1
|
Sharma R, Mukherjee A, Kumar A, Sarma HD. Evaluation of 177Lu-Labeled Pertuzumab F(ab') 2 Fragments for HER2-Positive Cancer Targeting: A Comparative In Vitro and In Vivo Study. Cancer Biother Radiopharm 2024; 39:64-74. [PMID: 38363819 DOI: 10.1089/cbr.2023.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024] Open
Abstract
Background: Radiolabeled antibody fragments present a promising opportunity as theranostic agents, offering distinct advantages over whole antibodies. In this study, the authors investigate the potential of [177Lu]Lu-DTPA-F(ab')2-pertuzumab as a theranostic agent for precise targeting of human epidermal growth factor receptor 2 (HER2)-positive cancers. Additionally, the authors aim to quantitatively assess the binding synergism in the presence of cold trastuzumab. Materials and Methods: F(ab')2-pertuzumab was prepared by pepsin digestion and conjugated with a bifunctional chelator. The immunoconjugate was radiolabeled with 177Lu and characterized by chromatography techniques. Binding parameters (affinity, specificity, and immunoreactivity) and cellular binding enhancement studies were evaluated in HER2-overexpressing and triple-negative cell lines. The in vivo enhancement in tumor uptake of the radiolabeled immunoformulation was assessed in severe combined immunodeficient (SCID) mice bearing tumors, both in the presence and absence of unlabeled trastuzumab. Results: The formulation of [177Lu]Lu-DTPA-F(ab')2-pertuzumab could be prepared in high yields and with consistent radiochemical purity, ensuring reproducibility. Comprehensive in vitro and in vivo evaluation studies confirmed high specificity and immunoreactivity of the formulation toward HER2 receptors. Binding synergism of radiolabeled pertuzumab fragments in the presence of trastuzumab to HER2 receptors was observed. Conclusions: The radioformulation of [177Lu]Lu-DTPA-F(ab')2-pertuzumab holds great promise as a targeted approach for addressing HER2-positive cancers. A potentially effective strategy to amplify therapeutic efficacy involves dual epitope targeting by combining radiolabeled pertuzumab with cold trastuzumab.
Collapse
Affiliation(s)
- Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Anuj Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| | - Haladhar Dev Sarma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| |
Collapse
|
2
|
Rathore Y, Shukla J, Laroiya I, Deep A, Lakhanpal T, Kumar R, Singh H, Bal A, Singh G, Gopal Thakur K, Mittal BR. Development 68Ga trastuzumab Fab and bioevaluation by PET imaging in HER2/neu expressing breast cancer patients. Nucl Med Commun 2022; 43:458-467. [PMID: 35131966 DOI: 10.1097/mnm.0000000000001521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Receptors on breast cancer cells play a crucial role in the management of patients. Trastuzumab is a widely used drug for the treatment of HER2/neu expressing tumors. ImmunoPET with trastuzumab is not feasible due to slow pharmacokinetics. Fragment of antigen-binding (Fab) radiolabeled with positron emitters can be used for immunoPET. METHODS Fab has been generated by papain digestion and conjugated with the bifunctional chelating agent NOTA. The SDS-PAGE and MALDI-TOF were used to see the integrity of Fab and conjugated Fab. In-vitro stability and target specificity for HER2/neu receptors were performed in plasma and receptor binding with bio-layer interferometry (BLI) techniques. Radiolabeling was standardized with 68GaCl3 and PET imaging was performed in seven patients showing 18F fluorodeoxyglucose (18F-FDG) uptake and correlated with HER2/neu expression by immunohistochemistry. RESULTS Fab production was optimized at molar ratio 23:1 of trastuzumab and papain at 37 °C with a constant stirrer at 850 rpm for 22-24 h, at pH 8. Conjugation with NOTA was standardized at molar ratio 1:25 of trastuzumab Fab and NOTA. Molecular mass of trastuzumab Fab-NOTA was found approximately 46.3 kDa (~1/3 of intact antibody). Trastuzumab Fab-NOTA showed radiolabelling efficiency of 48-70% with incubation time 15 min at 37-40 °C and pH 4.5-5.0. BLI demonstrated the affinity of trastuzumab, trastuzumab Fab and trastuzumab Fab-NOTA towards HER2/neu receptor with KD of <1pM, ~0.5nM and ~20nM, respectively. All immunohistochemistry proven patients showed uptake in primary breast lesion and lymph nodes. CONCLUSION Trastuzumab Fab-NOTA is suitable for radiolabelling with 68Ga and ImmunoPET imaging of HER2/neu receptor.
Collapse
Affiliation(s)
- Yogesh Rathore
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research
| | - Jaya Shukla
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research
| | - Ishita Laroiya
- Department of General Surgery, Post Graduate Institute of Medical Education and Research
| | - Amar Deep
- Structural Biology Laboratory, CSIR-Institute of Microbial Technology (IMTECH)
| | - Tamanna Lakhanpal
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research
| | - Rajender Kumar
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research
| | - Harmandeep Singh
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research
| | - Amanjit Bal
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh India
| | - Gurpreet Singh
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh India
| | | | - B R Mittal
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research
| |
Collapse
|
3
|
Radiotherapy enhances uptake and efficacy of 90Y-cetuximab: A preclinical trial. Radiother Oncol 2021; 155:285-292. [DOI: 10.1016/j.radonc.2020.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 01/09/2023]
|
4
|
Marquez J, Dong J, Dong C, Tian C, Serrero G. Identification of Prostaglandin F2 Receptor Negative Regulator (PTGFRN) as an internalizable target in cancer cells for antibody-drug conjugate development. PLoS One 2021; 16:e0246197. [PMID: 33503070 PMCID: PMC7840024 DOI: 10.1371/journal.pone.0246197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/14/2021] [Indexed: 11/19/2022] Open
Abstract
Antibody-drug conjugates (ADC) are effective antibody-based therapeutics for hematopoietic and lymphoid tumors. However, there is need to identify new targets for ADCs, particularly for solid tumors and cancers with unmet needs. From a hybridoma library developed against cancer cells, we selected the mouse monoclonal antibody 33B7, which was able to bind to, and internalize, cancer cell lines. This antibody was used for identification of the target by immunoprecipitation and mass spectrometric analysis, followed by target validation. After target validation, 33B7 binding and target positivity were tested by flow cytometry and western blot analysis in several cancer cell lines. The ability of 33B7 conjugated to saporin to inhibit in vitro proliferation of PTFRN positive cell lines was investigated, as well as the 33B7 ADC in vivo effect on tumor growth in athymic mice. All flow cytometry and in vitro internalization assays were analyzed for statistical significance using a Welsh's T-test. Animal studies were analyzed using Two-Way Analysis of Variance (ANOVA) utilizing post-hoc Bonferroni analysis, and/or Mixed Effects analysis. The 33B7 cell surface target was identified as Prostaglandin F2 Receptor Negative Regulator (PTGFRN), a transmembrane protein in the Tetraspanin family. This target was confirmed by showing that PTGFRN-expressing cells bound and internalized 33B7, compared to PTGFRN negative cells. Cells able to bind 33B7 were PTGFRN-positive by Western blot analysis. In vitro treatment PTGFRN-positive cancer cell lines with the 33B7-saporin ADC inhibited their proliferation in a dose-dependent fashion. 33B7 conjugated to saporin was also able to block tumor growth in vivo in mouse xenografts when compared to a control ADC. These findings show that screening antibody libraries for internalizing antibodies in cancer cell lines is a good approach to identify new cancer targets for ADC development. These results suggest PTGFRN is a possible therapeutic target via antibody-based approach for certain cancers.
Collapse
Affiliation(s)
- Jorge Marquez
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore School of Pharmacy, Baltimore, Maryland, United States of America
| | - Jianping Dong
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Chun Dong
- Precision Antibody Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Changsheng Tian
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Ginette Serrero
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
- Precision Antibody Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| |
Collapse
|
5
|
Sharma R, Kameswaran M, Dash A. Comparative In Vitro Cytotoxicity Studies of 177Lu-CHX-A″-DTPA-Trastuzumab and 177Lu-CHX-A″-DTPA-F(ab') 2-Trastuzumab in HER2-Positive Cancer Cell Lines. Cancer Biother Radiopharm 2020; 35:177-189. [PMID: 32196365 DOI: 10.1089/cbr.2019.2882] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Human epidermal growth factor receptor 2 (HER2) is found to be amplified in ∼15%-20% of breast cancers. In this study, the authors report the synthesis and comparative in vitro therapeutic efficacy of 177Lu-CHX-A″-DTPA-trastuzumab and 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab to determine their potential as theranostic agents for patients with breast cancer. Materials and Methods: Bivalent F(ab')2-trastuzumab was produced by enzymatic digestion of trastuzumab, conjugated with p-SCN-Bn-CHX-A″-DTPA and subsequently radiolabeled with 177Lu. Cell viability, membrane toxicity assays, and apoptosis analysis were carried out with 177Lu-CHX-A″-DTPA-trastuzumab and 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab in HER2-positive ovarian (SK-OV-3) and breast cancer (SK-BR-3 and MDA-MB-453) cells. Results: In vitro cell binding studies showed ∼20%-25% binding of 177Lu-CHX-A″-DTPA-trastuzumab and 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab to SK-OV-3, SK-BR-3, and MDA-MB-453 cells. The cells exhibited similar degree of membrane integrity and cellular toxicity when treated with same amount (activity) of 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab and 177Lu-CHX-A″-DTPA-trastuzumab, and the toxicity was dose dependent. The mode of cell death was predominantly by apoptosis and necrosis with both the radioimmunoconjugates. Conclusions: The results indicated that the efficacy of both the radioimmunoconjugates, in terms of inducing cell death, was similar thereby ascertaining their potential as good therapeutic agents for patients with breast cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Mythili Kameswaran
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
6
|
Hermanto S, Haryuni RD, Ramli M, Mutalib A, Hudiyono S. Synthesis and stability test of radioimmunoconjugate 177Lu-DOTA-F(ab′)2-trastuzumab for theranostic agent of HER2 positive breast cancer. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2019. [DOI: 10.1016/j.jrras.2016.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Sandra Hermanto
- Department of Chemistry, Faculty of Mathematics and Science, University of Indonesia, Depok, 16424, Indonesia
- Study Program of Chemistry, Faculty of Science and Technology, Syarif Hidayatullah State Islamic University, Jl. Ir, H. Juanda No. 95, Ciputat, Jakarta, 15412, Indonesia
| | - Ratna Dini Haryuni
- Centre for Radioisotopes and Radiopharmaceuticals, National Nuclear Energy Agency of Indonesia, Kawasan Puspiptek, Setu, Kota Tangerang Selatan, Banten, 15314, Indonesia
| | - Martalena Ramli
- Centre for Radioisotopes and Radiopharmaceuticals, National Nuclear Energy Agency of Indonesia, Kawasan Puspiptek, Setu, Kota Tangerang Selatan, Banten, 15314, Indonesia
| | - Abdul Mutalib
- Department of Chemistry, Faculty of Mathematics and Science, Padjadjaran University, Jl. Raya Bandung-Sumedang Km. 21, Jatinangor, Jawa Barat, Indonesia
| | - Sumi Hudiyono
- Department of Chemistry, Faculty of Mathematics and Science, University of Indonesia, Depok, 16424, Indonesia
| |
Collapse
|
7
|
Envisaging an alpha therapy programme in the atomic energy establishments: the priorities and the nuances. Eur J Nucl Med Mol Imaging 2017; 44:1244-1246. [PMID: 28364163 DOI: 10.1007/s00259-017-3686-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 01/22/2023]
|
8
|
Houghton JL, Membreno R, Abdel-Atti D, Cunanan KM, Carlin S, Scholz WW, Zanzonico PB, Lewis JS, Zeglis BM. Establishment of the In Vivo Efficacy of Pretargeted Radioimmunotherapy Utilizing Inverse Electron Demand Diels-Alder Click Chemistry. Mol Cancer Ther 2016; 16:124-133. [PMID: 28062708 DOI: 10.1158/1535-7163.mct-16-0503] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/20/2016] [Accepted: 11/01/2016] [Indexed: 12/25/2022]
Abstract
The pretargeting system based on the inverse electron demand Diels-Alder reaction (IEDDA) between trans-cyclooctene (TCO) and tetrazine (Tz) combines the favorable pharmacokinetic properties of radiolabeled small molecules with the affinity and specificity of antibodies. This strategy has proven to be an efficient method for the molecularly targeted delivery of pharmaceuticals, including isotopes for radiological imaging. Despite encouraging results from in vivo PET imaging studies, this promising system has yet to be thoroughly evaluated for pretargeted radioimmunotherapy (PRIT). Toward that end, we synthesized two novel 177Lu-labeled tetrazine-bearing radioligands. Next, we compared the usefulness of our ligands for PRIT when paired with TCO-modified 5B1-a human, anti-CA19.9 mAb-in preclinical murine models of pancreatic cancer. The exemplary ligand, 177Lu-DOTA-PEG7-Tz, showed rapid (4.6 ± 0.8% ID/g at 4 hours) and persistent (16.8 ± 3.9% ID/g at 120 hours) uptake in tumors while concurrently clearing from blood and nontarget tissues. Single-dose therapy studies using 5B1-TCO and varying amounts of 177Lu-DOTA-PEG7-Tz (400, 800, and 1,200 μCi) showed that our system elicits a dose-dependent therapeutic response in mice bearing human xenografts. Furthermore, dosimetry calculations suggest that our approach is amenable to clinical applications with its excellent dosimetric profile in organs of clearance (i.e., liver and kidneys) as well as in dose-limiting tissues, such as red marrow. This study established that a pretargeted methodology utilizing the IEDDA reaction can rapidly and specifically deliver a radiotherapeutic payload to tumor tissue, thus illustrating its excellent potential for clinical translation. Mol Cancer Ther; 16(1); 124-33. ©2016 AACR.
Collapse
Affiliation(s)
- Jacob L Houghton
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rosemery Membreno
- Department of Chemistry, Hunter College and the Graduate Center of the City University of New York, New York, New York.,Ph.D. Program in Chemistry of the Graduate Center of the City University of New York, New York, New York
| | - Dalya Abdel-Atti
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kristen M Cunanan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean Carlin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Pat B Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Chemistry, Hunter College and the Graduate Center of the City University of New York, New York, New York.,Ph.D. Program in Chemistry of the Graduate Center of the City University of New York, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
9
|
Abstract
Through years of evolutionary selection pressures, organisms have developed potent toxins that coincidentally have marked antineoplastic activity. These natural products have been vital for the development of multiagent treatment regimens currently employed in cancer chemotherapy, and are used in the treatment of a variety of malignancies. Therefore, this review catalogs recent advances in natural product-based drug discovery via the examination of mechanisms of action and available clinical data to highlight the utility of these novel compounds in the burgeoning age of precision medicine. The review also highlights the recent development of antibody-drug conjugates and other immunotoxins, which are capable of delivering highly cytotoxic agents previously deemed too toxic to elicit therapeutic benefit preferentially to neoplastic cells. Finally, the review examines natural products not currently used in the clinic that have novel mechanisms of action, and may serve to supplement current chemotherapeutic protocols.
Collapse
|
10
|
Gudkov SV, Shilyagina NY, Vodeneev VA, Zvyagin AV. Targeted Radionuclide Therapy of Human Tumors. Int J Mol Sci 2015; 17:E33. [PMID: 26729091 PMCID: PMC4730279 DOI: 10.3390/ijms17010033] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 12/07/2015] [Accepted: 12/22/2015] [Indexed: 12/12/2022] Open
Abstract
Targeted radionuclide therapy is one of the most intensively developing directions of nuclear medicine. Unlike conventional external beam therapy, the targeted radionuclide therapy causes less collateral damage to normal tissues and allows targeted drug delivery to a clinically diagnosed neoplastic malformations, as well as metastasized cells and cellular clusters, thus providing systemic therapy of cancer. The methods of targeted radionuclide therapy are based on the use of molecular carriers of radionuclides with high affinity to antigens on the surface of tumor cells. The potential of targeted radionuclide therapy has markedly grown nowadays due to the expanded knowledge base in cancer biology, bioengineering, and radiochemistry. In this review, progress in the radionuclide therapy of hematological malignancies and approaches for treatment of solid tumors is addressed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya St, 3, Pushchino, Moscow 142290, Russia.
- Prokhorov Institute of General Physics, Russian Academy of Sciences, Vavilova St, 38, Moscow 119991, Russia.
| | - Natalya Yu Shilyagina
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
| | - Vladimir A Vodeneev
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
| | - Andrei V Zvyagin
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, Sydney 2109, Australia.
| |
Collapse
|
11
|
Trendowski M. PU-H71: An improvement on nature's solutions to oncogenic Hsp90 addiction. Pharmacol Res 2015; 99:202-16. [DOI: 10.1016/j.phrs.2015.06.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 12/26/2022]
|
12
|
Reitkopf-Brodutch S, Confino H, Schmidt M, Cooks T, Efrati M, Arazi L, Rath-Wolfson L, Marshak G, Kelson I, Keisari Y. Ablation of experimental colon cancer by intratumoral 224Radium-loaded wires is mediated by alpha particles released from atoms which spread in the tumor and can be augmented by chemotherapy. Int J Radiat Biol 2015; 91:179-86. [PMID: 25179346 DOI: 10.3109/09553002.2015.959666] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE We developed (224)Ra-loaded wires, which release by recoil alpha emitting nuclei into solid tumors and cause tumor cell killing. This research examined if the major damage was inflicted by alpha particles emitted from these atoms or by direct gamma and beta emissions from the inserted wires. We also examined the efficacy of this treatment against colon cancer in combination with chemotherapy. MATERIALS AND METHODS Mouse colon carcinomas (CT-26 xenografts), treated by intra-tumoral radioactive wires loaded with (224)Ra atoms were monitored for effects on tumor growth, intratumoral tissue damage and distribution of alpha emitting atoms. The effects were compared with those of (224)Ra-loaded wires coated with poly methyl methacrylate (PMMA), which blocks atom recoil. Similar experiments were performed with radioactive wires combined with systemic 5-FU. RESULTS (224)Ra-loaded wires inhibited tumor growth and formed necrotic areas inside the tumor. PMMA coated wires did not inhibit tumor growth, and caused minor intratumoral damage. Autoradiography images of tumors treated with (224)Ra-loaded wires revealed a spread of alpha emitters over several mm, whereas PMMA-coated wires showed no such spread. Injection of 5-FU with (224)Ra-loaded wires augmented tumor growth retardation and cure. CONCLUSIONS (224)Ra-loaded wires ablate solid tumors by the release of alpha-particle emitting atoms inside the tissue, an effect that can be enhanced by combining this method with chemotherapy.
Collapse
Affiliation(s)
- Shira Reitkopf-Brodutch
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Sedlacek O, Kucka J, Mattova J, Parizek M, Studenovsky M, Zadinova M, Pouckova P, Hruby M. Multistage-targeted pH-responsive polymer conjugate of Auger electron emitter: Optimized design and in vivo activity. Eur J Pharm Sci 2014; 63:216-25. [DOI: 10.1016/j.ejps.2014.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/17/2014] [Accepted: 07/31/2014] [Indexed: 12/31/2022]
|
14
|
Zheng SG, Xu HX, Guo LH, Liu LN, Lu F. The safety and treatment response of combination therapy of radioimmunotherapy and radiofrequency ablation for solid tumor: a study in vivo. PLoS One 2014; 9:e96539. [PMID: 24787957 PMCID: PMC4008584 DOI: 10.1371/journal.pone.0096539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 04/08/2014] [Indexed: 12/18/2022] Open
Abstract
OBJECTION To investigate the safety and treatment response of radioimmunotherapy (RIT) in combination with radiofrequency ablation (RFA) for the treatment of VX2 tumor on rabbit. MATERIALS AND METHODS A total of 36 rabbits bearing VX2 tumor on the thigh were randomly assigned into 3 groups (group I: 1-2 cm; group II: 2-3 cm; group III: 3-4 cm) and 4 subgroups (A: as control, just puncture the tumor using the RFA electrode without power output; B: RFA alone; C: 131I-chTNT intratumoral injection alone; D: RFA+131I-chTNT intratumoral injection 3 days later). The variation of blood assay, weight and survival among different groups and subgroups were used to assess the treatment safety. Ultrasound (US) was used to monitor and assess the tumor response after treatment. RESULTS According to the results of the weight and the blood assay among different groups, subgroups, and at two time points (one day before and the 16th day after treatment), no damages to the liver, kidney function and myelosuppression resulting from the treatment were found. No significant differences in survivals among the four subgroups (p = 0.087) were found. In addition, 131I-chTNT did not show significant inhibition effect on VX2 tumor progression according to US measurements. CONCLUSION 131I-chTNT intratumoral injection alone or in combination with RFA is relatively safe for rabbit without significant toxicity and shows no significant effect on the survival. The treatment response is not as satisfactory as anticipated.
Collapse
Affiliation(s)
- Shu-Guang Zheng
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
- Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui-Xiong Xu
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
- Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Le-Hang Guo
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Lin-Na Liu
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Feng Lu
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
| |
Collapse
|
15
|
Elgström E, Ljungberg O, Eriksson SE, Orbom A, Strand SE, Ohlsson TG, Nilsson R, Tennvall J. Change in cell death markers during (177)Lu-mAb radioimmunotherapy-induced rejection of syngeneic rat colon carcinoma. Cancer Biother Radiopharm 2014; 29:143-52. [PMID: 24693940 DOI: 10.1089/cbr.2013.1576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE To monitor cell death in tumors during the rejection process after treatment with an antibody radiolabeled with a β-emitter. METHODS Tumors during rejection after treatment with (177)Lu-labeled antibody BR96 and after administration of unlabeled BR96 were compared with untreated tumors from the same immunocompetent syngeneic rat tumor model. Cell death was monitored with the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and immunohistochemical staining of activated caspase-3 and γH2AX. These data were evaluated together with histopathological morphology, BR96-binding antigen expression, and (177)Lu radioactivity distribution imaged by digital autoradiography. RESULTS The untreated tumors showed staining for all the markers, mainly in and around the necrotic areas. One to 2 days p.i. large areas were stained with anti-γH2AX, followed by a slight decrease. Staining of activated caspase-3 was intense and extensive 1-2 days p.i., while found in and around necrotic areas 3-8 days p.i. TUNEL staining was similar to activated caspase-3 staining 1-2 days p.i. but more extensive than activated caspase-3 staining 3-4 days p.i. Digital autoradiography revealed activity concentration in granulation tissue from 1 day p.i. CONCLUSION Following radioimmunotherapy in an immunocompetent syngeneic colon carcinoma model, tumor cells did not only die through caspase-3-dependent apoptosis, but also by other mechanisms.
Collapse
Affiliation(s)
- Erika Elgström
- 1 Division of Oncology, Department of Clinical Sciences, Lund University , Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Hillyar CRT, Cornelissen B, Vallis KA. Uptake, internalization and nuclear translocation of radioimmunotherapeutic agents. Ther Deliv 2014; 5:319-35. [PMID: 24592956 DOI: 10.4155/tde.14.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
Radioimmunotherapy (RIT) agents that incorporate short-range particle-emitting radionuclides exploit the high linear energy transfer of α-particles and Auger electrons. Both are densely ionizing, generate complex DNA double-strand breaks and so are profoundly cytotoxic. Internalizing RIT agents enter tumor cells through receptor-mediated endocytosis and by incorporation of cell-penetrating peptides. Once internalized, some RIT agents mediate escape from endosomes and/or translocate to the nucleus. In the classical nuclear import pathway, α/β-importins recognize nuclear localization sequences in RIT agents. Translocation through nuclear pores enables RIT agents to bind to nuclear targets induced by, for example, cellular stress, growth factors or anticancer therapy, such as γH2AX or p27(KIP-1). This review discusses RIT agents designed to exploit the mechanisms underlying these complex processes and compares them with noninternalizing RIT agents.
Collapse
Affiliation(s)
- Christopher R T Hillyar
- Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology & Biology, Department of Oncology, University of Oxford, OX3 7DQ, UK
| | | | | |
Collapse
|
17
|
Keisari Y, Hochman I, Confino H, Korenstein R, Kelson I. Activation of local and systemic anti-tumor immune responses by ablation of solid tumors with intratumoral electrochemical or alpha radiation treatments. Cancer Immunol Immunother 2014; 63:1-9. [PMID: 23955682 PMCID: PMC11029492 DOI: 10.1007/s00262-013-1462-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/27/2013] [Indexed: 12/12/2022]
Abstract
Cancer, the most devastating chronic disease affecting humankind, is treated primarily by surgery, chemotherapy, and radiation therapy. Surgery and radiotherapy are mainly used for debulking the primary tumor, while chemotherapy is the most efficient anti-metastatic treatment. To control better metastatic cancer, the host immune system should be stimulated. Yet, successful specific stimulation of the immune system against tumors was seldom achieved even in antigenic tumors. Our working hypothesis is that aggressive in situ tumor ablation can release tumor antigens and danger signals, which will enhance anti-tumor T cell responses resulting in the destruction of residual malignant cells in primary tumors and distant metastases. We developed two efficient in situ ablation treatments for solid cancer, which can be used to destroy the primary tumors and stimulate anti-tumor immune responses. The first treatment, electrochemical ablation, is applied through intratumoral electrodes, which deliver unipolar-pulsed electric currents. The second treatment, diffusing alpha-emitters radiation therapy (DaRT), is based on intratumoral (224)Ra-loaded wire(s) that release by recoil its daughter atoms. These short-lived alpha-emitting atoms spread in the tumor and spray it with lethal alpha particles. It was confirmed that these treatments effectively destroy various malignant animal and human primary solid tumors. As a consequence of such tumor ablation, tumor-derived antigenic material was released and provoked systemic T cell-dependent anti-tumor immunological reactions. These reactions conferred protection against a secondary tumor challenge and destroyed remaining malignant cells in the primary tumor as well as in distant metastases. Such anti-tumor immune responses could be further amplified by the immune adjuvant, CpG. Electrochemical ablation or DaRT together with chemotherapy and immunostimulatory agents can serve as treatment protocols for solid metastatic tumors and can be applied instead of or in combination with surgery.
Collapse
Affiliation(s)
- Yona Keisari
- The Roberts-Guthman Chair in Immunopharmacology, Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, P.O. Box 39040, Tel Aviv, 69978, Israel,
| | | | | | | | | |
Collapse
|
18
|
Heyerdahl H, Røe K, Brevik EM, Dahle J. Modifications in Dynamic Contrast-Enhanced Magnetic Resonance Imaging Parameters After α-Particle-Emitting 227Th-trastuzumab Therapy of HER2-Expressing Ovarian Cancer Xenografts. Int J Radiat Oncol Biol Phys 2013; 87:153-9. [DOI: 10.1016/j.ijrobp.2013.04.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/09/2013] [Accepted: 04/29/2013] [Indexed: 12/28/2022]
|
19
|
Zheng SG, Xu HX, Lu MD, Yue DC, Xie XY, Liu GJ. Radiofrequency ablation before intratumoral injection of (131)I-chTNT improves the tumor-to-normal tissue ratio in solid VX2 tumor. Cancer Biother Radiopharm 2013; 28:725-30. [PMID: 23964639 DOI: 10.1089/cbr.2012.1418] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE This study was aimed to investigate whether the tumor necrosis induced by radiofrequency ablation (RFA) can improve the ratio of tumor-to-normal tissue (T/NT) after intratumoral injection of (131)I-chTNT. MATERIALS AND METHOD Eighteen New Zealand rabbits bearing VX2 tumor on the thigh were randomly divided into two treatment groups (control group: intratumoral injection of (131)I-chTNT alone; RFA group: RFA + intratumoral injection of (131)I-chTNT 3 days after RFA) and each group was further divided into three subgroups I, II, and III (1-2 cm, 2-3 cm, and 3-4 cm in maximum diameter, respectively), by the tumor size. SPECT was performed to evaluate the T/NT on days 1, 8, and 15 after (131)I-chTNT injection. RESULTS After treatment, all rabbits underwent the SPECT whole-body scan and the T/NT was analyzed. The results showed that T/NT in the RFA group (55.45±41.83) was significantly higher compared with the control group (7.23±5.61) (F=18.89, p=0.001). Meanwhile, a linear ascending trend was found for T/NT in the RFA group along with the follow-up time (r=0.47, p=0.01). The tumor size or the dose of (131)I-TNT injection had no significant effect on the variation of T/NT in both groups (p>0.05). CONCLUSION RFA before intratumoral injection of (131)I-chTNT can dramatically improve T/NT, demonstrating the potential application of this combination therapy.
Collapse
Affiliation(s)
- Shu-Guang Zheng
- 1 Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| | | | | | | | | | | |
Collapse
|
20
|
Örbom A, Eriksson SE, Elgström E, Ohlsson T, Nilsson R, Tennvall J, Strand SE. The Intratumoral Distribution of Radiolabeled 177Lu-BR96 Monoclonal Antibodies Changes in Relation to Tumor Histology over Time in a Syngeneic Rat Colon Carcinoma Model. J Nucl Med 2013; 54:1404-10. [DOI: 10.2967/jnumed.112.117028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|