinoma and squamous cell variants of NSCLC. CONCLUSION The high sensitivity and specificity of [68Ga]Ga-Pentixafor PET/CT for in vivo targeting of CXCR4 receptors in lung cancer can thus be used effectively for the response assessment and development of CXCR4-based radioligand therapies in LC.
Collapse
Affiliation(s)
- Ankit Watts
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Baljinder Singh
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India.
| | - Harmandeep Singh
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Amanjit Bal
- Department of Histopathology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Harneet Kaur
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Ninjit Dhanota
- Department of Immunopathology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Sunil K Arora
- Department of Immunopathology, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Bhagwant R Mittal
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Digambar Behera
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| |
Collapse
|
22
|
Aboagye EO, Barwick TD, Haberkorn U. Radiotheranostics in oncology: Making precision medicine possible. CA Cancer J Clin 2023; 73:255-274. [PMID: 36622841 DOI: 10.3322/caac.21768] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/10/2022] [Accepted: 11/28/2022] [Indexed: 01/10/2023] Open
Abstract
A quintessential setting for precision medicine, theranostics refers to a rapidly evolving field of medicine in which disease is diagnosed followed by treatment of disease-positive patients using tools for the therapy identical or similar to those used for the diagnosis. Against the backdrop of only-treat-when-visualized, the goal is a high therapeutic index with efficacy markedly surpassing toxicity. Oncology leads the way in theranostics innovation, where the approach has become possible with the identification of unique proteins and other factors selectively expressed in cancer versus healthy tissue, advances in imaging technology able to report these tissue factors, and major understanding of targeting chemicals and nanodevices together with methods to attach labels or warheads for imaging and therapy. Radiotheranostics-using radiopharmaceuticals-is becoming routine in patients with prostate cancer and neuroendocrine tumors who express the proteins PSMA (prostate-specific membrane antigen) and SSTR2 (somatostatin receptor 2), respectively, on their cancer. The palpable excitement in the field stems from the finding that a proportion of patients with large metastatic burden show complete and partial responses, and this outcome is catalyzing the search for more radiotheranostics approaches. Not every patient will benefit from radiotheranostics; but, for those who cross the target-detected line, the likelihood of response is very high.
Collapse
Affiliation(s)
- Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Tara D Barwick
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- Department of Imaging, Imperial College Healthcare National Health Service Trust, Hammersmith Hospital, London, UK
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| |
Collapse
|
23
|
Predicting Microenvironment in CXCR4- and FAP-Positive Solid Tumors-A Pan-Cancer Machine Learning Workflow for Theranostic Target Structures. Cancers (Basel) 2023; 15:cancers15020392. [PMID: 36672341 PMCID: PMC9856808 DOI: 10.3390/cancers15020392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
(1) Background: C-X-C Motif Chemokine Receptor 4 (CXCR4) and Fibroblast Activation Protein Alpha (FAP) are promising theranostic targets. However, it is unclear whether CXCR4 and FAP positivity mark distinct microenvironments, especially in solid tumors. (2) Methods: Using Random Forest (RF) analysis, we searched for entity-independent mRNA and microRNA signatures related to CXCR4 and FAP overexpression in our pan-cancer cohort from The Cancer Genome Atlas (TCGA) database-representing n = 9242 specimens from 29 tumor entities. CXCR4- and FAP-positive samples were assessed via StringDB cluster analysis, EnrichR, Metascape, and Gene Set Enrichment Analysis (GSEA). Findings were validated via correlation analyses in n = 1541 tumor samples. TIMER2.0 analyzed the association of CXCR4 / FAP expression and infiltration levels of immune-related cells. (3) Results: We identified entity-independent CXCR4 and FAP gene signatures representative for the majority of solid cancers. While CXCR4 positivity marked an immune-related microenvironment, FAP overexpression highlighted an angiogenesis-associated niche. TIMER2.0 analysis confirmed characteristic infiltration levels of CD8+ cells for CXCR4-positive tumors and endothelial cells for FAP-positive tumors. (4) Conclusions: CXCR4- and FAP-directed PET imaging could provide a non-invasive decision aid for entity-agnostic treatment of microenvironment in solid malignancies. Moreover, this machine learning workflow can easily be transferred towards other theranostic targets.
Collapse
|
24
|
D’Alterio C, Giardino A, Scognamiglio G, Butturini G, Portella L, Guardascione G, Frigerio I, Montella M, Gobbo S, Martignoni G, Napolitano V, De Vita F, Tatangelo F, Franco R, Scala S. CXCR4-CXCL12-CXCR7 and PD-1/PD-L1 in Pancreatic Cancer: CXCL12 Predicts Survival of Radically Resected Patients. Cells 2022; 11:3340. [PMID: 36359736 PMCID: PMC9655815 DOI: 10.3390/cells11213340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 04/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is currently the most deadly cancer. Although characterized by 5-20% of neoplastic cells in the highly fibrotic stroma, immunotherapy is not a valid option in PDAC treatment. As CXCR4-CXCL12 regulates tumor invasion and T-cell access and PD-1/PD-L1 controls immune tolerance, 76 PDACs were evaluated for CXCR4-CXCL12-CXCR7 and PD-1/PD-L1 in the epithelial and stromal component. Neoplastic CXCR4 and CXCL12 discriminated PDACs for recurrence-free survival (RFS), while CXCL12 and CXCR7 discriminated patients for cancer-specific survival (CSS). Interestingly, among patients with radical resection (R0), high tumor CXCR4 clustered patients with worse RFS, high CXCL12 identified poor prognostic patients for both RFS and CSS, while stromal lymphocytic-monocytic PD-L1 associated with improved RFS and CSS. PD-1 was only sporadically expressed (<1%) in focal lymphocyte infiltrate and does not impact prognosis. In multivariate analysis, tumoral CXCL12, perineural invasion, and AJCC lymph node status were independent prognostic factors for RFS; tumoral CXCL12, AJCC Stage, and vascular invasion were independent prognostic factors for CSS. CXCL12's poor prognostic meaning was confirmed in an additional perspective-independent 13 fine-needle aspiration cytology advanced stage-PDACs. Thus, CXCR4-CXCL12 evaluation in PDAC identifies prognostic categories and could orient therapeutic approaches.
Collapse
Affiliation(s)
- Crescenzo D’Alterio
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Alessandro Giardino
- Unit of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Giosuè Scognamiglio
- Pathology Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giovanni Butturini
- Unit of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giuseppe Guardascione
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Isabella Frigerio
- Unit of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Marco Montella
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Stefano Gobbo
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Guido Martignoni
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Vincenzo Napolitano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Ferdinando De Vita
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Fabiana Tatangelo
- Pathology Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| |
Collapse
|
25
|
Current Status of 68Ga-Pentixafor in Solid Tumours. Diagnostics (Basel) 2022; 12:diagnostics12092135. [PMID: 36140541 PMCID: PMC9497673 DOI: 10.3390/diagnostics12092135] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022] Open
Abstract
Chemokine receptor CXCR4 is overexpressed in neoplasms and its expression is related to tumour invasion, metastasis and aggressiveness. 68Ga-Pentixafor is used to non-invasively image the expression of CXCR4 in tumours and has been widely used in haematological malignancies. Recent evidence shows that therapies targeting CXCR4 can increase the chemosensitivity of the tumour as well as inhibit tumour metastasis and aggressiveness. 68Ga-Pentixafor has shown promise as an elegant radiotracer to aid in the selection of patients whose tumours demonstrate CXCR4 overexpression and who therefore may benefit from novel therapies targeting CXCR4. In addition, its therapeutic partners 177Lu- and 90Y-Pentixather have been investigated in the treatment of patients with advanced haematological malignancies, and initial studies have shown a good treatment response in metabolically active lesions. 68Ga-Pentixafor in solid tumours complements 18F-FDG by providing prognostic information and selecting patients who may benefit from therapies targeting CXCR4. This review summarises the available literature on the potential applications of 68Ga-Pentixafor in solid tumours.
Collapse
|
26
|
Kraus S, Klassen P, Kircher M, Dierks A, Habringer S, Gäble A, Kortüm KM, Weinhold N, Ademaj-Kospiri V, Werner RA, Schirbel A, Buck AK, Herhaus P, Wester HJ, Rosenwald A, Weber WA, Einsele H, Keller U, Rasche L, Lapa C. Reduced splenic uptake on 68Ga-Pentixafor-PET/CT imaging in multiple myeloma - a potential imaging biomarker for disease prognosis. Am J Cancer Res 2022; 12:5986-5994. [PMID: 35966583 PMCID: PMC9373803 DOI: 10.7150/thno.75847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/24/2022] [Indexed: 11/18/2022] Open
Abstract
Beyond being a key factor for tumor growth and metastasis in human cancer, C-X-C motif chemokine receptor 4 (CXCR4) is also highly expressed by a number of immune cells, allowing for non-invasive read-out of inflammatory activity. With two recent studies reporting on prognostic implications of the spleen signal in diffusion-weighted magnetic resonance imaging in patients with plasma cell dyscrasias, the aim of this study was to correlate splenic 68Ga-Pentixafor uptake in multiple myeloma (MM) with clinical parameters and to evaluate its prognostic impact. Methods: Eighty-seven MM patients underwent molecular imaging with 68Ga-Pentixafor-PET/CT. Splenic CXCR4 expression was semi-quantitatively assessed by peak standardized uptake values (SUVpeak) and corresponding spleen-to-bloodpool ratios (TBR) and correlated with clinical and prognostic features as well as survival parameters. Results:68Ga-Pentixafor-PET/CT was visually positive in all MM patients with markedly heterogeneous tracer uptake in the spleen. CXCR4 expression determined by 68Ga-Pentixafor-PET/CT corresponded with advanced disease and was inversely associated with the number of previous treatment lines as compared to controls or untreated smouldering multiple myeloma patients (SUVpeakSpleen 4.06 ± 1.43 vs. 6.02 ± 1.16 vs. 7.33 ± 1.40; P < 0.001). Moreover, reduced splenic 68Ga-Pentixafor uptake was linked to unfavorable clinical outcome. Patients with a low SUVpeakSpleen (<3.35) experienced a significantly shorter overall survival of 5 months as compared to 62 months in patients with a high SUVpeakSpleen >5.79 (P < 0.001). Multivariate Cox analysis confirmed SUVpeakSpleen as an independent predictor of survival (HR 0.75; P = 0.009). Conclusion: These data suggest that splenic 68Ga-Pentixafor uptake might provide prognostic information in pre-treated MM patients similar to what was reported for diffusion-weighted magnetic resonance imaging. Further research to elucidate the underlying biologic implications is warranted.
Collapse
Affiliation(s)
- Sabrina Kraus
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Philipp Klassen
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Malte Kircher
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Alexander Dierks
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Stefan Habringer
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Gäble
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Klaus Martin Kortüm
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Valëza Ademaj-Kospiri
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany.,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Peter Herhaus
- Technical University Munich, School of Medicine, Klinikum rechts der Isar, Clinic and Policlinic for Internal Medicine III, Munich, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technical University of Munich, Munich, Germany
| | | | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
27
|
Ramachandran A, Krishnaraju VS, Kumar R, Shukla J, Laroya I. Ga-68 Chemokine Receptor-4 PET/CT Imaging in Schminke Type of Nasopharyngeal Carcinoma. Indian J Nucl Med 2022; 37:284-285. [PMID: 36686303 PMCID: PMC9855242 DOI: 10.4103/ijnm.ijnm_184_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/29/2022] [Indexed: 11/05/2022] Open
Abstract
Nasopharyngeal carcinoma is a rare malignancy of the head-and-neck region. It is associated with Epstein-Barr virus infection and smoking. Its association with breast cancer is also infrequent. Chemokine receptor (CXCR)-4 imaging is a newer agent for imaging many malignancies with a good diagnostic value. We present a case of a young female diagnosed with left breast carcinoma in whom Ga-68 CXCR-4 PET/CT demonstrated tracer avid lesion in the nasopharynx. Biopsy of the nasopharyngeal lesion revealed Schminke type of lymphoepithelial cancer, indicating CXCR-4 PET/CT as a potential imaging modality for lymphoepithelial malignancy.
Collapse
Affiliation(s)
- Arivan Ramachandran
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Rajender Kumar
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaya Shukla
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ishita Laroya
- Department of General Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
28
|
Abstract
A growing body of literature reports on the upregulation of C-X-C motif chemokine receptor 4 (CXCR4) in a variety of cancer entities, rendering this receptor as suitable target for molecular imaging and endoradiotherapy in a theranostic setting. For instance, the CXCR4-targeting positron emission tomography (PET) agent [68 Ga]PentixaFor has been proven useful for a comprehensive assessment of the current status quo of solid tumors, including adrenocortical carcinoma or small-cell lung cancer. In addition, [68 Ga]PentixaFor has also provided an excellent readout for hematological malignancies, such as multiple myeloma, marginal zone lymphoma, or mantle cell lymphoma. PET-based quantification of the CXCR4 capacities in vivo allows for selecting candidates that would be suitable for treatment using the theranostic equivalent [177Lu]/[90Y]PentixaTher. This CXCR4-directed theranostic concept has been used as a conditioning regimen prior to hematopoietic stem cell transplantation and to achieve sufficient anti-lymphoma/-tumor activity in particular for malignant tissues that are highly sensitive to radiation, such as the hematological system. Increasing the safety margin, pretherapeutic dosimetry is routinely performed to determine the optimal activity to enhance therapeutic efficacy and to reduce off-target adverse events. The present review will provide an overview of current applications for CXCR4-directed molecular imaging and will introduce the CXCR4-targeted theranostic concept for advanced hematological malignancies.
Collapse
|
29
|
Werner RA, Schirbel A, Buck AK, Fassnacht M, Hahner S. Adrenal functional imaging. Presse Med 2022; 51:104114. [PMID: 35131316 DOI: 10.1016/j.lpm.2022.104114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/03/2022] [Accepted: 01/28/2022] [Indexed: 12/16/2022] Open
Abstract
Given the more widespread use of conventional imaging techniques such as magnetic resonance imaging or computed tomography, recent years have witnessed an increased rate of incidental findings in the adrenal gland and those adrenal masses can be either of benign or malignant origin. In this regard, routinely conducted morphological imaging cannot always reliably distinguish between cancerous and noncancerous lesions. As such, those incidental adrenal masses trigger further diagnostic work-up, including molecular functional imaging providing a non-invasive read-out on a sub-cellular level. For instance, [18F]FDG positron emission tomography (PET) as a marker of glucose consumption has been widely utilized to distinguish between malignant vs benign adrenal lesions. In addition, more adrenal cortex-targeted radiotracers for PET or single photon emission computed tomography have entered the clinical arena, e.g., Iodometomidate or IMAZA, which are targeting CYP11B enzymes, or Pentixafor identifying CXCR4 in adrenal tissue. All these tracers are used for diagnosing tumors deriving from the adrenal cortex. Furthermore, radiolabeled MIBG, DOPA, and DOTATOC/-TATE are radiotracers that are quite helpful in detecting pheochromocytomas originating from the adrenal medulla. Of note, after having quantified the retention capacities of the target in-vivo, such radiotracers have the potential to be used as anti-cancer therapeutics by using their therapeutic equivalents in a theranostic setting. The present review will summarize the current advent of established and recently introduced molecular image biomarkers for investigating adrenal masses and highlight its transformation beyond providing functional status towards image-guided therapeutic approaches, in particular in patients afflicted with adrenocortical carcinoma.
Collapse
Affiliation(s)
- Rudolf A Werner
- University Hospital, University of Würzburg, Department of Nuclear Medicine, Germany
| | - Andreas Schirbel
- University Hospital, University of Würzburg, Department of Nuclear Medicine, Germany
| | - Andreas K Buck
- University Hospital, University of Würzburg, Department of Nuclear Medicine, Germany
| | - Martin Fassnacht
- University Hospital, University of Würzburg, Division of Endocrinology and Diabetes, Department of Medicine I, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Stefanie Hahner
- University Hospital, University of Würzburg, Division of Endocrinology and Diabetes, Department of Medicine I, Oberdürrbacher Str. 6, Würzburg 97080, Germany.
| |
Collapse
|
30
|
Establishment of Novel Neuroendocrine Carcinoma Patient-Derived Xenograft Models for Receptor Peptide-Targeted Therapy. Cancers (Basel) 2022; 14:cancers14081910. [PMID: 35454817 PMCID: PMC9033026 DOI: 10.3390/cancers14081910] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 03/30/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Gastroenteropancreatic neuroendocrine neoplasms (GEP NENs) are a family of rare cancers with rising incidence in recent years. GEP NEN tumor cells are difficult to propagate, and few cellular and patient-derived xenograft (PDX) models are available for testing new therapies and studying the heterogeneous nature of these cancers. Here, we described the establishment and characterization of two novel NEC cellular and PDX models (NEC913 and NEC1452). NEC913 PDX tumors express somatostatin receptor 2 (SSTR2), whereas NEC1452 PDX tumors are SSTR2 negative. As a proof-of-concept study, we demonstrated how these PDX models can be used for peptide imaging experiments targeting SSTR2 using fluorescently labelled octreotide. The NEC913 and NEC1452 PDX lines represent valuable new tools for accelerating the process of drug discovery for GEP NENs. Abstract Gastroenteropancreatic neuroendocrine neoplasms (GEP NENs) are rare cancers consisting of neuroendocrine carcinomas (NECs) and neuroendocrine tumors (NETs), which have been increasing in incidence in recent years. Few cell lines and pre-clinical models exist for studying GEP NECs and NETs, limiting the ability to discover novel imaging and treatment modalities. To address this gap, we isolated tumor cells from cryopreserved patient GEP NECs and NETs and injected them into the flanks of immunocompromised mice to establish patient-derived xenograft (PDX) models. Two of six mice developed tumors (NEC913 and NEC1452). Over 80% of NEC913 and NEC1452 tumor cells stained positive for Ki67. NEC913 PDX tumors expressed neuroendocrine markers such as chromogranin A (CgA), synaptophysin (SYP), and somatostatin receptor-2 (SSTR2), whereas NEC1452 PDX tumors did not express SSTR2. Exome sequencing revealed loss of TP53 and RB1 in both NEC tumors. To demonstrate an application of these novel NEC PDX models for SSTR2-targeted peptide imaging, the NEC913 and NEC1452 cells were bilaterally injected into mice. Near infrared-labelled octreotide was administered and the fluorescent signal was specifically observed for the NEC913 SSTR2 positive tumors. These 2 GEP NEC PDX models serve as a valuable resource for GEP NEN therapy testing.
Collapse
|
31
|
Serfling SE, Lapa C, Dreher N, Hartrampf PE, Rowe SP, Higuchi T, Schirbel A, Weich A, Hahner S, Fassnacht M, Buck AK, Werner RA. Impact of Tumor Burden on Normal Organ Distribution in Patients Imaged with CXCR4-Targeted [68Ga]Ga-PentixaFor PET/CT. Mol Imaging Biol 2022; 24:659-665. [PMID: 35312939 PMCID: PMC9296404 DOI: 10.1007/s11307-022-01717-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 12/16/2022]
Abstract
Abstract
Background
CXCR4-directed positron emission tomography/computed tomography (PET/CT) has been used as a diagnostic tool in patients with solid tumors. We aimed to determine a potential correlation between tumor burden and radiotracer accumulation in normal organs.
Methods
Ninety patients with histologically proven solid cancers underwent CXCR4-targeted [68Ga]Ga-PentixaFor PET/CT. Volumes of interest (VOIs) were placed in normal organs (heart, liver, spleen, bone marrow, and kidneys) and tumor lesions. Mean standardized uptake values (SUVmean) for normal organs were determined. For CXCR4-positive tumor burden, maximum SUV (SUVmax), tumor volume (TV), and fractional tumor activity (FTA, defined as SUVmean x TV), were calculated. We used a Spearman's rank correlation coefficient (ρ) to derive correlative indices between normal organ uptake and tumor burden.
Results
Median SUVmean in unaffected organs was 5.2 for the spleen (range, 2.44 – 10.55), 3.27 for the kidneys (range, 1.52 – 17.4), followed by bone marrow (1.76, range, 0.84 – 3.98), heart (1.66, range, 0.88 – 2.89), and liver (1.28, range, 0.73 – 2.45). No significant correlation between SUVmax in tumor lesions (ρ ≤ 0.189, P ≥ 0.07), TV (ρ ≥ -0.204, P ≥ 0.06) or FTA (ρ ≥ -0.142, P ≥ 0.18) with the investigated organs was found.
Conclusions
In patients with solid tumors imaged with [68Ga]Ga-PentixaFor PET/CT, no relevant tumor sink effect was noted. This observation may be of relevance for therapies with radioactive and non-radioactive CXCR4-directed drugs, as with increasing tumor burden, the dose to normal organs may remain unchanged.
Collapse
|
32
|
Lepareur N. Cold Kit Labeling: The Future of 68Ga Radiopharmaceuticals? Front Med (Lausanne) 2022; 9:812050. [PMID: 35223907 PMCID: PMC8869247 DOI: 10.3389/fmed.2022.812050] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Over the last couple of decades, gallium-68 (68Ga) has gained a formidable interest for PET molecular imaging of various conditions, from cancer to infection, through cardiac pathologies or neuropathies. It has gained routine use, with successful radiopharmaceuticals such as somatostatin analogs ([68Ga]Ga-DOTATOC and [68Ga]GaDOTATATE) for neuroendocrine tumors, and PSMA ligands for prostate cancer. It represents a major clinical impact, particularly in the context of theranostics, coupled with their 177Lu-labeled counterparts. Beside those, a bunch of new 68Ga-labeled molecules are in the preclinical and clinical pipelines, with some of them showing great promise for patient care. Increasing clinical demand and regulatory issues have led to the development of automated procedures for the production of 68Ga radiopharmaceuticals. However, the widespread use of these radiopharmaceuticals may rely on simple and efficient radiolabeling methods, undemanding in terms of equipment and infrastructure. To make them technically and economically accessible to the medical community and its patients, it appears mandatory to develop a procedure similar to the well-established kit-based 99mTc chemistry. Already available commercial kits for the production of 68Ga radiopharmaceuticals have demonstrated the feasibility of using such an approach, thus paving the way for more kit-based 68Ga radiopharmaceuticals to be developed. This article discusses the development of 68Ga cold kit radiopharmacy, including technical issues, and regulatory aspects.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, Rennes, France
- Univ Rennes, Inrae, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer), UMR_A 1341, UMR_S 1241, Rennes, France
| |
Collapse
|
33
|
Liu J, Ren WX, Shu J. Multimodal molecular imaging evaluation for early diagnosis and prognosis of cholangiocarcinoma. Insights Imaging 2022; 13:10. [PMID: 35050416 PMCID: PMC8776965 DOI: 10.1186/s13244-021-01147-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 12/15/2021] [Indexed: 11/23/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive and lethal malignancy with limited therapeutic options. Despite recent advances in diagnostic imaging for CCA, the early diagnosis of CCA and evaluation of tumor invasion into the bile duct and its surrounding tissues remain challenging. Most patients with CCA are diagnosed at an advanced stage, at which treatment options are limited. Molecular imaging is a promising diagnostic method for noninvasive imaging of biological events at the cellular and molecular level in vivo. Molecular imaging plays a key role in the early diagnosis, staging, and treatment-related evaluation and management of cancer. This review will describe different methods for molecular imaging of CCA, including nuclear medicine, magnetic resonance imaging, optical imaging, and multimodal imaging. The main challenges and future directions in this field are also discussed.
Collapse
Affiliation(s)
- Jiong Liu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No 25 Taiping St, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China
| | - Wen Xiu Ren
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No 25 Taiping St, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jian Shu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China. .,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No 25 Taiping St, Jiangyang District, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
34
|
Schottelius M, Herrmann K, Lapa C. In Vivo Targeting of CXCR4-New Horizons. Cancers (Basel) 2021; 13:5920. [PMID: 34885030 PMCID: PMC8656854 DOI: 10.3390/cancers13235920] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/23/2023] Open
Abstract
Given its pre-eminent role in the context of tumor cell growth as well as metastasis, the C-X-C motif chemokine receptor 4 (CXCR4) has attracted a lot of interest in the field of nuclear oncology, and clinical evidence on the high potential of CXCR4-targeted theranostics is constantly accumulating. Additionally, since CXCR4 also represents a key player in the orchestration of inflammatory responses to inflammatory stimuli, based on its expression on a variety of pro- and anti-inflammatory immune cells (e.g., macrophages and T-cells), CXCR4-targeted inflammation imaging has recently gained considerable attention. Therefore, after briefly summarizing the current clinical status quo of CXCR4-targeted theranostics in cancer, this review primarily focuses on imaging of a broad spectrum of inflammatory diseases via the quantification of tissue infiltration with CXCR4-expressing immune cells. An up-to-date overview of the ongoing preclinical and clinical efforts to visualize inflammation and its resolution over time is provided, and the predictive value of the CXCR4-associated imaging signal for disease outcome is discussed. Since the sensitivity and specificity of CXCR4-targeted immune cell imaging greatly relies on the availability of suitable, tailored imaging probes, recent developments in the field of CXCR4-targeted imaging agents for various applications are also addressed.
Collapse
Affiliation(s)
- Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, German Cancer Consortium (DKTK)-University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| |
Collapse
|
35
|
Lewis R, Habringer S, Kircher M, Hefter M, Peuker CA, Werner R, Ademaj-Kospiri V, Gäble A, Weber W, Wester HJ, Buck A, Herhaus P, Lapa C, Keller U. Investigation of spleen CXCR4 expression by [ 68Ga]Pentixafor PET in a cohort of 145 solid cancer patients. EJNMMI Res 2021; 11:77. [PMID: 34417915 PMCID: PMC8380222 DOI: 10.1186/s13550-021-00822-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/07/2021] [Indexed: 02/08/2023] Open
Abstract
Background The chemokine receptor CXCR4 is frequently overexpressed and associated with adverse prognosis in most hematopoietic malignancies and solid cancers. Recently, CXCR4 molecular imaging using the CXCR4-specific positron emission tomography (PET) tracer Pentixafor ([68Ga]Pentixafor) has become a well-established method to non-invasively measure CXCR4 expression in vivo. In previous Pentixafor imaging studies, highly variable CXCR4 tracer uptake to the spleen was observed.
Results We investigated the hypothesis that enhanced spleen [68Ga]Pentixafor uptake and thus CXCR4 expression in patients with solid tumors would indicate an activated spleen state and/or an association with clinical and prognostic features and survival parameters. In this retrospective study, [68Ga]Pentixafor-PET images and patient records of 145 solid tumor patients representing 27 cancer entities were investigated for an association of spleen [68Ga]Pentixafor uptake and clinical characteristics and outcome. Based on this assessment, we did not observe differences in clinical outcomes, measured by progression-free survival, overall survival and remission status neither within the entire cohort nor within subgroups of adrenal cancer, desmoplastic small round cell tumor, neuroendocrine tumors, non-small cell lung cancer, small cell lung cancer and pancreatic adenocarcinoma patients. No tumor entity showed especially high levels of spleen [68Ga]Pentixafor uptake compared to others or a control cohort. However, when investigating laboratory parameters, there was a positive correlation of high spleen [68Ga]Pentixafor uptake with leukocyte and/or platelet counts in neuroendocrine tumors, non-small cell lung cancer and small cell lung cancer. Conclusion Spleen [68Ga]Pentixafor uptake was not associated with stage of disease and clinical outcomes in solid tumor patients. We identified positively associated platelet and/or leukocyte counts with spleen [68Ga]Pentixafor uptake in neuroendocrine tumors, non-small cell lung cancer and small cell lung cancer, suggesting that splenic CXCR4 expression could possibly play a role in systemic immunity/inflammation in some types of solid tumors or a subgroup of patients within solid tumor entities. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00822-6.
Collapse
Affiliation(s)
- Richard Lewis
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Stefan Habringer
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Malte Kircher
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Maike Hefter
- Clinic and Policlinic for Internal Medicine III, School of Medicine, Technical University of Munich, Munich, Germany
| | - Caroline Anna Peuker
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Rudolf Werner
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Valëza Ademaj-Kospiri
- Clinic for Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Gäble
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Wolfgang Weber
- Clinic for Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hans-Jürgen Wester
- Chair of Pharmaceutical Chemistry, Technical University of Munich, Garching, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Peter Herhaus
- Clinic and Policlinic for Internal Medicine III, School of Medicine, Technical University of Munich, Munich, Germany
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany. .,German Cancer Consortium (DKTK), Partner Site Berlin; and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
36
|
Weich A, Werner RA, Buck AK, Hartrampf PE, Serfling SE, Scheurlen M, Wester HJ, Meining A, Kircher S, Higuchi T, Pomper MG, Rowe SP, Lapa C, Kircher M. CXCR4-Directed PET/CT in Patients with Newly Diagnosed Neuroendocrine Carcinomas. Diagnostics (Basel) 2021; 11:diagnostics11040605. [PMID: 33805264 PMCID: PMC8067200 DOI: 10.3390/diagnostics11040605] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 01/29/2023] Open
Abstract
We aimed to elucidate the diagnostic potential of the C-X-C motif chemokine receptor 4 (CXCR4)-directed positron emission tomography (PET) tracer 68Ga-Pentixafor in patients with poorly differentiated neuroendocrine carcinomas (NEC), relative to the established reference standard 18F-FDG PET/computed tomography (CT). In our database, we retrospectively identified 11 treatment-naïve patients with histologically proven NEC, who underwent 18F-FDG and CXCR4-directed PET/CT for staging and therapy planning. The images were analyzed on a per-patient and per-lesion basis and compared to immunohistochemical staining (IHC) of CXCR4 from PET-guided biopsies. 68Ga-Pentixafor visualized tumor lesions in 10/11 subjects, while18F-FDG revealed sites of disease in all 11 patients. Although weak to moderate CXCR4 expression could be corroborated by IHC in 10/11 cases, 18F-FDG PET/CT detected significantly more tumor lesions (102 vs. 42; total lesions, n = 107; p < 0.001). Semi-quantitative analysis revealed markedly higher 18F-FDG uptake as compared to 68Ga-Pentixafor (maximum and mean standardized uptake values (SUV) and tumor-to-background ratios (TBR) of cancerous lesions, SUVmax: 12.8 ± 9.8 vs. 5.2 ± 3.7; SUVmean: 7.4 ± 5.4 vs. 3.1 ± 3.2, p < 0.001; and, TBR 7.2 ± 7.9 vs. 3.4 ± 3.0, p < 0.001). Non-invasive imaging of CXCR4 expression in NEC is inferior to the reference standard 18F-FDG PET/CT.
Collapse
Affiliation(s)
- Alexander Weich
- Department of Internal Medicine I, Gastroenterology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.W.); (M.S.); (A.M.)
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080 Würzburg, Germany; (R.A.W.); (A.K.B.); (C.L.)
| | - Rudolf A. Werner
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080 Würzburg, Germany; (R.A.W.); (A.K.B.); (C.L.)
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (P.E.H.); (S.E.S.); (T.H.)
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.G.P.); (S.P.R.)
| | - Andreas K. Buck
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080 Würzburg, Germany; (R.A.W.); (A.K.B.); (C.L.)
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (P.E.H.); (S.E.S.); (T.H.)
| | - Philipp E. Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (P.E.H.); (S.E.S.); (T.H.)
| | - Sebastian E. Serfling
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (P.E.H.); (S.E.S.); (T.H.)
| | - Michael Scheurlen
- Department of Internal Medicine I, Gastroenterology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.W.); (M.S.); (A.M.)
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080 Würzburg, Germany; (R.A.W.); (A.K.B.); (C.L.)
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, 80333 München, Germany;
| | - Alexander Meining
- Department of Internal Medicine I, Gastroenterology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.W.); (M.S.); (A.M.)
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080 Würzburg, Germany; (R.A.W.); (A.K.B.); (C.L.)
| | - Stefan Kircher
- Institute of Pathology, University of Würzburg, 97080 Würzburg, Germany;
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (P.E.H.); (S.E.S.); (T.H.)
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
- Comprehensive Heart Failure Center, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.G.P.); (S.P.R.)
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (M.G.P.); (S.P.R.)
| | - Constantin Lapa
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080 Würzburg, Germany; (R.A.W.); (A.K.B.); (C.L.)
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (P.E.H.); (S.E.S.); (T.H.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Malte Kircher
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, NET Zentrum, University Hospital Würzburg, 97080 Würzburg, Germany; (R.A.W.); (A.K.B.); (C.L.)
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (P.E.H.); (S.E.S.); (T.H.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
- Correspondence:
| |
Collapse
|
37
|
Linde P, Baues C, Wegen S, Trommer M, Quaas A, Rosenbrock J, Celik E, Marnitz S, Bruns CJ, Fischer T, Schomaecker K, Wester HJ, Drzezga A, van Heek L, Kobe C. Pentixafor PET/CT for imaging of chemokine receptor 4 expression in esophageal cancer - a first clinical approach. Cancer Imaging 2021; 21:22. [PMID: 33579381 PMCID: PMC7881561 DOI: 10.1186/s40644-021-00391-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/05/2021] [Indexed: 12/24/2022] Open
Abstract
Background Expression of CXCR4, a chemokine (C-X-C motif) receptor that plays a central role in tumor growth and metastasis of circulating tumor cells, has been described in a variety of solid tumors. A high expression of CXCR4 has a prognostic significance with regard to overall and progression-free survival and offers a starting point for targeted therapies. In this context, [68]Ga-Pentixafor-Positron Emission Tomography/Computer Tomography (PET/CT) offers promising possibility of imaging the CXCR4 expression profile. We set out to compare a [18F] fluorodeoxyglucose (FDG)-PET/CT and a [68Ga]Pentixafor-PET/CT in (re-)staging and radiation planning of patients with localized esophageal cancer. Materials and methods In this retrospective analysis, ten patients, with adeno- or squamous cell carcinoma of the esophagus (n = 3 and n = 7, respectively), which were scheduled for radio (chemo) therapy, were imaged using both Pentixafor and FDG PET/CT examinations. All lesions were visually rated as Pentixafor and FDG positive or negative. For both tracers, SUVmax was measured all lesions and compared to background. Additionally, immunohistochemistry of CXCR4 was obtained in patients undergoing surgery. Results FDG-positive tumor-suspicious lesions were detected in all patients and a total of 26 lesions were counted. The lesion-based analysis brought equal status in 14 lesions which were positive for both tracers while five lesions were FDG positive and Pentixafor negative and seven lesions were FDG negative, but Pentixafor positive. Histopathologic correlation was available in seven patients. The CXCR4 expression of four non-pretreated tumour lesion samples was confirmed immunohistochemically. Conclusion Our data shows that additional PET/CT imaging with Pentixafor for imaging the CXCR4 chemokine receptor is feasible but heterogeneous in both newly diagnosed and pretreated recurrent esophageal cancer. In addition, the Pentixafor PET/CT may serve as complementary tool for radiation field expansion in radiooncology. Supplementary Information The online version contains supplementary material available at 10.1186/s40644-021-00391-w.
Collapse
Affiliation(s)
- Philipp Linde
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany.
| | - Christian Baues
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Simone Wegen
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Maike Trommer
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Alexander Quaas
- Department of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Johannes Rosenbrock
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Eren Celik
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Simone Marnitz
- Department of Radiation Oncology, University Hospital of Cologne, University of Cologne, Kerpener St 62, 50937, Cologne, Germany
| | - Christiane J Bruns
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Thomas Fischer
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Klaus Schomaecker
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Hans-Juergen Wester
- Department of Radiochemistry, Technische Universität München, Garching, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Lutz van Heek
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
38
|
Yordanova A, Biersack HJ, Ahmadzadehfar H. Advances in Molecular Imaging and Radionuclide Therapy of Neuroendocrine Tumors. J Clin Med 2020; 9:E3679. [PMID: 33207788 PMCID: PMC7697910 DOI: 10.3390/jcm9113679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroendocrine neoplasms make up a heterogeneous group of tumors with inter-patient and intra-patient variabilities. Molecular imaging can help to identify and characterize neuroendocrine tumors (NETs). Furthermore, imaging and treatment with novel theranostics agents offers a new, tailored approach to managing NETs. Recent advances in the management of NETs aim to enhance the effectiveness of targeted treatment with either modifications of known substances or the development of new substances with better targeting features. There have been several attempts to increase the detectability of NET lesions via positron emission tomography (PET) imaging and improvements in pretreatment planning using dosimetry. Especially notable is PET imaging with the radionuclide Copper-64. Increasing interest is also being paid to theranostics of grade 3 and purely differentiated NETs, for example, via targeting of the C-X-C motif chemokine receptor 4 (CXCR4). The aim of this review is to summarize the most relevant recent studies, which present promising new agents in molecular imaging and therapy for NETs, novel combination therapies and new applications of existing molecular imaging modalities in nuclear medicine.
Collapse
Affiliation(s)
- Anna Yordanova
- Department of Radiology, St. Marien Hospital Bonn, 53115 Bonn, Germany;
| | | | | |
Collapse
|
39
|
Si Y, Guan J, Xu Y, Chen K, Kim S, Zhou L, Jaskula-Sztul R, Liu XM. Dual-Targeted Extracellular Vesicles to Facilitate Combined Therapies for Neuroendocrine Cancer Treatment. Pharmaceutics 2020; 12:E1079. [PMID: 33187322 PMCID: PMC7696983 DOI: 10.3390/pharmaceutics12111079] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022] Open
Abstract
Neuroendocrine (NE) cancers arise from cells within the neuroendocrine system. Chemotherapies and endoradiotherapy have been developed, but their clinical efficacy is limited. The objective of this study was to develop a dual-targeted extracellular vesicles (EV)-delivered combined therapies to treat NE cancer. Specifically, we produced EV in stirred-tank bioreactors and surface tagged both anti-somatostatin receptor 2 (SSTR 2) monoclonal antibody (mAb) and anti-C-X-C motif chemokine receptor 4 (CXCR4) mAb to generate mAbs-EV. Both live-cell confocal microscopy imaging and In Vivo Imaging System (IVIS) imaging confirmed that mAbs-EV specifically targeted and accumulated in NE cancer cells and NE tumor xenografts. Then the highly potent natural cytotoxic marine compound verrucarin A (Ver-A) with IC50 of 2.2-2.8 nM and microtubule polymerization inhibitor mertansine (DM1) with IC50 of 3.1-4.2 nM were packed into mAbs-EV. The in vivo maximum tolerated dose study performed in non-tumor-bearing mice indicated minimal systemic toxicity of mAbs-EV-Ver-A/DM1. Finally, the in vivo anticancer efficacy study demonstrated that the SSTR2/CXCR4 dual-targeted EV-Ver-A/DM1 is more effective to inhibit NE tumor growth than the single targeting and single drug. The results from this study could expand the application of EV to targeting deliver the combined potent chemotherapies for cancer treatment.
Collapse
Affiliation(s)
- Yingnan Si
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA; (Y.S.); (J.G.); (Y.X.); (K.C.); (S.K.)
| | - JiaShiung Guan
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA; (Y.S.); (J.G.); (Y.X.); (K.C.); (S.K.)
| | - Yuanxin Xu
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA; (Y.S.); (J.G.); (Y.X.); (K.C.); (S.K.)
| | - Kai Chen
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA; (Y.S.); (J.G.); (Y.X.); (K.C.); (S.K.)
| | - Seulhee Kim
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA; (Y.S.); (J.G.); (Y.X.); (K.C.); (S.K.)
| | - Lufang Zhou
- Department of Medicine, University of Alabama at Birmingham, 703 19th Street South, Birmingham, AL 35294, USA;
| | - Renata Jaskula-Sztul
- Department of Surgery, University of Alabama at Birmingham, 1808 7th Avenue South, Birmingham, AL 35294, USA;
| | - X. Margaret Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA; (Y.S.); (J.G.); (Y.X.); (K.C.); (S.K.)
| |
Collapse
|
40
|
Panda A, Goenka AH, Hope TA, Veit-Haibach P. PET/Magnetic Resonance Imaging Applications in Abdomen and Pelvis. Magn Reson Imaging Clin N Am 2020; 28:369-380. [DOI: 10.1016/j.mric.2020.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
41
|
Osl T, Schmidt A, Schwaiger M, Schottelius M, Wester HJ. A new class of PentixaFor- and PentixaTher-based theranostic agents with enhanced CXCR4-targeting efficiency. Am J Cancer Res 2020; 10:8264-8280. [PMID: 32724470 PMCID: PMC7381729 DOI: 10.7150/thno.45537] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Non-invasive PET imaging of CXCR4 expression in cancer and inflammation as well as CXCR4-targeted radioligand therapy (RLT) have recently found their way into clinical research by the development of the theranostic agents [68Ga]PentixaFor (cyclo(D-Tyr1-D-[NMe]Orn2(AMBS-[68Ga]DOTA)-Arg3-Nal4-Gly5) = [68Ga]DOTA-AMBS-CPCR4) and [177Lu/90Y]PentixaTher (cyclo(D-3-iodo-Tyr1-D-[NMe]Orn2(AMBS-[177Lu/90Y]DOTA)-Arg3-Nal4-Gly5) = [177Lu/90Y]DOTA-AMBS-iodoCPCR4). Although convincing clinical results have already been obtained with both agents, this study was designed to further investigate the required structural elements for improved ligand-receptor interaction for both peptide cores (CPCR4 and iodoCPCR4). To this aim, a series of DOTA-conjugated CPCR4- and iodoCPCR4-based ligands with new linker structures, replacing the AMBA-linker in PentixaFor and PentixaTher, were synthesized and evaluated. Methods: The in vitro investigation of the novel compounds alongside with the reference peptides PentixaFor and PentixaTher encompassed the determination of hCXCR4 and mCXCR4 affinity (IC50) of the respective natGa-, natLu-, natY- and natBi-complexes in Jurkat and Eμ-myc 1080 cells using [125I]FC-131 and [125I]CPCR4.3 as radioligands, respectively, as well as the evaluation of the internalization and externalization kinetics of selected 68Ga- and 177Lu-labeled compounds in hCXCR4-transfected Chem-1 cells. Comparative small animal PET imaging studies (1h p.i.) as well as in vivo biodistribution studies (1, 6 and 48h p.i.) were performed in Daudi (human B cell lymphoma) xenograft bearing CB17 SCID mice. Results: Based on the affinity data and cellular uptake studies, [68Ga/177Lu]DOTA-r-a-ABA-CPCR4 and [68Ga/177Lu]DOTA-r-a-ABA-iodoCPCR4 (with r-a-ABA = D-Arg-D-Ala-4-aminobenzoyl-) were selected for further evaluation. Both analogs show app. 10-fold enhanced hCXCR4 affinity compared to the respective references [68Ga]PentixaFor and [177Lu]PentixaTher, four times higher cellular uptake in hCXCR4 expressing cells and improved cellular retention. Unfortunately, the improved in vitro binding and uptake characteristics of [68Ga]DOTA-r-a-ABA-CPCR4 and -iodoCPCR4 could not be recapitulated in initial PET imaging studies; both compounds showed similar uptake in the Daudi xenografts as [68Ga]PentixaFor, alongside with higher background accumulation, especially in the kidneys. However, the subsequent biodistribution studies performed for the corresponding 177Lu-labeled analogs revealed a clear superiority of [177Lu]DOTA-r-a-ABA-CPCR4 and [177Lu]DOTA-r-a-ABA-iodoCPCR4 over [177Lu]PentixaTher with respect to tumor uptake (18.3±3.7 and 17.2±2.0 %iD/g, respectively, at 1h p.i. vs 12.4±3.7%iD/g for [177Lu]PentixaTher) as well as activity retention in tumor up to 48h. Especially for [177Lu]DOTA-r-a-ABA-CPCR4 with its low background accumulation, tumor/organ ratios at 48h were 2- to 4-fold higher than those obtained for [177Lu]PentixaTher (except for kidney). Conclusions: The in-depth evaluation of a series of novel CPCR4- and iodoCPCR4 analogs with modified linker structure has yielded reliable structure-activity relationships. It was generally observed that a) AMBA-by-ABA-substitution leads to enhanced ligand internalization, b) the extension of the ABA-linker by two additional amino acids (DOTA-Xaa2-Xaa1-ABA-) provides sufficient linker length to minimize the interaction of the [M3+]DOTA-chelate with the receptor, and that c) introduction of a cationic side chain (Xaa2) greatly enhances receptor affinity of the constructs, obliterating the necessity for Tyr1-iodination of the pentapeptide core to maintain high receptor affinity (such as in [177Lu]PentixaTher). As a result, [177Lu]DOTA-r-a-ABA-CPCR4 has emerged from this study as a powerful second-generation therapeutic CXCR4 ligand with greatly improved targeting efficiency and tumor retention and will be further evaluated in preclinical and clinical CXCR4-targeted dosimetry and RLT studies.
Collapse
|
42
|
|
43
|
Rangger C, Haubner R. Radiolabelled Peptides for Positron Emission Tomography and Endoradiotherapy in Oncology. Pharmaceuticals (Basel) 2020; 13:E22. [PMID: 32019275 PMCID: PMC7169460 DOI: 10.3390/ph13020022] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
This review deals with the development of peptide-based radiopharmaceuticals for the use with positron emission tomography and peptide receptor radiotherapy. It discusses the pros and cons of this class of radiopharmaceuticals as well as the different labelling strategies, and summarises approaches to optimise metabolic stability. Additionally, it presents different target structures and addresses corresponding tracers, which are already used in clinical routine or are being investigated in clinical trials.
Collapse
Affiliation(s)
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria;
| |
Collapse
|