1
|
Alati S, Singh R, Pomper MG, Rowe SP, Banerjee SR. Preclinical Development in Radiopharmaceutical Therapy for Prostate Cancer. Semin Nucl Med 2023; 53:663-686. [PMID: 37468417 DOI: 10.1053/j.semnuclmed.2023.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Prostate cancer is a leading cause of cancer death in men worldwide. Among the various treatment options, radiopharmaceutical therapy has shown notable success in metastatic, castration-resistant disease. Radiopharmaceutical therapy is a systemic approach that delivers cytotoxic radiation doses precisely to the malignant tumors and/or tumor microenvironment. Therapeutic radiopharmaceuticals are composed of a therapeutic radionuclide and a high-affinity, tumor-targeting carrier molecule. Therapeutic radionuclides used in preclinical prostate cancer studies are primarily α-, β--, or Auger-electron-emitting radiometals or radiohalogens. Monoclonal antibodies, antibody-derived fragments, peptides, and small molecules are frequently used as tumor-targeting molecules. Over the years, several important membrane-associated proteases and receptors have been identified, validated, and subsequently used for preclinical radiotherapeutic development for prostate cancer. Prostate-specific membrane antigen (PSMA) is the most well-studied prostate cancer-associated protease in preclinical literature. PSMA-targeting radiotherapeutic agents are being investigated using high-affinity antibody- and small-molecule-based agents for safety and efficacy. Early generations of such agents were developed simply by replacing radionuclides of the imaging agents with therapeutic ones. Later, extensive structure-activity relationship studies were conducted to address the safety and efficacy issues obtained from initial patient data. Recent regulatory approval of the 177Lu-labeled low-molecular-weight agent, 177Lu-PSMA-617, is a significant accomplishment. Current preclinical experiments are focused on the structural modification of 177Lu-PSMA-617 and relevant investigational agents to increase tumor targeting and reduce off-target binding and toxicity in healthy organs. While lutetium-177 (177Lu) remains the most widely used radionuclide, radiolabeled analogs with iodine-131 (128I), yttrium-90 (89Y), copper-67 (67Cu), and terbium-161 (161Tb) have been evaluated as potential alternatives in recent years. In addition, agents carrying the α-particle-emitting radiohalogen, astatine-211 (211At), or radiometals, actinium-225 (225Ac), lead-212 (212Pb), radium-223 (223Ra), and thorium-227 (227Th), have been increasingly investigated in preclinical research. Besides PSMA-based radiotherapeutics, other prominent prostate cancer-related proteases, for example, human kallikrein peptidases (HK2 and HK3), have been explored using monoclonal-antibody-(mAb)-based targeting platforms. Several promising mAbs targeting receptors overexpressed on the different stages of prostate cancer have also been developed for radiopharmaceutical therapy, for example, Delta-like ligand 3 (DLL-3), CD46, and CUB domain-containing protein 1 (CDCP1). Progress is also being made using peptide-based targeting platforms for the gastrin-releasing peptide receptor (GRPR), a well-established membrane-associated receptor expressed in localized and metastatic prostate cancers. Furthermore, mechanism-driven combination therapies appear to be a burgeoning area in the context of preclinical prostate cancer radiotherapeutics. Here, we review the current developments related to the preclinical radiopharmaceutical therapy of prostate cancer. These are summarized in two major topics: (1) therapeutic radionuclides and (2) tumor-targeting approaches using monoclonal antibodies, small molecules, and peptides.
Collapse
Affiliation(s)
- Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
2
|
|
3
|
Antibody-Drug Conjugates in Uro-Oncology. Target Oncol 2022; 17:203-221. [PMID: 35567672 DOI: 10.1007/s11523-022-00872-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Abstract
Currently available treatment options for patients with refractory metastatic prostate, bladder, or kidney cancers are limited with the prognosis remaining poor. Advances in the pathobiology of tumors has led to the discovery of cancer antigens that may be used as the target for cancer treatment. Antibody-drug conjugates (ADCs) are a relatively new concept in cancer treatment that broaden therapeutic landscape. ADCs are examples of a 'drug delivery into the tumor' system composed of an antigen-directed antibody linked to a cytotoxic drug that may release cytotoxic components after binding to the antigen located on the surface of tumor cells. The clinical properties of drugs are influenced by every component of ADCs. Regarding uro-oncology, enfortumab vedotin (EV) and sacituzumab govitecan (SG) are currently registered for patients with locally advanced or metastatic urothelial cancer following previous treatment with an immune checkpoint inhibitor (iCPI; programmed death receptor-1 [PD-1] or programmed death-ligand 1 [PD-L1]) inhibitor) and platinum-containing chemotherapy. The EV-301 trial showed that EV significantly prolonged the overall survival compared with classic chemotherapy. The TROPHY-U-01 trial conducted to evaluate SG demonstrated promising results as regards the objective response rate and duration of response. The safety and efficacy of ADCs in monotherapy and polytherapy (mainly with iCPIs) for different cancer stages and tumor types are assessed in numerous ongoing clinical trials. The aim of this review is to present new molecular biomarkers, specific mechanisms of action, and ongoing clinical trials of ADCs in genitourinary cancers. In the expert discussion, we assess the place of ADCs in uro-oncology and discuss their clinical value.
Collapse
|
4
|
Zhu X, Ma X, Zhao S, Cao Z. DLX6-AS1 accelerates cell proliferation through regulating miR-497-5p/SNCG pathway in prostate cancer. ENVIRONMENTAL TOXICOLOGY 2021; 36:308-319. [PMID: 33035382 DOI: 10.1002/tox.23036] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 08/10/2020] [Accepted: 09/22/2020] [Indexed: 05/02/2023]
Abstract
Prostate cancer (PCa) has become the second leading cause of cancer-related mortality in males worldwide. Although the long noncoding RNA DLX6-AS1 has been recognized to be an oncogene in multiple cancers, the biological function and regulatory mechanism of DLX6-AS1 in prostate cancer are still obscure. In the present study, we observed that DLX6-AS1 was significantly upregulated in PCa tissues and cells. Knockdown of DLX6-AS1 inhibited PCa progression by suppressing cell proliferation and accelerating cell apoptosis. Molecular mechanism exploration indicated that DLX6-AS1 acted as a sponge for miR-497-5p and synuclein gamma (SNCG) was a downstream target gene of miR-497-5p. In addition, there was a negative correlation between DLX6-AS1 and miR-497-5p in PCa tissues. Rescue assays showed that SNCG overexpression could partially recover DLX6-AS1 knockdown-mediated inhibition of progression in PCa. Furthermore, xenograft tumor model was established to determine the role of DLX6-AS1 in PCa tumor growth and the results suggested that DLX6-AS1 could facilitate tumor growth by regulating SNCG in vivo. In conclusion, our study investigated the biological function and underlying mechanism of DLX6-AS1 in PCa and validated that DLX6-AS1 functioned as an oncogene through miR-497-5p/SNCG axis.
Collapse
Affiliation(s)
- Xu Zhu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xingxin Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shuli Zhao
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhigang Cao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Choi B, Choi H, Yu B, Kim DH. Synergistic Local Combination of Radiation and Anti-Programmed Death Ligand 1 Immunotherapy Using Radiation-Responsive Splintery Metallic Nanocarriers. ACS NANO 2020; 14:13115-13126. [PMID: 32885958 DOI: 10.1021/acsnano.0c04701] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Radiation therapy (RT)-mediated tumor immunogenicity offers an opportunity for synergistic combination RT and immunotherapy. One of the challenges in the clinic is to attain the optimum efficacy of combination RT and immunotherapy with minimized overlapping toxicities. Here, to achieve synergistic therapeutic efficacy of combinational RT and anti-programmed death ligand 1 (aPD-L1) immunotherapy, RT-responsive splintery snowflake-like Au nanocarriers (S-AuNC) were synthesized with a method for hierarchical bimetallic supra-nanostructures. Primary Au nanocrystals interconnected with Ag nanocrystals in S-AuNC showed RT-responsive structural deformation resulting in RT-triggered release of cargo aPD-LI in S-AuNC. The local combination of RT and aPD-L1 loaded S-AuNC significantly enhanced the immunogenic cell death for tumor microenvironment conversion. RT-triggered local aPD-L1 release allowed a controlled spatiotemporal combination RT and aPD-L1 immunotherapy resulting in a synergistic anticancer adaptive immune response with minimized systemic immune-related adverse effects.
Collapse
Affiliation(s)
- Bongseo Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Hyunjun Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Bo Yu
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
- Department of Biomedical Engineering, McCormick School of Engineering, Evanston, Illinois 60208, United States
- Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois 60611, United States
| |
Collapse
|
6
|
Orozco JJ, Kenoyer AL, Lin Y, O'Steen S, Guel R, Nartea ME, Hernandez AH, Hylarides MD, Fisher DR, Balkin ER, Hamlin DK, Wilbur DS, Orcutt KD, Wittrup KD, Green DJ, Gopal AK, Till BG, Sandmaier B, Press OW, Pagel JM. Therapy of Myeloid Leukemia using Novel Bispecific Fusion Proteins Targeting CD45 and 90Y-DOTA. Mol Cancer Ther 2020; 19:2575-2584. [PMID: 33082277 DOI: 10.1158/1535-7163.mct-20-0306] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/13/2020] [Accepted: 09/28/2020] [Indexed: 11/16/2022]
Abstract
Pretargeted radioimmunotherapy (PRIT) has been investigated as a multi-step approach to decrease relapse and toxicity for high-risk acute myeloid leukemia (AML). Relevant factors including endogenous biotin and immunogenicity, however, have limited the use of PRIT with an anti-CD45 antibody streptavidin conjugate and radiolabeled DOTA-biotin. To overcome these limitations we designed anti-murine and anti-human CD45 bispecific antibody constructs using 30F11 and BC8 antibodies, respectively, combined with an anti-yttrium (Y)-DOTA single-chain variable fragment (C825) to capture a radiolabeled ligand. The bispecific construct targeting human CD45 (BC8-Fc-C825) had high uptake in leukemia HEL xenografts [7.8 ± 0.02% percent injected dose/gram of tissue (% ID/g)]. Therapy studies showed that 70% of mice with HEL human xenografts treated with BC8-Fc-C825 followed by 44.4 MBq (1,200 μCi) of 90Y-DOTA-biotin survived at least 170 days after therapy, while all nontreated controls required euthanasia because of tumor progression by day 32. High uptake at sites of leukemia (spleen and bone marrow) was also seen with 30F11-IgG1-C825 in a syngeneic disseminated SJL murine leukemia model (spleen, 9.0 ± 1.5% ID/g and bone marrow, 8.1 ± 1.2% ID/g), with minimal uptake in all other normal organs (<0.5% ID/g) at 24 hours after 90Y-DOTA injections. SJL leukemia mice treated with the bispecific 30F11-IgG1-C825 and 29.6 MBq (800 μCi) of 90Y-DOTA-biotin had a survival advantage compared with untreated leukemic mice (median, 43 vs. 30 days, respectively; P < 0.0001). These data suggest bispecific antibody-mediated PRIT may be highly effective for leukemia therapy and translation to human studies.
Collapse
Affiliation(s)
- Johnnie J Orozco
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Aimee L Kenoyer
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Yukang Lin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Shyril O'Steen
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rosario Guel
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Margaret E Nartea
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alexandra H Hernandez
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark D Hylarides
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Darrell R Fisher
- Versant Medical Physics and Radiation Dosimetry, Richland, Washington
| | - Ethan R Balkin
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Donald K Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | | | - K Dane Wittrup
- Massachusetts Institute of Technology, Boston, Massachusetts
| | - Damian J Green
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Ajay K Gopal
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Brian G Till
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Brenda Sandmaier
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Oliver W Press
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | | |
Collapse
|
7
|
Tsai WTK, Zettlitz KA, Dahlbom M, Reiter RE, Wu AM. Evaluation of [ 131I]I- and [ 177Lu]Lu-DTPA-A11 Minibody for Radioimmunotherapy in a Preclinical Model of PSCA-Expressing Prostate Cancer. Mol Imaging Biol 2020; 22:1380-1391. [PMID: 32661830 PMCID: PMC7688013 DOI: 10.1007/s11307-020-01518-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Radioimmunotherapy uses tumor-specific antibodies to deliver therapeutic radionuclides, but hematological toxicity due to the long serum half-life of intact antibodies remains a challenge. We evaluated a smaller antibody fragment, the minibody, with faster kinetics and a potentially improved therapeutic index. PROCEDURES The anti-prostate stem cell antigen (PSCA) minibody (A11 Mb) was radiolabeled with iodine-124 ([124I]I-A11 Mb) or conjugated with deferoxamine (DFO) and labeled with zirconium-89 ([89Zr]Zr-DFO-A11 Mb) for surrogate immunoPET to profile pharmacokinetics in a human prostate cancer xenograft model. Subsequently, minibodies labeled with two therapeutic beta emitters, directly iodinated [131I]I-A11 Mb (non-residualizing) and 177Lu chelated using DTPA ([177Lu]Lu-DTPA-A11 Mb) (residualizing), were compared for in vitro antigen-specific cytotoxicity. Full biodistribution studies (in 22Rv1-PSCA tumor bearing and hPSCA knock-in mice) were conducted for dosimetry calculations. Finally, the lead candidate [131I]I-A11 Mb was evaluated in a radioimmunotherapy experiment. Escalating single doses (3.7, 11, or 37 MBq) and saline control were administered to 22Rv1-PSCA tumor bearing mice and anti-tumor effects (tumor volume) and toxicity (body weight) were monitored. RESULTS Minibodies radiolabeled with therapeutic beta emitters [131I]I-A11 Mb and [177Lu]Lu-DTPA-A11 Mb exhibited comparable tumor cell growth inhibition in vitro. In vivo surrogate immunoPET imaging using [89Zr]Zr-DFO-A11 Mb showed activity retention in liver and kidney up to 72 h, while [124I]I-A11 Mb cleared from liver, kidney, and blood by 48 h. Based on full biodistribution and dosimetry calculations, administering 37 MBq [131I]I-A11 Mb was predicted to deliver a favorable dose to the tumor (35 Gy), with a therapeutic index of 22 (tumor:bone marrow). For [177Lu]Lu-DTPA-A11 Mb, the kidneys would be dose-limiting, and the maximum tolerated activity (7.4 MBq) was not predicted to deliver an effective radiation dose to tumor. Radioimmunotherapy with a single dose of [131I]I-A11 Mb showed dose-dependent tumor inhibition with minimal off-target toxicity and improved median survival (19 and 24 days, P < 0.001) compared with untreated mice (12 days). CONCLUSIONS These findings show the potential of the anti-PSCA minibody for targeted radioimmunotherapy with minimal toxicity, and the application of immunoPET and dosimetry for personalized treatment.
Collapse
Affiliation(s)
- Wen-Ting K Tsai
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, UC Los Angeles, Los Angeles, CA, USA
- Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Kirstin A Zettlitz
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, UC Los Angeles, Los Angeles, CA, USA
- Department of Molecular Imaging and Therapy, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Magnus Dahlbom
- Department of Molecular and Medical Pharmacology, Ahmanson Translational Imaging Division, David Geffen School of Medicine, UC Los Angeles, Los Angeles, CA, USA
| | - Robert E Reiter
- Department of Urology, David Geffen School of Medicine, UC Los Angeles, Los Angeles, CA, USA
| | - Anna M Wu
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, UC Los Angeles, Los Angeles, CA, USA.
- Department of Molecular Imaging and Therapy, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
8
|
Hu M, Yang J. Down-regulation of lncRNA UCA1 enhances radiosensitivity in prostate cancer by suppressing EIF4G1 expression via sponging miR-331-3p. Cancer Cell Int 2020; 20:449. [PMID: 32943997 PMCID: PMC7488500 DOI: 10.1186/s12935-020-01538-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND We aimed to explore the role of long noncoding RNA urothelial carcinoma-associated 1 (lncRNA UCA1) and its underlying mechanism in the radioresistance of prostate cancer (PCa). METHODS QRT-PCR was conducted to measure the expression of UCA1, microRNA-331-3p (miR-331-3p) and eukaryotic translation initiation factor 4 gamma 1 (EIF4G1) in PCa tissues and cells. The relative protein level was determined by western blot assay. Cell proliferation and apoptosis were detected by MTT, colony formation assay, and flow cytometry, respectively. The target interaction between miR-331-3p and UCA1 or EIF4G1 was predicted through bioinformatics analysis, and verified by dual-luciferase reporter gene assay system. RESULTS The high levels of UCA1 and EIF4G1 as well as the low level of miR-331-3p were observed in PCa tissues and cell lines. UCA1 and EIF4G1 expression were significantly upregulated by Gy radiation treatement. UCA1 or EIF4G1 knockdown repressed cell growth and enhanced cell apoptosis in 22RV1 and DU145 cells under radiation. Moreover, overexpression of EIF4G1 abolished UCA1 knockdown-induced effect on 6 Gy irradiated PCa cells. UCA1 sponged miR-331-3p to regulate EIF4G1 expression. CONCLUSIONS LncRNA UCA1 deletion suppressed the radioresistance to PCa by suppressing EIF4G1 expression via miR-331-3p. UCA1 acted as a potential regulator of radioresistance of PCa, providing a promising therapeutic target for PCa.
Collapse
Affiliation(s)
- Minhua Hu
- Department of Nursing College, Xi’an Medical University, Xi’an, 710021 Shaanxi Province China
| | - Jincheng Yang
- Department of Urology Surgery, The First People’s Hospital of Yinchuan, No. 4, Liqun West Street, Xingqing District, Yinchuan, 750004 Ningxia China
| |
Collapse
|
9
|
Kasten BB, Ferrone S, Zinn KR, Buchsbaum DJ. B7-H3-targeted Radioimmunotherapy of Human Cancer. Curr Med Chem 2020; 27:4016-4038. [PMID: 30836909 PMCID: PMC8668195 DOI: 10.2174/0929867326666190228120908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Targeted Radioimmunotherapy (RIT) is an attractive approach to selectively localize therapeutic radionuclides to malignant cells within primary and metastatic tumors while sparing normal tissues from the effects of radiation. Many human malignancies express B7-H3 on the tumor cell surface, while expression on the majority of normal tissues is limited, presenting B7-H3 as a candidate target for RIT. This review provides an overview of the general principles of targeted RIT and discusses publications that have used radiolabeled B7-H3-targeted antibodies for RIT of cancer in preclinical or clinical studies. METHODS Databases including PubMed, Scopus, and Google Scholar were searched for publications through June 2018 using a combination of terms including "B7-H3", "radioimmunotherapy", "targeted", "radiotherapy", and "cancer". After screening search results for relevancy, ten publications were included for discussion. RESULTS B7-H3-targeted RIT studies to date range from antibody development and assessment of novel Radioimmunoconjugates (RICs) in animal models of human cancer to phase II/III trials in humans. The majority of clinical studies have used B7-H3-targeted RICs for intra- compartment RIT of central nervous system malignancies. The results of these studies have indicated high tolerability and favorable efficacy outcomes, supporting further assessment of B7-H3-targeted RIT in larger trials. Preclinical B7-H3-targeted RIT studies have also shown encouraging therapeutic outcomes in a variety of solid malignancies. CONCLUSION B7-H3-targeted RIT studies over the last 15 years have demonstrated feasibility for clinical development and support future assessment in a broader array of human malignancies. Future directions worthy of exploration include strategies that combine B7-H3- targeted RIT with chemotherapy or immunotherapy.
Collapse
Affiliation(s)
- Benjamin B. Kasten
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Kurt R. Zinn
- Institute for Quantitative Health Science and Engineering, Department of Radiology, Michigan State University, East Lansing, Michigan, U.S.A
| | - Donald J. Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| |
Collapse
|
10
|
Wu C, Miao C, Tang Q, Zhou X, Xi P, Chang P, Hua L, Ni H. MiR-129-5p promotes docetaxel resistance in prostate cancer by down-regulating CAMK2N1 expression. J Cell Mol Med 2019; 24:2098-2108. [PMID: 31876385 PMCID: PMC7011149 DOI: 10.1111/jcmm.14050] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/11/2018] [Accepted: 11/05/2018] [Indexed: 12/22/2022] Open
Abstract
This study focuses on the effect of miR‐129‐5p on docetaxel‐resistant (DR) prostate cancer (PCa) cells invasion, migration and apoptosis. In our study, the expression of CAMK2N1 was assessed by qRT‐PCR in PCa patient tissues and cell lines including PC‐3 and PC‐3‐DR. Cells transfected with miR‐129‐5p mimics, inhibitor, CAMK2N1 or negative controls (NC) were used to interrogate their effects on DR cell invasions, migrations and apoptosis during docetaxel (DTX) treatments. The apoptosis rate of the PCa cells was validated by flow cytometry. Relationships between miR‐129‐5p and CAMK2N1 levels were identified by qRT‐PCR and dual‐luciferase reporter assay. CAMK2N1 was found to be down‐expressed in DR PCa tissue sample, and low levels of CAMK2N1 were correlated with high docetaxel resistance and clinical prediction of poor survival. CAMK2N1 levels were decreased in DR PCa cells treated with DXT. We further explored that up‐regulation of miR‐129‐5p could promote DR PCa cells viability, invasion and migration but demote apoptosis. Involved molecular mechanism studies revealed that miR‐129‐5p reduced downstream CAMK2N1 expression to further impact on chemoresistance to docetaxel of PCa cells, indicating its vital role in PCa docetaxel resistance. Our findings revealed that miR‐129‐5p contributed to the resistance of PC‐3‐DR cells to docetaxel through suppressing CAMK2N1 expression, and thus targeting miR‐129‐5p may provide a novel therapeutic approach in sensitizing PCa to future docetaxel treatment.
Collapse
Affiliation(s)
- Cheng Wu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunqing Miao
- Department of Urology, People's Hospital of Dongtai City, Dongtai, Jiangsu, China
| | - Qingsheng Tang
- Department of Urology, People's Hospital of Dongtai City, Dongtai, Jiangsu, China
| | - Xunrong Zhou
- Department of Urology, People's Hospital of Dongtai City, Dongtai, Jiangsu, China
| | - Pengshan Xi
- Department of Urology, People's Hospital of Dongtai City, Dongtai, Jiangsu, China
| | - Ping'an Chang
- Department of Urology, People's Hospital of Dongtai City, Dongtai, Jiangsu, China
| | - Lixin Hua
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haodong Ni
- Department of Urology, People's Hospital of Dongtai City, Dongtai, Jiangsu, China
| |
Collapse
|
11
|
Yang J, Hao T, Sun J, Wei P, Zhang H. Long noncoding RNA GAS5 modulates α-Solanine-induced radiosensitivity by negatively regulating miR-18a in human prostate cancer cells. Biomed Pharmacother 2019; 112:108656. [PMID: 30970507 DOI: 10.1016/j.biopha.2019.108656] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 01/23/2019] [Accepted: 02/01/2019] [Indexed: 12/25/2022] Open
Abstract
Radiotherapy is an adjuvant treatment of surgery in prostate cancer, while radioresistance has been the challenge of treatment. It has been reported that α-Solanine exhibits anti-cancer activity and enhances the chemo- and radio-sensitivity in several human cancers, whereas the role of α-Solanine on radiosensitivity to PCa remains to be uncovered yet. We found α-Solanine decreased cell viability in human PCa cells rather than normal prostate epithelial cells in vitro. Functional experiments showed that cell viability and colonies formation were declined & apoptosis rate and DNA double strand breaks (DSBs) marker γ-H2AX expressions were elevated by α-Solanine in PCa cells treated with X-ray irradiation, compared with X-ray irradiation treatment only. GAS5 was down-regulated & miR-18a was up-regulated in PCa cells, which was reversed in the presence of α-Solanine. Effects of ectopic GAS5 on inhibiting cell viability and survival & promoting apoptosis and DNA damage were reversed by miR-18a overexpression in PCa cells. Moreover, GAS5 regulated miR-18a expression by target binding during α-Solanine treatment. Collectively, α-Solanine suppresses cell proliferation and promotes radiosensitivity through up-regulating GAS5/miR-18a pathway in PCa. Our results provide a novel mechanism of α-Solanine treatment in human prostate cancer and help to develop a new approach to sensitizing radioresistant prostate cancer cells by targeting GAS5/miR-18a.
Collapse
Affiliation(s)
- Jinhui Yang
- Department of Urology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Tongtong Hao
- Department of Urology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Jiantao Sun
- Department of Urology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China.
| | - Pengtao Wei
- Department of Urology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Han Zhang
- Department of Urology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| |
Collapse
|
12
|
Zhang C, Yan L, Song H, Ma Z, Chen D, Yang F, Fang L, Li Z, Li K, Li D, Yu N, Liu H, Xu Z. Elevated Serum Sialic Acid Levels Predict Prostate Cancer As Well As Bone Metastases. J Cancer 2019; 10:449-457. [PMID: 30719139 PMCID: PMC6360313 DOI: 10.7150/jca.27700] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/02/2018] [Indexed: 01/08/2023] Open
Abstract
Objective: To evaluate the value of serum sialic acid (SA) in diagnosis of benign prostatic hyperplasia (BPH), prostate cancer (PCa), and bone metastases in PCa patients. Materials and Methods: Data from 540 patients who were newly diagnosed with PCa or BPH between November 2014 and March 2018 were retrospectively collected and analyzed. Pretreatment SA levels were compared across various groups, then, associations between SA levels and clinic parameters of patients were analyzed as well. Univariate and multivariate logistic regression analyses were further used to identify independent associations. Results: The mean SA levels in patients with PCa were significantly higher than with BPH (p = 0.013). Furthermore, PCa patients with bone metastases showed elevated SA levels compared with PCa without bone metastases (p < 0.001). A multivariate logistic regression model showed that: SA level > 52.35 mg/dL was identified to be independently associated with the diagnosis of PCa (HR = 1.645, p = 0.036), and SA level > 59 mg/dL was identified to be independent association with the presence of bone metastases in PCa patients (HR = 6.421, p = 0.012). Conclusions: Elevated SA level is an independent predictor of prostate cancer as well as its bone metastases. Therefore, SA level may be a promising diagnostic and prognostic biomarker for prostate cancer and bone metastases.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Lei Yan
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Hongkai Song
- Department of Bone and Soft Tissue Tumor, Shandong Cancer Hospital affiliated to Shandong University, Wenhuaxi Road 440#, Jinan, 250117, P.R.China
| | - Zheng Ma
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China.,Department of Urology, The People's Hospital of Liaocheng, Dongchangxi Road 67#, Liaocheng, 252000, P.R.China
| | - Dongshan Chen
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Feilong Yang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Liang Fang
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Zeyan Li
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Kui Li
- Department of Urology, The people's Hospital of Yucheng, Kaituo Road 753#, Dezhou, 251200, P.R.China
| | - Dawei Li
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Nengwang Yu
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Hainan Liu
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| | - Zhonghua Xu
- Department of Urology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, P.R.China
| |
Collapse
|
13
|
Bartholomä MD. Radioimmunotherapy of solid tumors: Approaches on the verge of clinical application. J Labelled Comp Radiopharm 2018. [PMID: 29524233 DOI: 10.1002/jlcr.3619] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
While radioimmunotherapy (RIT) for the treatment of hematological malignancies such as indolent B-cell lymphoma has proven quite successful, clinical results of RIT in solid tumors have only been moderate in the past. The reasons were manifold and can be mostly attributed to the different biological properties of solid tumors vs hematological cancers. Furthermore, the slow clearance of the radiolabelled antibody prevents the use of radiation doses necessary to achieve clinical responses. The long biological half-life of radioimmunoconjugates results in high background levels and is the main reason for radiation related toxicities. In recent years, researchers and clinicians have developed solutions for the successful application of RIT for the treatment of solid tumors. These include compartmental route of administration, neoadjuvant therapies, and pretargeting approaches. In this review, recent developments in RIT for the treatment of solid tumors that address these restrictions as well as future perspectives will be highlighted from a clinical perspective.
Collapse
Affiliation(s)
- Mark D Bartholomä
- Department of Nuclear Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Yan H, Kapoor V, Nguyen K, Akers WJ, Li H, Scott J, Laforest R, Rogers B, Thotala D, Hallahan D. Anti-tax interacting protein-1 (TIP-1) monoclonal antibody targets human cancers. Oncotarget 2017; 7:43352-43362. [PMID: 27270318 PMCID: PMC5190028 DOI: 10.18632/oncotarget.9713] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/13/2016] [Indexed: 02/04/2023] Open
Abstract
Radiation-inducible neo-antigens are proteins expressed on cancer cell surface after exposure to ionizing radiation (IR). These neo-antigens provide opportunities to specifically target cancers while sparing normal tissues. Tax interacting protein-1 (TIP-1) is induced by irradiation and is translocated to the surface of cancer cells. We have developed a monoclonal antibody, 2C6F3, against TIP-1. Epitope mapping revealed that 2C6F3 binds to the QPVTAVVQRV epitope of the TIP-1 protein. 2C6F3 binds to the surface of lung cancer (A549, LLC) and glioma (D54, GL261) cell lines. 2C6F3 binds specifically to TIP-1 and ELISA analysis showed that unconjugated 2C6F3 efficiently blocked binding of radiolabeled 2C6F3 to purified TIP-1 protein. To study in vivo tumor binding, we injected near infrared (NIR) fluorochrome-conjugated 2C6F3 via tail vein in mice bearing subcutaneous LLC and GL261 heterotopic tumors. The NIR images indicated that 2C6F3 bound specifically to irradiated LLC and GL261 tumors, with little or no binding in un-irradiated tumors. We also determined the specificity of 2C6F3 to bind tumors in vivo using SPECT/CT imaging. 2C6F3 was conjugated with diethylene triamine penta acetic acid (DTPA) chelator and radiolabeled with 111Indium (111In). SPECT/CT imaging revealed that 111In-2C6F3 bound more to the irradiated LLC tumors compared to un-irradiated tumors. Furthermore, injection of DTPA-2C6F3 labeled with the therapeutic radioisotope, 90Y, (90Y-DTPA-2C6F3) significantly delayed LLC tumor growth. 2C6F3 mediated antibody dependent cell-mediated cytotoxicity (ADCC) and antibody dependent cell-mediated phagocytosis (ADCP) in vitro. In conclusion, the monoclonal antibody 2C6F3 binds specifically to TIP-1 on cancer and radio-immunoconjugated 2C6F3 improves tumor control.
Collapse
Affiliation(s)
- Heping Yan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Vaishali Kapoor
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kim Nguyen
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Walter J Akers
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Hua Li
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jalen Scott
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Richard Laforest
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Buck Rogers
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Dennis Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA.,Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
15
|
Radioimmunotherapy in Oncology. CURRENT RADIOLOGY REPORTS 2017. [DOI: 10.1007/s40134-017-0258-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
16
|
Elgqvist J. Nanoparticles as Theranostic Vehicles in Experimental and Clinical Applications-Focus on Prostate and Breast Cancer. Int J Mol Sci 2017; 18:E1102. [PMID: 28531102 PMCID: PMC5455010 DOI: 10.3390/ijms18051102] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/13/2017] [Accepted: 05/15/2017] [Indexed: 12/27/2022] Open
Abstract
Prostate and breast cancer are the second most and most commonly diagnosed cancer in men and women worldwide, respectively. The American Cancer Society estimates that during 2016 in the USA around 430,000 individuals were diagnosed with one of these two types of cancers, and approximately 15% of them will die from the disease. In Europe, the rate of incidences and deaths are similar to those in the USA. Several different more or less successful diagnostic and therapeutic approaches have been developed and evaluated in order to tackle this issue and thereby decrease the death rates. By using nanoparticles as vehicles carrying both diagnostic and therapeutic molecular entities, individualized targeted theranostic nanomedicine has emerged as a promising option to increase the sensitivity and the specificity during diagnosis, as well as the likelihood of survival or prolonged survival after therapy. This article presents and discusses important and promising different kinds of nanoparticles, as well as imaging and therapy options, suitable for theranostic applications. The presentation of different nanoparticles and theranostic applications is quite general, but there is a special focus on prostate cancer. Some references and aspects regarding breast cancer are however also presented and discussed. Finally, the prostate cancer case is presented in more detail regarding diagnosis, staging, recurrence, metastases, and treatment options available today, followed by possible ways to move forward applying theranostics for both prostate and breast cancer based on promising experiments performed until today.
Collapse
Affiliation(s)
- Jörgen Elgqvist
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden.
- Department of Physics, University of Gothenburg, 412 96 Gothenburg, Sweden.
| |
Collapse
|
17
|
Lutetium-177-labelled anti-prostate-specific membrane antigen antibody and ligands for the treatment of metastatic castrate-resistant prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis 2017; 20:352-360. [DOI: 10.1038/pcan.2017.23] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/04/2017] [Accepted: 03/16/2017] [Indexed: 11/08/2022]
|
18
|
Ceder J, Elgqvist J. Targeting Prostate Cancer Stem Cells with Alpha-Particle Therapy. Front Oncol 2017; 6:273. [PMID: 28119854 PMCID: PMC5220245 DOI: 10.3389/fonc.2016.00273] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/22/2016] [Indexed: 01/16/2023] Open
Abstract
Modern molecular and radiopharmaceutical development has brought the promise of tumor-selective delivery of antibody-drug conjugates to tumor cells for the diagnosis and treatment of primary and disseminated tumor disease. The classical mode of discourse regarding targeted therapy has been that the antigen targeted must be highly and homogenously expressed in the tumor cell population, and at the same time exhibit low expression in healthy tissue. However, there is increasing evidence that the reason cancer patients are not cured by current protocols is that there exist subpopulations of cancer cells that are resistant to conventional therapy including radioresistance and that these cells express other target antigens than the bulk of the tumor cells. These types of cells are often referred to as cancer stem cells (CSCs). The CSCs are tumorigenic and have the ability to give rise to all types of cells found in a cancerous disease through the processes of self-renewal and differentiation. If the CSCs are not eradicated, the cancer is likely to recur after therapy. Due to some of the characteristics of alpha particles, such as short path length and high density of energy depositions per distance traveled in tissue, they are especially well suited for use in targeted therapies against microscopic cancerous disease. The characteristics of alpha particles further make it possible to minimize the irradiation of non-targeted surrounding healthy tissue, but most importantly, make it possible to deliver high-absorbed doses locally and therefore eradicating small tumor cell clusters on the submillimeter level, or even single tumor cells. When alpha particles pass through a cell, they cause severe damage to the cell membrane, cytoplasm, and nucleus, including double-strand breaks of DNA that are very difficult to repair for the cell. This means that very few hits to a cell by alpha particles are needed in order to cause cell death, enabling killing of cells, such as CSCs, exhibiting cellular resistance mechanisms to conventional therapy. This paper presents and evaluates the possibility of using alpha-particle emitting radionuclides in the treatment of prostate cancer (PCa) and discusses the parameters that have to be considered as well as pros and cons of targeted alpha-particle therapy in the treatment of PCa. By targeting and eradicating the CSCs responsible of tumor recurrence in patients who no longer respond to conventional therapies, including androgen deprivation and castration, it may be possible to cure the disease, or prolong survival significantly.
Collapse
Affiliation(s)
- Jens Ceder
- Department of Translational Medicine, Lund University, Skåne University Hospital , Malmö , Sweden
| | - Jörgen Elgqvist
- Department of Physics, University of Gothenburg, Gothenburg, Sweden; Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
19
|
Timmermand OV, Nilsson J, Strand SE, Elgqvist J. High resolution digital autoradiographic and dosimetric analysis of heterogeneous radioactivity distribution in xenografted prostate tumors. Med Phys 2016; 43:6632. [PMID: 27908170 DOI: 10.1118/1.4967877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The first main aim of this study was to illustrate the absorbed dose rate distribution from 177Lu in sections of xenografted prostate cancer (PCa) tumors using high resolution digital autoradiography (DAR) and compare it with hypothetical identical radioactivity distributions of 90Y or 7 MeV alpha-particles. Three dosimetry models based on either dose point kernels or Monte Carlo simulations were used and evaluated. The second and overlapping aim, was to perform DAR imaging and dosimetric analysis of the distribution of radioactivity, and hence the absorbed dose rate, in tumor sections at an early time point after injection during radioimmunotherapy using 177Lu-h11B6, directed against the human kallikrein 2 antigen. METHODS Male immunodeficient BALB/c nude mice, aged 6-8 w, were inoculated by subcutaneous injection of ∼107 LNCaP cells in a 200 μl suspension of a 1:1 mixture of medium and Matrigel. The antibody h11B6 was conjugated with the chelator CHX-A″-DTPA after which conjugated h11B6 was mixed with 177LuCl3. The incubation was performed at room temperature for 2 h, after which the labeling was terminated and the solution was purified on a NAP-5 column. About 20 MBq 177Lu-h11B6 was injected intravenously in the tail vein. At approximately 10 h postinjection (hpi), the mice were sacrificed and one tumor was collected from each of the five animals and cryosectioned into 10 μm thick slices. The tumor slices were measured and imaged using the DAR MicroImager system and the M3Vision software. Then the absorbed dose rate was calculated using a dose point kernel generated with the Monte Carlo code gate v7.0. RESULTS The DAR system produced high resolution images of the radioactivity distribution, close to the resolution of single PCa cells. The DAR images revealed a pronounced heterogeneous radioactivity distribution, i.e., count rate per area, in the tumors, indicated by the normalized intensity variations along cross sections as mean ± SD: 0.15 ± 0.15, 0.20 ± 0.18, 0.12 ± 0.17, 0.15 ± 0.16, and 0.23 ± 0.22, for each tumor section, respectively. The absorbed dose rate distribution for 177Lu at the time of dissection 10 hpi showed a maximum value of 2.9 ± 0.4 Gy/h (mean ± SD), compared to 6.0 ± 0.9 and 159 ± 25 Gy/h for the hypothetical 90Y and 7 MeV alpha-particle cases assuming the same count rate densities. Mean absorbed dose rate values were 0.13, 0.53, and 6.43 Gy/h for 177Lu, 90Y, and alpha-particles, respectively. CONCLUSIONS The initial uptake of 177Lu-h11B6 produces a high absorbed dose rate, which is important for a successful therapeutic outcome. The hypothetical 90Y case indicates a less heterogeneous absorbed dose rate distribution and a higher mean absorbed dose rate compared to 177Lu, although with a potentially increased irradiation of surrounding healthy tissue. The hypothetical alpha-particle case indicates the possibility of a higher maximum absorbed dose rate, although with a more heterogeneous absorbed dose rate distribution.
Collapse
Affiliation(s)
- Oskar V Timmermand
- Faculty of Medicine, Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Lund 22185, Sweden
| | - Jenny Nilsson
- Sahlgrenska Academy, Institute of Clinical Sciences, Department Radiation Physics, University of Gothenburg, Gothenburg 41345, Sweden
| | - Sven-Erik Strand
- Faculty of Medicine, Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Lund 22185, Sweden and Faculty of Medicine, Department of Clinical Sciences Lund, Medical Radiation Physics, Lund University, Lund 22185, Sweden
| | - Jörgen Elgqvist
- Faculty of Science, Department of Physics, University of Gothenburg, Gothenburg 41296, Sweden
| |
Collapse
|
20
|
Abstract
Prostate cancer (PCa) is the most common malignancy in men and a major cause of cancer death. Accurate imaging plays an important role in diagnosis, staging, restaging, detection of biochemical recurrence, and for therapy of patients with PCa. Because no effective treatment is available for advanced PCa, there is an urgent need to develop new and more effective therapeutic strategies. To optimize treatment outcome, especially in high-risk patients with PCa, therapy for PCa is moving rapidly toward personalization. Medical imaging, including positron emission tomography (PET)/computed tomography (CT), plays an important role in personalized medicine in oncology. In the recent years, much focus has been on prostate-specific membrane antigen (PSMA) as a promising target for imaging and therapy with radionuclides, as it is upregulated in most PCa. In the prostate, one potential role for PSMA PET imaging is to help guide focal therapy. Several studies have shown great potential of PSMA PET/CT for initial staging, lymph node staging, and detection of recurrence of PCa, even at very low prostate-specific antigen values after primary therapy. Furthermore, studies have shown that PSMA PET/CT has a higher detection rate than choline PET/CT. Radiolabeled PSMA ligands for therapy show promise in several studies with metastatic PCa and is an area of active investigation. The "image and treat" strategy, with radiolabeled PSMA ligands, has the potential to improve the treatment outcome of patients with PCa and is paving the way for precision medicine in PCa. The aim of this review is to give an overview of recent advancement in PSMA PET and radionuclide therapy for PCa.
Collapse
Affiliation(s)
- Kirsten Bouchelouche
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark.
| | - Baris Turkbey
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD
| |
Collapse
|