1
|
Sabanathan D, Lund ME, Campbell DH, Walsh BJ, Gurney H. Radioimmunotherapy for solid tumors: spotlight on Glypican-1 as a radioimmunotherapy target. Ther Adv Med Oncol 2021; 13:17588359211022918. [PMID: 34646364 PMCID: PMC8504276 DOI: 10.1177/17588359211022918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Radioimmunotherapy (i.e., the use of radiolabeled tumor targeting antibodies) is an emerging approach for the diagnosis, therapy, and monitoring of solid tumors. Often using paired agents, each targeting the same tumor molecule, but labelled with an imaging or therapeutic isotope, radioimmunotherapy has achieved promising clinical results in relatively radio-resistant solid tumors such as prostate. Several approaches to optimize therapeutic efficacy, such as dose fractionation and personalized dosimetry, have seen clinical success. The clinical use and optimization of a radioimmunotherapy approach is, in part, influenced by the targeted tumor antigen, several of which have been proposed for different solid tumors. Glypican-1 (GPC-1) is a heparan sulfate proteoglycan that is expressed in a variety of solid tumors, but whose expression is restricted in normal adult tissue. Here, we discuss the preclinical and clinical evidence for the potential of GPC-1 as a radioimmunotherapy target. We describe the current treatment paradigm for several solid tumors expressing GPC-1 and suggest the potential clinical utility of a GPC-1 directed radioimmunotherapy for these tumors.
Collapse
Affiliation(s)
- Dhanusha Sabanathan
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | | | | | | | - Howard Gurney
- Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, Sydney, NSW 2109, Australia
| |
Collapse
|
2
|
Sabanathan D, Campbell DH, Velonas VM, Wissmueller S, Mazure H, Trifunovic M, Poursoltan P, Ho Shon K, Mackay TR, Lund ME, Lu Y, Roach PJ, Bailey DL, Walsh BJ, Gillatt D, Gurney H. Safety and tolerability of Miltuximab ® - a first in human study in patients with advanced solid cancers. ASIA OCEANIA JOURNAL OF NUCLEAR MEDICINE & BIOLOGY 2021; 9:86-100. [PMID: 34250138 PMCID: PMC8255523 DOI: 10.22038/aojnmb.2021.55600.1386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 01/31/2023]
Abstract
OBJECTIVES Miltuximab® is a chimeric antibody targeting Glypican-1 (GPC-1), a cell surface antigen which is overexpressed in solid cancers. Miltuximab® has shown promising safety and efficacy in radioimmunotherapy models of prostate cancer. This first in human study used Miltuximab® radiolabelled with Gallium-67 ([67Ga]Ga-DOTA-Miltuximab®). The primary study endpoint was to establish safety and tolerability of Miltuximab®. Secondary endpoints were biodistribution, tumour targeting and pharmacokinetic analysis. METHODS Four cohorts of three patients (9 with advanced prostate cancer, 2 with pancreatic and 1 with bladder cancer) were dosed with 1 mg, ~250 MBq of [67Ga]Ga-DOTA-Miltuximab®. Cohort 1 received [67Ga]Ga-DOTA-Miltuximab® alone, while cohorts 2-4 were pre-infused with increasing doses (3.5, 11.5 and 24 mg, respectively) of unlabelled Miltuximab®-DOTA 1 hour prior to [67Ga]Ga-DOTA-Miltuximab®. Safety and tolerability were assessed by clinical and standard laboratory assessments. Patients underwent whole body gamma-camera scans and SPECT/CT scans up to 144 h post-infusion. Total organ radiation exposure was determined by dosimetry of whole-body gamma scans. RESULTS The dosing regimen was well tolerated, with no drug-related adverse events observed. Liver and spleen uptake of [67Ga]Ga-DOTA-Miltuximab® was observed. Liver uptake was reduced by pre-infusion of unlabelled Miltuximab®-DOTA. Dosimetry analysis showed a favorable exposure profile. [67Ga]Ga-DOTA-Miltuximab® targeting to tumour sites was observed in two prostate cancer patients who had failed enzalutamide treatment. Higher doses of unlabelled antibody achieved lower liver uptake and increased antibody serum half life. CONCLUSIONS This study is the first in human for Miltuximab® a first in class antibody targeting GPC-1. The trial met its primary endpoint of safety, demonstrating its potential as a safe and tolerable monoclonal antibody. This safety data, together with targeting to tumour lesions and biodistribution information supports the further clinical development of Miltuximab® as a theranostic agent in a planned Phase I human trial.
Collapse
Affiliation(s)
- Dhanusha Sabanathan
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | | | - Vicki M. Velonas
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | | | - Hubert Mazure
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | | | - Pirooz Poursoltan
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Kevin Ho Shon
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | | | - Maria E. Lund
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | - Yanling Lu
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | | | | | - Bradley J. Walsh
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | - David Gillatt
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Howard Gurney
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
3
|
Yeh MC, Tse BWC, Fletcher NL, Houston ZH, Lund M, Volpert M, Stewart C, Sokolowski KA, Jeet V, Thurecht KJ, Campbell DH, Walsh BJ, Nelson CC, Russell PJ. Targeted beta therapy of prostate cancer with 177Lu-labelled Miltuximab® antibody against glypican-1 (GPC-1). EJNMMI Res 2020; 10:46. [PMID: 32382920 PMCID: PMC7206480 DOI: 10.1186/s13550-020-00637-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Chimeric antibody Miltuximab®, a human IgG1 engineered from the parent antibody MIL-38, is in clinical development for solid tumour therapy. Miltuximab® targets glypican-1 (GPC-1), a cell surface protein involved in tumour growth, which is overexpressed in solid tumours, including prostate cancer (PCa). This study investigated the potential of 89Zr-labelled Miltuximab® as an imaging agent, and 177Lu-labelled Miltuximab® as a targeted beta therapy, in a mouse xenograft model of human prostate cancer. METHODS Male BALB/c nude mice were inoculated subcutaneously with GPC-1-positive DU-145 PCa cells. In imaging and biodistribution studies, mice bearing palpable tumours received (a) 2.62 MBq [89Zr]Zr-DFO-Miltuximab® followed by PET-CT imaging, or (b) 6 MBq [177Lu]Lu-DOTA-Miltuximab® by Cerenkov imaging, and ex vivo assessment of biodistribution. In an initial tumour efficacy study, mice bearing DU-145 tumours were administered intravenously with 6 MBq [177Lu]Lu-DOTA-Miltuximab® or control DOTA-Miltuximab® then euthanised after 27 days. In a subsequent survival efficacy study, tumour-bearing mice were given 3 or 10 MBq of [177Lu]Lu-DOTA-Miltuximab®, or control, and followed up to 120 days. RESULTS Antibody accumulation in DU-145 xenografts was detected by PET-CT imaging using [89Zr]Zr-DFO-Miltuximab® and confirmed by Cerenkov luminescence imaging post injection of [177Lu]Lu-DOTA-Miltuximab®. Antibody accumulation was higher (% IA/g) in tumours than other organs across multiple time points. A single injection with 6 MBq of [177Lu]Lu-DOTA-Miltuximab® significantly inhibited tumour growth as compared with DOTA-Miltuximab® (control). In the survival study, mice treated with 10 MBq [177Lu]Lu-DOTA-Miltuximab® had significantly prolonged survival (mean 85 days) versus control (45 days), an effect associated with increased cancer cell apoptosis. Tissue histopathology assessment showed no abnormalities associated with [177Lu]Lu-DOTA-Miltuximab®, in line with other observations of tolerability, including body weight stability. CONCLUSION These findings demonstrate the potential utility of Miltuximab® as a PET imaging agent ([89Zr]Zr-DFO-Miltuximab®) and a beta therapy ([177Lu]Lu-DOTA-Miltuximab®) in patients with PCa or other GPC-1 expressing tumours.
Collapse
Affiliation(s)
- Mei-Chun Yeh
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Brian W C Tse
- Preclinical Imaging Facility, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre in Biomedical Imaging Technology, University of Queensland, Building 57 University Drive, St Lucia, Queensland, 4072, Australia
| | - Zachary H Houston
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre in Biomedical Imaging Technology, University of Queensland, Building 57 University Drive, St Lucia, Queensland, 4072, Australia
| | - Maria Lund
- Glytherix Ltd, Suite 2, Ground Floor 75 Talavera Road, Macquarie Park, New South Wales, 2113, Australia
| | - Marianna Volpert
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Chelsea Stewart
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Varinder Jeet
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre in Biomedical Imaging Technology, University of Queensland, Building 57 University Drive, St Lucia, Queensland, 4072, Australia
| | - Douglas H Campbell
- Glytherix Ltd, Suite 2, Ground Floor 75 Talavera Road, Macquarie Park, New South Wales, 2113, Australia
| | - Bradley J Walsh
- Glytherix Ltd, Suite 2, Ground Floor 75 Talavera Road, Macquarie Park, New South Wales, 2113, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Pamela J Russell
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia.
| |
Collapse
|
4
|
Jadvar H. Targeted α-Therapy in Cancer Management: Synopsis of Preclinical and Clinical Studies. Cancer Biother Radiopharm 2020; 35:475-484. [PMID: 32202923 DOI: 10.1089/cbr.2019.3340] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The approval of 223Ra dichloride (223RaCl2) in 2013 was a principal event in introducing targeted α-therapy as a form of safe and effective management strategy in cancer. There is an increasing interest in research and development of new targeted α-therapy agents spearheaded by advancements in cancer biology, radiochemistry, and availability of clinically relevant α particles. There are active clinical studies on sequencing or combining 223RaCl2 with other drug regimens in the setting of metastatic prostate cancer and in other cancers such as osteosarcoma and bone-dominant breast cancer. Targeted α-therapy strategy is also being actively explored through many preclinical and few early clinical studies using 225Ac, 213Bi, 211At, 227Th, and 212Pb. Investigations incorporating 225Ac are more robust and active at this time with promising results. The author provide a brief synopsis of the preclinical and clinical studies in the rapidly evolving field of targeted α-therapy in cancer management.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
5
|
Tafreshi NK, Doligalski ML, Tichacek CJ, Pandya DN, Budzevich MM, El-Haddad G, Khushalani NI, Moros EG, McLaughlin ML, Wadas TJ, Morse DL. Development of Targeted Alpha Particle Therapy for Solid Tumors. Molecules 2019; 24:molecules24234314. [PMID: 31779154 PMCID: PMC6930656 DOI: 10.3390/molecules24234314] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Targeted alpha-particle therapy (TAT) aims to selectively deliver radionuclides emitting α-particles (cytotoxic payload) to tumors by chelation to monoclonal antibodies, peptides or small molecules that recognize tumor-associated antigens or cell-surface receptors. Because of the high linear energy transfer (LET) and short range of alpha (α) particles in tissue, cancer cells can be significantly damaged while causing minimal toxicity to surrounding healthy cells. Recent clinical studies have demonstrated the remarkable efficacy of TAT in the treatment of metastatic, castration-resistant prostate cancer. In this comprehensive review, we discuss the current consensus regarding the properties of the α-particle-emitting radionuclides that are potentially relevant for use in the clinic; the TAT-mediated mechanisms responsible for cell death; the different classes of targeting moieties and radiometal chelators available for TAT development; current approaches to calculating radiation dosimetry for TATs; and lead optimization via medicinal chemistry to improve the TAT radiopharmaceutical properties. We have also summarized the use of TATs in pre-clinical and clinical studies to date.
Collapse
Affiliation(s)
- Narges K. Tafreshi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Michael L. Doligalski
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Christopher J. Tichacek
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Darpan N. Pandya
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - Mikalai M. Budzevich
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Ghassan El-Haddad
- Depts. of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nikhil I. Khushalani
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Eduardo G. Moros
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Mark L. McLaughlin
- Department of Pharmaceutical Sciences, West Virginia University, Health Sciences Center, Morgantown, WV & Modulation Therapeutics Inc., 64 Medical Center Drive, Morgantown, WV 26506, USA;
| | - Thaddeus J. Wadas
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - David L. Morse
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-745-8948; Fax: +1-813-745-8375
| |
Collapse
|
6
|
Ni J, Bongers A, Chamoli U, Bucci J, Graham P, Li Y. In Vivo 3D MRI Measurement of Tumour Volume in an Orthotopic Mouse Model of Prostate Cancer. Cancer Control 2019; 26:1073274819846590. [PMID: 31032634 PMCID: PMC6488786 DOI: 10.1177/1073274819846590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed cancer in males in western
countries. Orthotopic implantation is considered as an ideal xenograft model for
CaP study, and noninvasive measurement of tumor volume changes is important for
monitoring responses to anticancer therapies. In this study, the T2-weighted
fast spin echo sequence magnetic resonance imaging (MRI) was performed on a CaP
orthotopic non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mouse
model weekly for 6 weeks post PC-3 CaP cell inoculation, and the fat signal was
suppressed using a chemical shift-selective pulse. Subsequently, the MRI data
were imported into the image processing software Avizo Standard and stacked into
three-dimensional (3D) volumes. Our results demonstrate that MRI, combined with
3D reconstruction, is a feasible and sensitive method to assess tumor growth in
a PC-3 orthotopic CaP mouse model and this established monitoring approach is
promising for longitudinal observation of CaP xenograft development after
anticancer therapy in vivo. Further investigation is needed to
validate this protocol in a larger cohort of mice to generate enough statistical
power.
Collapse
Affiliation(s)
- Jie Ni
- 1 Cancer Care Centre, St George Hospital, Kogarah, New South Wales, Australia.,2 St George and Sutherland Clinical School, UNSW Sydney, New South Wales, Australia
| | - Andre Bongers
- 3 Biological Resource Imaging Laboratory, UNSW Sydney, New South Wales, Australia
| | - Uphar Chamoli
- 4 Spine Service, Department of Orthopaedic Surgery, St George and Sutherland Clinical School, UNSW Sydney, Kogarah, New South Wales, Australia.,5 School of Biomedical Engineering, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Joseph Bucci
- 1 Cancer Care Centre, St George Hospital, Kogarah, New South Wales, Australia.,2 St George and Sutherland Clinical School, UNSW Sydney, New South Wales, Australia
| | - Peter Graham
- 1 Cancer Care Centre, St George Hospital, Kogarah, New South Wales, Australia.,2 St George and Sutherland Clinical School, UNSW Sydney, New South Wales, Australia
| | - Yong Li
- 1 Cancer Care Centre, St George Hospital, Kogarah, New South Wales, Australia.,2 St George and Sutherland Clinical School, UNSW Sydney, New South Wales, Australia.,6 School of Basic Medical Sciences, Zhengzhou University, Henan, China
| |
Collapse
|
7
|
Ni J, Cozzi P, Beretov J, Duan W, Bucci J, Graham P, Li Y. Epithelial cell adhesion molecule (EpCAM) is involved in prostate cancer chemotherapy/radiotherapy response in vivo. BMC Cancer 2018; 18:1092. [PMID: 30419852 PMCID: PMC6233586 DOI: 10.1186/s12885-018-5010-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022] Open
Abstract
Background Development of chemo−/radioresistance is a major challenge for the current prostate cancer (CaP) therapy. We have previously demonstrated that epithelial cell adhesion molecule (EpCAM) is associated with CaP growth and therapeutic resistance in vitro, however, the role of EpCAM in CaP in vivo is not fully elucidated. Here, we aimed to investigate how expression of EpCAM is involved in CaP growth and chemo−/radiotherapy response in NOD/SCID mouse models in vivo and to validate its role as a therapeutic target for CaP therapy. Methods EpCAM was knocked down in PC-3 CaP cell line using short hairpin RNA (shRNA). The effect of EpCAM-knockdown (KD) on tumour growth, chemo−/radiotherapy response and animal survival was evaluated on subcutaneous (s.c) and orthotopic mouse models. Results We found that KD of EpCAM significantly inhibited tumour growth, increased xenograft sensitivity to chemotherapy/radiotherapy, and prolonged the survival of tumour-bearing mice. In addition, we demonstrated that KD of EpCAM is associated with downregulation of the PI3K/Akt/mTOR pathway. Conclusions In conclusion, our data confirms that CaP growth and chemo−/radioresistance in vivo is associated with over-expression of EpCAM, which serves both a functional biomarker and promising therapeutic target. Electronic supplementary material The online version of this article (10.1186/s12885-018-5010-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Paul Cozzi
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Department of Surgery, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Julia Beretov
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Anatomical Pathology, NSW Health Pathology, St George Hospital, Gray St, Kogarah, NSW, 2217, Australia
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - Joseph Bucci
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Peter Graham
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia. .,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia. .,School of Basic Medical Sciences, Zhengzhou University, Henan, 450001, China.
| |
Collapse
|
8
|
Hao J, Graham P, Chang L, Ni J, Wasinger V, Beretov J, Deng J, Duan W, Bucci J, Malouf D, Gillatt D, Li Y. Proteomic identification of the lactate dehydrogenase A in a radioresistant prostate cancer xenograft mouse model for improving radiotherapy. Oncotarget 2018; 7:74269-74285. [PMID: 27708237 PMCID: PMC5342052 DOI: 10.18632/oncotarget.12368] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/15/2016] [Indexed: 12/11/2022] Open
Abstract
Radioresistance is a major challenge for prostate cancer (CaP) metastasis and recurrence after radiotherapy. This study aimed to identify potential protein markers and signaling pathways associated with radioresistance using a PC-3 radioresistant (RR) subcutaneous xenograft mouse model and verify the radiosensitization effect from a selected potential candidate. PC-3RR and PC-3 xenograft tumors were established and differential protein expression profiles from two groups of xenografts were analyzed using liquid chromatography tandem-mass spectrometry. One selected glycolysis marker, lactate dehydrogenase A (LDHA) was validated, and further investigated for its role in CaP radioresistance. We found that 378 proteins and 51 pathways were significantly differentially expressed between PC-3RR and PC-3 xenograft tumors, and that the glycolysis pathway is closely linked with CaP radioresistance. In addition, we also demonstrated that knock down of LDHA with siRNA or inhibition of LDHA activity with a LDHA specific inhibitor (FX-11), could sensitize PC-3RR cells to radiotherapy with reduced epithelial-mesenchymal transition, hypoxia, DNA repair ability and autophagy, as well as increased DNA double strand breaks and apoptosis. In summary, we identified a list of potential RR protein markers and important signaling pathways from a PC-3RR xenograft mouse model, and demonstrate that targeting LDHA combined with radiotherapy could increase radiosensitivity in RR CaP cells, suggesting that LDHA is an ideal therapeutic target to develop combination therapy for overcoming CaP radioresistance.
Collapse
Affiliation(s)
- Jingli Hao
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter Graham
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lei Chang
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jie Ni
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Valerie Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, Sydney, NSW 2052, Australia.,School of Medical Sciences, Sydney, NSW 2052, Australia
| | - Julia Beretov
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.,SEALS, Anatomical Pathology, St George Hospital, Kogarah, NSW 2217, Australia
| | - Junli Deng
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3217, Australia
| | - Joseph Bucci
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - David Malouf
- Department of Urology, St George Hospital, Kogarah, NSW 2217, Australia
| | - David Gillatt
- Department of Urology, St George Hospital, Kogarah, NSW 2217, Australia.,Australian School of Advanced Medicine, Macquarie University, Sydney, NSW 2019, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
9
|
Identification of protein biomarkers and signaling pathways associated with prostate cancer radioresistance using label-free LC-MS/MS proteomic approach. Sci Rep 2017; 7:41834. [PMID: 28225015 PMCID: PMC5320484 DOI: 10.1038/srep41834] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/28/2016] [Indexed: 12/20/2022] Open
Abstract
Identifying biomarkers and signaling pathways are important for the management of prostate cancer (CaP) radioresistance. In this study, we identified differential proteins and signaling pathways from parental CaP cell lines and CaP radioresistant (RR) sublines using a label-free LC-MS/MS proteomics approach. A total of 309 signaling pathway proteins were identified to be significantly altered between CaP and CaP-RR cells (p ≤ 0.05, fold differences >1.5, ≥80% power). Among these proteins, nineteen are common among three paired CaP cell lines and associated with metastasis, progression and radioresistance. The PI3K/Akt, VEGF and glucose metabolism pathways were identified as the main pathways associated with CaP radioresistance. In addition, the identified potential protein markers were further validated in CaP-RR cell lines and subcutaneous (s.c) animal xenografts by western blotting and immunohistochemistry, respectively and protein aldolase A (ALDOA) was selected for a radiosensitivity study. We found the depletion of ALDOA combined with radiotherapy effectively reduced colony formation, induced more apoptosis and increased radiosensitivity in CaP-RR cells. Our findings indicate that CaP radioresistance is caused by multifactorial traits and downregulation of ALDOA increases radiosensitivity in CaP-RR cells, suggesting that controlling these identified proteins or signaling pathways in combination with radiotherapy may hold promise to overcome CaP radioresistance.
Collapse
|
10
|
Truong Q, Justiniano IO, Nocon AL, Soon JT, Wissmueller S, Campbell DH, Walsh BJ. Glypican-1 as a Biomarker for Prostate Cancer: Isolation and Characterization. J Cancer 2016; 7:1002-9. [PMID: 27313791 PMCID: PMC4910593 DOI: 10.7150/jca.14645] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/22/2016] [Indexed: 01/21/2023] Open
Abstract
Prostate cancer is the most frequently diagnosed male visceral cancer and the second leading cause of cancer death in the United States. Standard tests such as prostate-specific antigen (PSA) measurement have poor specificity (33%) resulting in a high number of false positive reports. Consequently there is a need for new biomarkers to address this problem. The MIL-38 antibody was first described nearly thirty years ago, however, until now, the identification of the target antigen remained elusive. By a series of molecular techniques and mass spectrometry, the MIL-38 antigen was identified to be the highly glycosylated proteoglycan Glypican-1 (GPC-1). This protein is present in two forms; a membrane bound core protein of 55-60 kDa and secreted soluble forms of 40 kDa and 52 kDa. GPC-1 identification was confirmed by immuno-precipitation, western blots and ELISA. An ELISA platform is currently being developed to assess the levels of GPC-1 in normal, benign prostatic hyperplasia (BPH) and prostate cancer patients to determine whether secreted GPC-1 may represent a clinically relevant biomarker for prostate cancer diagnosis.
Collapse
Affiliation(s)
- Quach Truong
- Minomic International Ltd, Suite 2, Ground Floor, 75 Talavera Rd, Macquarie Park, NSW 2113, Australia
| | - Irene O Justiniano
- Minomic International Ltd, Suite 2, Ground Floor, 75 Talavera Rd, Macquarie Park, NSW 2113, Australia
| | - Aline L Nocon
- Minomic International Ltd, Suite 2, Ground Floor, 75 Talavera Rd, Macquarie Park, NSW 2113, Australia
| | - Julie T Soon
- Minomic International Ltd, Suite 2, Ground Floor, 75 Talavera Rd, Macquarie Park, NSW 2113, Australia
| | - Sandra Wissmueller
- Minomic International Ltd, Suite 2, Ground Floor, 75 Talavera Rd, Macquarie Park, NSW 2113, Australia
| | - Douglas H Campbell
- Minomic International Ltd, Suite 2, Ground Floor, 75 Talavera Rd, Macquarie Park, NSW 2113, Australia
| | - Bradley J Walsh
- Minomic International Ltd, Suite 2, Ground Floor, 75 Talavera Rd, Macquarie Park, NSW 2113, Australia
| |
Collapse
|
11
|
Ni J, Cozzi P, Hung TT, Hao J, Graham P, Li Y. Monitoring Prostate Tumor Growth in an Orthotopic Mouse Model Using Three-Dimensional Ultrasound Imaging Technique. Transl Oncol 2016; 9:41-45. [PMID: 26947880 PMCID: PMC4800064 DOI: 10.1016/j.tranon.2015.11.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/10/2015] [Indexed: 02/03/2023] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed and the second leading cause of death from cancer in males in USA. Prostate orthotopic mouse model has been widely used to study human CaP in preclinical settings. Measurement of changes in tumor size obtained from noninvasive diagnostic images is a standard method for monitoring responses to anticancer modalities. This article reports for the first time the usage of a three-dimensional (3D) ultrasound system equipped with photoacoustic (PA) imaging in monitoring longitudinal prostate tumor growth in a PC-3 orthotopic NODSCID mouse model (n = 8). Two-dimensional and 3D modes of ultrasound show great ability in accurately depicting the size and shape of prostate tumors. PA function on two-dimensional and 3D images showed average oxygen saturation and average hemoglobin concentration of the tumor. Results showed a good fit in representative exponential tumor growth curves (n = 3; r2 = 0.948, 0.955, and 0.953, respectively) and a good correlation of tumor volume measurements performed in vivo with autopsy (n = 8, r = 0.95, P < .001). The application of 3D ultrasound imaging proved to be a useful imaging modality in monitoring tumor growth in an orthotopic mouse model, with advantages such as high contrast, uncomplicated protocols, economical equipment, and nonharmfulness to animals. PA mode also enabled display of blood oxygenation surrounding the tumor and tumor vasculature and angiogenesis, making 3D ultrasound imaging an ideal tool for preclinical cancer research.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, the University of New South Wales (UNSW), Kensington, NSW 2052, Australia
| | - Paul Cozzi
- St George and Sutherland Clinical School, the University of New South Wales (UNSW), Kensington, NSW 2052, Australia; Department of Surgery, St George Hospital, Kogarah, NSW 2217, Australia
| | - Tzong-Tyng Hung
- Biological Resource Imaging Laboratory, the University of New South Wales (UNSW), Kensington, NSW 2052, Australia
| | - Jingli Hao
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, the University of New South Wales (UNSW), Kensington, NSW 2052, Australia
| | - Peter Graham
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, the University of New South Wales (UNSW), Kensington, NSW 2052, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, the University of New South Wales (UNSW), Kensington, NSW 2052, Australia.
| |
Collapse
|
12
|
|
13
|
Guseva LI. Radioisotope generators of short-lived α-emitting radionuclides promising for use in nuclear medicine. RADIOCHEMISTRY 2014. [DOI: 10.1134/s1066362214050014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Eriksson SE, Elgström E, Bäck T, Ohlsson T, Jensen H, Nilsson R, Lindegren S, Tennvall J. Sequential Radioimmunotherapy with 177Lu- and 211At-Labeled Monoclonal Antibody BR96 in a Syngeneic Rat Colon Carcinoma Model. Cancer Biother Radiopharm 2014; 29:238-46. [DOI: 10.1089/cbr.2014.1625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Erika Elgström
- Department of Oncology and Pathology, Lund University, Lund, Sweden
| | - Tom Bäck
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tomas Ohlsson
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Holger Jensen
- Cyclotron and PET Unit, Rigshospitalet, Copenhagen, Denmark
| | - Rune Nilsson
- Department of Oncology and Pathology, Lund University, Lund, Sweden
| | - Sture Lindegren
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Tennvall
- Department of Oncology and Pathology, Lund University, Lund, Sweden
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
15
|
Clarke RA, Allen BJ. Next-generation therapy for residual prostate cancer. Immunotherapy 2014; 5:1235-41. [PMID: 24188677 DOI: 10.2217/imt.13.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prostate cancer claimed an estimated 136,500 lives globally in 2011. Ironically, the best existing treatment strategies provide survival benefits and missed opportunities to further improve outcomes. Prostatectomy provides the greatest survival benefit, albeit the risk of systemic recurrence increases dramatically with extracapsular involvement. To date, further systemic treatment is not generally prescribed for these 'high-risk' patients until such time as advanced disease is diagnosed based on persistent high PSA levels and/or when larger tumors are confirmed by imaging. This recurrent form of the disease is most often terminal. Androgen deprivation therapy (ADT) provides outstanding early control for these patients, which is rather tragic as the early benefits of ADT are lost within 2 years for most men, as the cancer again progresses to an incurable 'late-stage' castration-resistant form of the disease with a median survival of approximately 18 months. We review the potential of targeted α-therapy as an adjuvant with minimal side effects for early-stage high-risk patients to be administered immediately following prostatectomy and/or during ADT.
Collapse
Affiliation(s)
- Raymond A Clarke
- Ingham Institute, School of Medicine, University of Western Sydney, Liverpool 2170, NSW, Australia.
| | | |
Collapse
|
16
|
Abstract
α-particle-emitting radionuclides are highly cytotoxic and are thus promising candidates for use in targeted radioimmunotherapy of cancer. Due to their high linear energy transfer (LET) combined with a short path length in tissue, α-particles cause severe DNA double-strand breaks that are repaired inaccurately and finally trigger cell death. For radioimmunotherapy, α-emitters such as 225Ac, 211At, 212Bi/212Pb, 213Bi and 227Th are coupled to antibodies via appropriate chelating agents. The α-emitter immunoconjugates preferably target proteins that are overexpressed or exclusively expressed on cancer cells. Application of α-emitter immunoconjugates seems particularly promising in treatment of disseminated cancer cells and small tumor cell clusters that are released during the resection of a primary tumor. α-emitter immunoconjugates have been successfully administered in numerous experimental studies for therapy of ovarian, colon, gastric, blood, breast and bladder cancer. Initial clinical trials evaluating α-emitter immunoconjugates in terms of toxicity and therapeutic efficacy have also shown positive results in patients with melanoma, ovarian cancer, acute myeloid lymphoma and glioma. The present problems in terms of availability of therapeutically effiective α-emitters will presumably be solved by use of alternative production routes and installation of additional production facilities in the near future. Therefore, clinical establishment of targeted α-emitter radioimmunotherapy as one part of a multimodal concept for therapy of cancer is a promising, middle-term concept.
Collapse
Affiliation(s)
- Christof Seidl
- Technische Universität München, Department of Nuclear Medicine, Ismaninger Strasse 22, 81675 Munich, Germany
| |
Collapse
|
17
|
Comparison between internalizing anti-HER2 mAbs and non-internalizing anti-CEA mAbs in alpha-radioimmunotherapy of small volume peritoneal carcinomatosis using 212Pb. PLoS One 2013; 8:e69613. [PMID: 23922757 PMCID: PMC3726680 DOI: 10.1371/journal.pone.0069613] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 06/10/2013] [Indexed: 11/25/2022] Open
Abstract
Background and Purpose We assessed the contribution of antibody internalization in the efficacy and toxicity of intraperitoneal α-radioimmunotherapy (RIT) of small volume carcinomatosis using 212Pb-labeled monoclonal antibodies (mAbs) that target HER2 (internalizing) or CEA (non-internalizing) receptors. Materials and Methods Athymic nude mice bearing 2–3 mm intraperitoneal tumor xenografts were intraperitoneally injected with similar activities (370, 740 and 1480 kBq; 37 MBq/mg) of 212Pb-labeled 35A7 (anti-CEA), trastuzumab (anti-HER2) or PX (non-specific) mAbs, or with equivalent amounts of unlabeled mAbs, or with NaCl. Tumor volume was monitored by bioluminescence and survival was reported. Hematologic toxicity and body weight were assessed. Biodistribution of 212Pb-labeled mAbs and absorbed dose-effect relationships using MIRD formalism were established. Results Transient hematological toxicity, as revealed by white blood cells and platelets numbering, was reported in mice treated with the highest activities of 212Pb-labeled mAbs. The median survival (MS) was significantly higher in mice injected with 1.48 MBq of 212Pb-35A7 (non-internalizing mAbs) (MS = 94 days) than in animals treated with the same activity of 212Pb-PX mAbs or with NaCl (MS = 18 days). MS was even not reached after 130 days when follow-up was discontinued in mice treated with 1.48 MBq of 212Pb-trastuzumab. The later efficacy was unexpected since final absorbed dose resulting from injection of 1.48 MBq, was higher for 212Pb-35A7 (35.5 Gy) than for 212Pb-trastuzumab (27.6 Gy). These results also highlight the lack of absorbed dose-effect relationship when mean absorbed dose was calculated using MIRD formalism and the requirement to perform small-scale dosimetry. Conclusions These data indicate that it might be an advantage of using internalizing anti-HER2 compared with non-internalizing anti-CEA 212Pb-labeled mAbs in the therapy of small volume xenograft tumors. They support clinical investigations of 212Pb-mAbs RIT as an adjuvant treatment after cytoreductive surgery in patients with peritoneal carcinomatosis.
Collapse
|
18
|
Rizvi S, Allen B, Lee C, Bruchertseifer F, Apostolidis C, Morgenstern A, Clarke R. Orthotopic administration of (213)Bi-bevacizumab inhibits progression of PC3 xenografts in the prostate. Immunotherapy 2012; 4:549-54. [PMID: 22642336 DOI: 10.2217/imt.12.42] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To investigate orthotopic targeted α-radioimmunotherapy for the control of early-stage PC3 prostate cancer nude mouse xenografts using the radiolabeled bevacizumab (BZ) immunoconjugate ((213)Bi-BZ), which emits short-range α-radiation. MATERIALS & METHODS 10(6) PC3 human prostate cancer cells were injected into the lower capsule of the mouse prostate gland 1 week prior to α-radioimmunotherapy. Mice were euthanized and assessed for tumour growth at 2 (two mice), 4 (two mice) and 6 weeks (three mice) post-therapy. The no-therapy control mice received a saline injection in equal volume to each BZ administration. RESULTS (213)Bi-BZ is significantly more efficacious in inhibiting xenograft progression in the prostate gland compared with BZ alone (p = 0.009) and when compared with the 'no therapy' protocol (p < 0.0001). CONCLUSION Orthotopic administration of (213)Bi-BZ greatly improves the early control of organ-confined prostate cancer compared with BZ alone (p < 0.01).
Collapse
Affiliation(s)
- Sma Rizvi
- Cancer Pathology & Cell Biology Laboratory, Ingham Institute of Applied Medical Research, & Discipline of Pathology, University of Western Sydney School of Medicine, Liverpool 2170, Australia
| | | | | | | | | | | | | |
Collapse
|
19
|
Essler M, Gärtner FC, Neff F, Blechert B, Senekowitsch-Schmidtke R, Bruchertseifer F, Morgenstern A, Seidl C. Therapeutic efficacy and toxicity of 225Ac-labelled vs. 213Bi-labelled tumour-homing peptides in a preclinical mouse model of peritoneal carcinomatosis. Eur J Nucl Med Mol Imaging 2012; 39:602-12. [DOI: 10.1007/s00259-011-2023-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 12/02/2011] [Indexed: 10/14/2022]
|