1
|
Folate-based radiotracers for nuclear imaging and radionuclide therapy. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
2
|
Carpanese D, Zorz A, Evangelista L, Salvarese N. Targeting prostate cancer with the anti-PSMA scFvD2B: a theranostic promise for nuclear medicine. Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00337-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
3
|
Farran B, Pavitra E, Kasa P, Peela S, Rama Raju GS, Nagaraju GP. Folate-targeted immunotherapies: Passive and active strategies for cancer. Cytokine Growth Factor Rev 2019; 45:45-52. [PMID: 30770191 DOI: 10.1016/j.cytogfr.2019.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 01/23/2023]
Abstract
The glycoprotein FRα is a membrane-attached transport protein that is shielded from the immune system in healthy cells. However, it is upregulated in various malignancies, involved in cancer development and is also immunogenic. Furthermore, FRα is a tumor-associated antigen endowed with unique properties, thus rendering it a suitable target for immunotherapeutic development in cancer. Various anti- FRα immunotherapeutic strategies are thus currently being developed and clinically assessed for the treatment of various solid tumors. These approaches include passive anti-FRα immunotherapies, such as monoclonal antibodies, or active immunotherapies, such as CART, folate haptens and vaccines. In this review, we will explore the advances in the field of FRα-based immune therapies and discuss both their successes and shortcomings in the clinical setting.
Collapse
Affiliation(s)
- Batoul Farran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Eluri Pavitra
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, 100, Inha-ro, Incheon, 22212, Republic of Korea
| | - Prameswari Kasa
- Dr. LV Prasad Diagnostics and Research Laboratory, Khairtabad, Hyderabad, 500004, India
| | - Sujatha Peela
- Department of Biotechnology, Dr. B.R. Ambedkar University, Srikakulam, Andhra Pradesh, 532410, India
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
4
|
Jindal V, Arora E, Gupta S, Lal A, Masab M, Potdar R. Prospects of chimeric antigen receptor T cell therapy in ovarian cancer. Med Oncol 2018; 35:70. [PMID: 29651744 DOI: 10.1007/s12032-018-1131-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 04/08/2018] [Indexed: 01/12/2023]
Abstract
Despite advances in various chemotherapy regimens, current therapeutic options are limited for ovarian cancer patients. Immunotherapy provides a promising and novel treatment option for ovarian cancer. Recently, chimeric antigen receptor (CAR) T cell therapy has shown promising results in hematological tumors and current research is going on in various solid tumors like ovarian cancer. CAR T cells are genetically engineered T cells with major histocompatibility complex-independent, tumor-specific, immune-mediated cytolytic actions against cancer cells. Initial studies of CAR T cell therapy have shown promising results in ovarian cancer, but there are some obstacles like impaired T cell trafficking, lack of antigenic targets, cytokine release syndrome and most important immunosuppressive tumor microenvironment. Optimization of design, improving tumor microenvironment and combinations with other therapies may help us in improving CAR T cell efficacy. In this review article, we highlight the current knowledge regarding CAR T cell therapy in ovarian cancer. We have discussed basic functioning of CAR T cells, their rationale and clinical outcome in ovarian cancer with limitations.
Collapse
Affiliation(s)
- Vishal Jindal
- Department of Internal Medicine, Saint Vincent Hospital, 123 Summer Street, Worcester, 01608, USA.
| | - Ena Arora
- Department of Internal Medicine, Government Medical College and Hospital, Chandigarh, India
| | - Sorab Gupta
- Department of Hematology and Oncology, Einstein Healthcare Network, Philadelphia, USA
| | - Amos Lal
- Department of Internal Medicine, Saint Vincent Hospital, 123 Summer Street, Worcester, 01608, USA
| | - Muhammad Masab
- Department of Internal Medicine, Einstein Healthcare Network, Philadelphia, USA
| | - Rashmika Potdar
- Department of Hematology and Oncology, Einstein Healthcare Network, Philadelphia, USA
| |
Collapse
|
5
|
Duan S, Guo L, Shi D, Shang M, Meng D, Li J. Development of a novel folate-modified nanobubbles with improved targeting ability to tumor cells. ULTRASONICS SONOCHEMISTRY 2017; 37:235-243. [PMID: 28427629 DOI: 10.1016/j.ultsonch.2017.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 06/07/2023]
Abstract
Conjugation of folate (FOL) to nanobubbles could enhance the selective targeting to tumors expressing high levels of folate receptor (FR). To further improve the selective targeting ability of FOL-modified nanobubbles, a novel FOL-targeted nanobubble ((FOL)2-NB) with increasing FOL content (accomplished by linking two FOL molecules per DSPE-PEG2000 chain) was synthesized, through the methods of mechanical shaking and low-speed centrifugation based on lipid-stabilized perfluoropropane. The bubble size and distribution range were measured by dynamic light scattering (DLS). Enhanced imaging ability was evaluated using a custom-made agarose mold with a clinical US imaging system at mechanical indices of up to 0.12 at a center frequency of 9.0MHz. Targeted ability was also carried out in human breast cancer MCF-7 cells, which over-express the FR, by fluorescence activated cell sorting (FACS) and fluorescence microscopy, respectively. (FOL)2-NB with a particle size of 286.87±22.96nm were successfully prepared, and they exhibited superior contrast imaging effect. FACS and fluorescence microscopy studies showed greater cellular targeting ability in the group of (FOL)2-NB than in their control group of Non-targeted-NB (no FOL targeted nanobubbles) and FOL-NB (one FOL molecule per DSPE-PEG2000 chain). These results suggest that a new type of stronger targeted nanobubble was successfully prepared by increasing the FOL content per DSPE-PEG2000 chain. This novel (FOL)2-NBs are potentially useful for ultrasound molecular imaging and treatment of FR-positive tumors and are worthy for further investigation.
Collapse
Affiliation(s)
- Sujuan Duan
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China.
| |
Collapse
|
6
|
Yung MMH, Ngan HYS, Chan DW. Targeting AMPK signaling in combating ovarian cancers: opportunities and challenges. Acta Biochim Biophys Sin (Shanghai) 2016; 48:301-17. [PMID: 26764240 PMCID: PMC4886241 DOI: 10.1093/abbs/gmv128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/29/2015] [Indexed: 12/25/2022] Open
Abstract
The development and strategic application of effective anticancer therapies have turned out to be one of the most critical approaches of managing human cancers. Nevertheless, drug resistance is the major obstacle for clinical management of these diseases especially ovarian cancer. In the past years, substantial studies have been carried out with the aim of exploring alternative therapeutic approaches to enhance efficacy of current chemotherapeutic regimes and reduce the side effects caused in order to produce significant advantages in overall survival and to improve patients' quality of life. Targeting cancer cell metabolism by the application of AMP-activated protein kinase (AMPK)-activating agents is believed to be one of the most plausible attempts. AMPK activators such as 5-aminoimidazole-4-carboxamide 1-β-d-ribofuranoside, A23187, metformin, and bitter melon extract not only prevent cancer progression and metastasis but can also be applied as a supplement to enhance the efficacy of cisplatin-based chemotherapy in human cancers such as ovarian cancer. However, because of the undesirable outcomes along with the frequent toxic side effects of most pharmaceutical AMPK activators that have been utilized in clinical trials, attentions of current studies have been aimed at the identification of replaceable reagents from nutraceuticals or traditional medicines. However, the underlying molecular mechanisms of many nutraceuticals in anticancer still remain obscure. Therefore, better understanding of the functional characterization and regulatory mechanism of natural AMPK activators would help pharmaceutical development in opening an area to intervene ovarian cancer and other human cancers.
Collapse
Affiliation(s)
- Mingo M H Yung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - David W Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
7
|
Drerup JM, Liu Y, Padron AS, Murthy K, Hurez V, Zhang B, Curiel TJ. Immunotherapy for ovarian cancer. Curr Treat Options Oncol 2015; 16:317. [PMID: 25648541 DOI: 10.1007/s11864-014-0317-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OPINION STATEMENT All work referenced herein relates to treatment of epithelial ovarian carcinomas, as their treatment differs from ovarian germ cell cancers and other rare ovarian cancers, the treatments of which are addressed elsewhere. Fallopian tube cancers and primary peritoneal adenocarcinomatosis are also generally treated as epithelial ovarian cancers. The standard of care initial treatment of advanced stage epithelial ovarian cancer is optimal debulking surgery as feasible plus chemotherapy with a platinum plus a taxane agent. If this front-line approach fails, as it too often the case, several FDA-approved agents are available for salvage therapy. However, because no second-line therapy for advanced-stage epithelial ovarian cancer is typically curative, we prefer referral to clinical trials as logistically feasible, even if it means referring patients outside our system. Immune therapy has a sound theoretical basis for treating carcinomas generally, and for treating ovarian cancer in particular. Advances in understanding the immunopathogenic basis of ovarian cancer, and the immunopathologic basis for prior failures of immunotherapy for it and other carcinomas promises to afford novel treatment approaches with potential for significant efficacy, and reduced toxicities compared with cytotoxic agents. Thus, referral to early phase immunotherapy trials for ovarian cancer patients that fail conventional treatment merits consideration.
Collapse
Affiliation(s)
- Justin M Drerup
- Department of Cellular and Structural Biology, School of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | | | | | | | | | | | | |
Collapse
|
8
|
Quarta A, Bernareggi D, Benigni F, Luison E, Nano G, Nitti S, Cesta MC, Di Ciccio L, Canevari S, Pellegrino T, Figini M. Targeting FR-expressing cells in ovarian cancer with Fab-functionalized nanoparticles: a full study to provide the proof of principle from in vitro to in vivo. NANOSCALE 2015; 7:2336-2351. [PMID: 25504081 DOI: 10.1039/c4nr04426f] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Efficient targeting in tumor therapies is still an open issue: systemic biodistribution and poor specific accumulation of drugs weaken efficacy of treatments. Engineered nanoparticles are expected to bring benefits by allowing specific delivery of drug to the tumor or acting themselves as localized therapeutic agents. In this study we have targeted epithelial ovarian cancer with inorganic nanoparticles conjugated to a human antibody fragment against the folate receptor over-expressed on cancer cells. The conjugation approach is generally applicable. Indeed several types of nanoparticles (either magnetic or fluorescent) were engineered with the fragment, and their biological activity was preserved as demonstrated by biochemical methods in vitro. In vivo studies with mice bearing orthotopic and subcutaneous tumors were performed. Elemental and histological analyses showed that the conjugated magnetic nanoparticles accumulated specifically and were retained at tumor sites longer than the non-conjugated nanoparticles.
Collapse
Affiliation(s)
- Alessandra Quarta
- Nanoscience Institute of CNR, National Nanotechnology Laboratory, via Arnesano, 73100, Lecce, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Müller C, Schibli R. Prospects in folate receptor-targeted radionuclide therapy. Front Oncol 2013; 3:249. [PMID: 24069581 PMCID: PMC3781362 DOI: 10.3389/fonc.2013.00249] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 09/09/2013] [Indexed: 11/18/2022] Open
Abstract
Targeted radionuclide therapy is based on systemic application of particle-emitting radiopharmaceuticals which are directed toward a specific tumor-associated target. Accumulation of the radiopharmaceutical in targeted cancer cells results in high doses of absorbed radiation energy whereas toxicity to non-targeted healthy tissue is limited. This strategy has found widespread application in the palliative treatment of neuroendocrine tumors using somatostatin-based radiopeptides. The folate receptor (FR) has been identified as a target associated with a variety of frequent tumor types (e.g., ovarian, lung, brain, renal, and colorectal cancer). In healthy organs and tissue FR-expression is restricted to only a few sites such as for instance the kidneys. This demonstrates why FR-targeting is an attractive strategy for the development of new therapy concepts. Due to its high FR-binding affinity (KD < 10−9 M) the vitamin folic acid has emerged as an almost ideal targeting agent. Therefore, a variety of folic acid radioconjugates for nuclear imaging have been developed. However, in spite of the large number of cancer patients who could benefit of a folate-based radionuclide therapy, a therapeutic concept with folate radioconjugates has not yet been envisaged for clinical application. The reason is the generally high accumulation of folate radioconjugates in the kidneys where emission of particle-radiation may result in damage to the renal tissue. Therefore, the design of more sophisticated folate radioconjugates providing improved tissue distribution profiles are needed. This review article summarizes recent developments with regard to a therapeutic application of folate radioconjugates. A new construct of a folate radioconjugate and an application protocol which makes use of a pharmacological interaction allowed the first preclinical therapy experiments with radiofolates. These results raise hope for future application of such new concepts also in the clinic.
Collapse
Affiliation(s)
- Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute , Villigen-PSI , Switzerland
| | | |
Collapse
|
10
|
Frigerio B, Fracasso G, Luison E, Cingarlini S, Mortarino M, Coliva A, Seregni E, Bombardieri E, Zuccolotto G, Rosato A, Colombatti M, Canevari S, Figini M. A single-chain fragment against prostate specific membrane antigen as a tool to build theranostic reagents for prostate cancer. Eur J Cancer 2013; 49:2223-32. [PMID: 23433847 DOI: 10.1016/j.ejca.2013.01.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 01/24/2013] [Accepted: 01/26/2013] [Indexed: 10/27/2022]
Abstract
Prostate carcinoma is the most common non-cutaneous cancer in developed countries and represents the second leading cause of death. Early stage androgen dependent prostate carcinoma responds well to conventional therapies, but relatively few treatment options exist for patients with hormone-refractory prostate cancer. One of the most suitable targets for antibody-mediated approaches is prostate specific membrane antigen (PSMA) which is a well known tumour associated antigen. PSMA is a type II integral cell-surface membrane protein that is not secreted, and its expression density and enzymatic activity are increased progressively in prostate cancer compared to normal prostate epithelium, thereby making PSMA an ideal target for monoclonal antibody imaging and therapy. To obtain a small protein that can better penetrate tissue, we have engineered a single-chain variable fragment (scFv) starting from the variable heavy and light domains of the murine anti-PSMA monoclonal antibody D2B. scFvD2B was analysed in vitro for activity, stability, internalisation ability and in vivo for targeting specificity. Maintenance of function and immunoreactivity as well as extremely high radiolabelling efficiency and radiochemical purity were demonstrated by in vitro assays and under different experimental conditions. Despite its monovalent binding, scFvD2B retained a good strength of binding and was able to internalise around 40% of bound antigen. In vivo we showed its ability to specifically target only PSMA expressing prostate cancer xenografts. Due to these advantageous properties, scFvD2B has the potential to become a good theranostic reagent for early detection and therapy of prostate cancers.
Collapse
Affiliation(s)
- B Frigerio
- Molecular Therapies Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - G Fracasso
- Department of Pathology and Diagnostics, Section of Immunology, University of Verona, Verona, Italy
| | - E Luison
- Molecular Therapies Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - S Cingarlini
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata (AOUI), Verona, Italy
| | - M Mortarino
- Molecular Therapies Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - A Coliva
- Department of Diagnostic Imaging and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - E Seregni
- Department of Diagnostic Imaging and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - E Bombardieri
- Department of Diagnostic Imaging and Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - A Rosato
- Istituto Oncologico Veneto IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - M Colombatti
- Department of Pathology and Diagnostics, Section of Immunology, University of Verona, Verona, Italy
| | - S Canevari
- Molecular Therapies Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - M Figini
- Molecular Therapies Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| |
Collapse
|
11
|
Beckford Vera DR, Eigner S, Eigner Henke K, Leyva Montaña R, Melichar F, Beran M. (177)Lu/ (90)Y intermediate-affinity monoclonal antibodies targeting EGFR and HER2/c-neu: preparation and preclinical evaluation. Recent Results Cancer Res 2013; 194:301-317. [PMID: 22918766 DOI: 10.1007/978-3-642-27994-2_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The epidermal growth factor receptor (EGFR) is a rational target of anticancer therapies due to its overexpression in a variety of malignant epithelial tumors. Nevertheless, this antigen is also present in normal tissues. Consequently, monoclonal antibodies which selectively bind to EGFR-overexpressing tumors will be choice drug candidates for development of radioimmunoconjugates (RIC). Nimotuzumab (h-R3) and trastuzumab are monoclonal antibodies (mAbs) which would preferentially target tissues with EGFR and HER2 overexpression, respectively. In this chapter, we describe preparation and evaluation of the targeting properties of RIC formed by (177)Lu/(90)Y and monoclonal antibodies which selectively target EGFR- and HER2/c-neu-overexpressing tissues. mAbs were labeled with n.c.a. (177)Lu/(90)Y using bifunctional chelating agents. RIC binding properties and toxicity were evaluated in vitro using cell lines with varying antigen expression. In vivo tumor targeting properties of RIC were evaluated in mice bearing colorectal (SNU-C2B) and A431 tumor xenografts. RICs were prepared with specific activities up to 2 GBq/mg without significant loss in biological activity. (90)Y-h-R3/trastuzumab increased cell growth inhibition compared with unmodified mAbs or (90)YCl(3) alone in cell lines with overexpression of the target antigen. (177)Lu-h-R3 showed significantly higher uptake in A431 (22.8 ± 3.1% ID/g) than in SNU-C2B (8.8 ± 4.1% ID/g) xenografts at 72 h post injection, indicating strong association between tumor uptake and EGFR expression levels.
Collapse
Affiliation(s)
- Denis R Beckford Vera
- Department of Radiopharmaceuticals, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
12
|
Karagiannis SN, Josephs DH, Karagiannis P, Gilbert AE, Saul L, Rudman SM, Dodev T, Koers A, Blower PJ, Corrigan C, Beavil AJ, Spicer JF, Nestle FO, Gould HJ. Recombinant IgE antibodies for passive immunotherapy of solid tumours: from concept towards clinical application. Cancer Immunol Immunother 2012; 61:1547-64. [PMID: 22139135 PMCID: PMC11028906 DOI: 10.1007/s00262-011-1162-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 11/11/2011] [Indexed: 01/18/2023]
Abstract
Therapeutic antibodies have revolutionised treatment of some cancers and improved prognosis for many patients. Over half of those available are approved for haematological malignancies, but efficacious antibodies for solid tumours are still urgently needed. Clinically available antibodies belong to the IgG class, the most prevalent antibody class in human blood, while other classes have not been extensively considered. We hypothesised that the unique properties of IgE, a class of tissue-resident antibodies commonly associated with allergies, which can trigger powerful immune responses through strong affinity for their particular receptors on effector cells, could be employed for passive immunotherapy of solid tumours such as ovarian and breast carcinomas. Our laboratory has examined this concept by evaluating two chimaeric antibodies of the same specificity (MOv18) but different isotype, an IgG1 and an IgE against the tumour antigen folate receptor α (FRα). The latter demonstrates the potency of IgE to mount superior immune responses against tumours in disease-relevant models. We identified Fcε receptor-expressing cells, monocytes/macrophages and eosinophils, activated by MOv18 IgE to kill tumour cells by mechanisms such as ADCC and ADCP. We also applied this notion to a marketed therapeutic, the humanised IgG1 antibody trastuzumab and engineered an IgE counterpart, which retained the functions of trastuzumab in restricting proliferation of HER2/neu-expressing tumour cells but also activated effector cells to kill tumour cells by different mechanisms. On-going efficacy, safety evaluations and future first-in-man clinical studies of IgE therapeutics constitute key metrics for this concept, providing new scope for antibody immunotherapies for solid tumours.
Collapse
Affiliation(s)
- Sophia N Karagiannis
- NIHR Biomedical Research Centre at Guy's and St Thomas's Hospitals and King's College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Beckford Vera DR, Eigner S, Henke KE, Lebeda O, Melichar F, Beran M. Preparation and preclinical evaluation of 177Lu-nimotuzumab targeting epidermal growth factor receptor overexpressing tumors. Nucl Med Biol 2012; 39:3-13. [DOI: 10.1016/j.nucmedbio.2011.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/30/2011] [Accepted: 07/03/2011] [Indexed: 10/17/2022]
|
14
|
Wittrup KD, Thurber GM, Schmidt MM, Rhoden JJ. Practical theoretic guidance for the design of tumor-targeting agents. Methods Enzymol 2012; 503:255-68. [PMID: 22230572 DOI: 10.1016/b978-0-12-396962-0.00010-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Theoretical analyses of targeting agent pharmacokinetics provides specific guidance with respect to desirable design objectives such as agent size, affinity, and target antigen. These analyses suggest that IgG-sized macromolecular constructs exhibit the most favorable balance between systemic clearance and vascular extravasation, resulting in maximal tumor uptake. Quantitative predictions of the effects of dose and binding affinity on tumor uptake and penetration are also provided. The single bolus dose required for saturation of xenografted tumors in mice can be predicted from knowledge of antigen expression level and metabolic half-life. The role of high binding affinity in tumor uptake can be summarized as: essential for small peptides, less important for antibodies, and negligible for nanoparticles.
Collapse
Affiliation(s)
- K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
15
|
Zacchetti A, Martin F, Luison E, Coliva A, Bombardieri E, Allegretti M, Figini M, Canevari S. Antitumor effects of a human dimeric antibody fragment 131I-AFRA-DFM5.3 in a mouse model for ovarian cancer. J Nucl Med 2011; 52:1938-46. [PMID: 22068897 DOI: 10.2967/jnumed.110.086819] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED AFRA-DMF5.3 is a human antibody fragment that, as a dimer, specifically binds to the α-folate receptor (FR) on ovary cancer cells. Pharmacokinetic and biodistribution parameters of (131)I-AFRA-DFM5.3 after intravenous administration in animal models support its potential therapeutic use. We evaluated its preclinical specificity and therapeutic efficacy in tumor models. METHODS A negative control, AFRA-DFM6.1, was obtained by protein engineering. The activity and specificity of (131)I-AFRA-DFMs were evaluated by systemic administration (intravenous) in subcutaneous tumor xenograft-bearing nude mice. Pharmacokinetics, biodistribution, and efficacy were assessed by intraperitoneal administration of (131)I-AFRA-DFM5.3 in nude mice bearing 2 different intraperitoneal ovarian carcinoma xenografts. Treatments were tested at different doses and as single or double administrations 1 wk apart. RESULTS In subcutaneous models, (131)I-AFRA-DFM5.3, but not the negative control, was found to reside on FR-positive tumor masses and significantly reduced tumor growth. In intraperitoneal models, early accumulation on free-floating clumps of ovarian cancer cells and solid peritoneal masses was evident after 1 h, and tumor uptake was stable for up to 3 h. The high tumor uptake determined the efficacy of (131)I-AFRA-DFM5.3. The best antitumor activity, with more than 50% of treated animals cured, was achieved with 2 locoregional treatments of intraperitoneally growing tumors on days 2 and 9. CONCLUSION These results suggest that radioimmunotherapy with (131)I-AFRA-DFM5.3 is feasible and leads to significantly prolonged survival. These preclinical data provide the basis for the rationale design of therapeutic treatments of ovarian cancer patients with a radiolabeled anti-FR antibody fragment.
Collapse
Affiliation(s)
- Alberto Zacchetti
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Thibodeaux SR, Curiel TJ. Immune Therapy for Ovarian Cancer: Promise and Pitfalls. Int Rev Immunol 2011; 30:102-19. [DOI: 10.3109/08830185.2011.567361] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
17
|
Mantia-Smaldone GM, Edwards RP, Vlad AM. Targeted treatment of recurrent platinum-resistant ovarian cancer: current and emerging therapies. Cancer Manag Res 2010; 3:25-38. [PMID: 21734812 PMCID: PMC3130354 DOI: 10.2147/cmr.s8759] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
With advances in surgical techniques and chemotherapeutic agents, mortality rates from epithelial ovarian cancer (EOC) have slightly decreased over the last 30 years. However, EOC still ranks as the most deadly gynecologic cancer with an overall 5-year survival rate of 45%. Prognosis is especially disappointing for women with platinum-resistant disease, where 80% of patients will fail to respond to available therapies. Emerging treatment strategies have sub-sequently focused on targets which are integral to tumor growth and metastasis. In this review, we will focus on those innovative agents currently under investigation in clinical trials.
Collapse
Affiliation(s)
- Gina M Mantia-Smaldone
- Division of Gynecologic Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
18
|
Liu B, Nash J, Runowicz C, Swede H, Stevens R, Li Z. Ovarian cancer immunotherapy: opportunities, progresses and challenges. J Hematol Oncol 2010; 3:7. [PMID: 20146807 PMCID: PMC2831814 DOI: 10.1186/1756-8722-3-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 02/10/2010] [Indexed: 12/20/2022] Open
Abstract
Due to the low survival rates from invasive ovarian cancer, new effective treatment modalities are urgently needed. Compelling evidence indicates that the immune response against ovarian cancer may play an important role in controlling this disease. We herein summarize multiple immune-based strategies that have been proposed and tested for potential therapeutic benefit against advanced stage ovarian cancer. We will examine the evidence for the premise that an effective therapeutic vaccine against ovarian cancer is useful not only for inducing remission of the disease but also for preventing disease relapse. We will also highlight the questions and challenges in the development of ovarian cancer vaccines, and critically discuss the limitations of some of the existing immunotherapeutic strategies. Finally, we will summarize our own experience on the use of patient-specific tumor-derived heat shock protein-peptide complex for the treatment of advanced ovarian cancer.
Collapse
Affiliation(s)
- Bei Liu
- Department of Immunology, University of Connecticut School of Medicine, Farmington, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Lu Y, Klein PJ, Westrick E, Xu LC, Santhapuram HKR, Bloomfield A, Howard SJ, Vlahov IR, Ellis PR, Low PS, Leamon CP. Strategy to prevent drug-related hypersensitivity in folate-targeted hapten immunotherapy of cancer. AAPS JOURNAL 2009; 11:628-38. [PMID: 19728104 DOI: 10.1208/s12248-009-9139-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 08/05/2009] [Indexed: 11/30/2022]
Abstract
Cancer vaccine/immunotherapy rarely involves systemic administration of an immunogenic compound to an actively immunized host. We have developed such a strategy that utilizes folate to deliver antigenic haptens [e.g., fluorescein (FITC) and dinitrophenyl] to folate receptor-positive tumors in a hapten-pre-vaccinated host. Here, we investigated the safety of this novel approach and developed strategies to prevent drug-related hypersensitivity. Using FITC as the model hapten, we identified a potential source of allergic species in folate-FITC preparations by LC-MS/MS. In mice and guinea pigs, we tested the significance of this impurity by passive cutaneous anaphylaxis and active systemic anaphylaxis assays. We studied the effect of immunogen (e.g., KLH-FITC) dose and derived a desensitization regimen that was further evaluated in a murine tumor model. Administration of folate-FITC with low multi-haptenated contaminants (e.g. bis-FITC) resulted in hypersensitivity in underimmunized animals. However, this drug-related hypersensitivity may be independently prevented by (1) increasing the immunogen dose and/or (2) desensitizing animals with folate-FITC during vaccination. In addition, such manipulation in vivo did not appear to negatively alter the effectiveness of immunotherapy. This study provided confidence on the safety of folate-hapten-targeted cancer immunotherapy in an actively immunized host.
Collapse
|
20
|
Zacchetti A, Coliva A, Luison E, Seregni E, Bombardieri E, Giussani A, Figini M, Canevari S. 177Lu- labeled MOv18 as compared to 131I- or 90Y-labeled MOv18 has the better therapeutic effect in eradication of alpha folate receptor-expressing tumor xenografts. Nucl Med Biol 2009; 36:759-70. [DOI: 10.1016/j.nucmedbio.2009.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 05/13/2009] [Accepted: 05/15/2009] [Indexed: 01/29/2023]
|
21
|
Figini M, Martin F, Ferri R, Luison E, Ripamonti E, Zacchetti A, Mortarino M, Di Cioccio V, Maurizi G, Allegretti M, Canevari S. Conversion of murine antibodies to human antibodies and their optimization for ovarian cancer therapy targeted to the folate receptor. Cancer Immunol Immunother 2009; 58:531-46. [PMID: 18704410 PMCID: PMC11030786 DOI: 10.1007/s00262-008-0575-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 07/28/2008] [Indexed: 12/16/2022]
Abstract
We previously developed murine and chimeric antibodies against a specific epithelial ovarian carcinoma (EOC) marker, named folate receptor (FR), and promising results were obtained in phase II trials. More recently, we successfully generated a completely human Fab fragment, C4, by conversion of one of the murine anti-FR antibodies to human antibody using phage display and guided selection. However, subsequent efforts to obtain C4 in a dimer format, which seems especially desirable for EOC locoregional treatment, resulted in a highly heterogeneous product upon natural dimerization and in a very poor production yield upon chemical dimerization by a non-hydrolyzable linker to a di-Fab-maleimide (DFM). We therefore designed, constructed and characterized a large Fab dual combinatorial human antibody phage display library obtained from EOC patients and potentially biased toward an anti-tumor response in an effort to obtain new anti-FR human antibodies suitable for therapy. Using this library and guiding the selection on FR-expressing cells with murine/human antibody chains, we generated four new human anti-FR antibody (AFRA) Fab fragments, one of which was genetically and chemically manipulated to obtain a chemical dimer, designated AFRA-DFM5.3, with high yield production and the capability for purification scaled-up to clinical grade. Overall affinity of AFRA-DFM5.3 was in the 2-digit nanomolar range, and immunohistochemistry indicated that the reagent recognized the FR expressed on EOC samples. (131)I-AFRA-DFM5.3 showed high immunoreactivity, in vitro stability and integrity, and specifically accumulated only in FR-expressing tumors in subcutaneous preclinical in vivo models. Overall, our studies demonstrate the successful conversion of murine to completely human anti-FR antibodies through the combined use of antibody phage display libraries biased toward an anti-tumor response, guided selection and chain shuffling, and point to the suitability of AFRA5.3 for future clinical application in ovarian cancer.
Collapse
Affiliation(s)
- Mariangela Figini
- Unit of Molecular Therapies, Department of Experimental Oncology and Laboratories, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zavaleta CL, Goins BA, Bao A, McManus LM, McMahan CA, Phillips WT. Imaging of 186Re-liposome therapy in ovarian cancer xenograft model of peritoneal carcinomatosis. J Drug Target 2009; 16:626-37. [PMID: 18686134 DOI: 10.1080/10611860802230372] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This study determined the biodistribution of rhenium-186 ((186)Re) encapsulated in biotin-liposomes containing patent blue dye, injected intraperitoneally (IP) with avidin in an OVCAR-3 ovarian cancer xenograft model and evaluated tumor response of this therapy with fluorine-18-fluorodeoxyglucose ((18)F-FDG) microPET imaging. Treated rats (n = 8) received an IP injection of (186)Re-blue-biotin-liposomes (1000 MBq/kg) 30 min before an IP injection of avidin (5 mg), whereas control rats (n = 4) received a sham IP injection of saline. Scintigraphic images showed that (186)Re-blue-biotin liposomes/avidin were retained in the peritoneal cavity with 18% of the original activity remaining after 5 days. From 1 to 4 weeks post-treatment, peritoneal (18)F-FDG standard uptake values decreased 30% in treatment group, yet increased 44% in control group. Total number of cells in ascites was significantly higher in control versus treatment group. Omental fat in control rats had numerous tumor cells compared with treated rats. Results show the potential for (186)Re-blue-biotin-liposome/avidin system in treating advanced ovarian cancer involving peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Cristina L Zavaleta
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Smith-Jones PM, Pandit-Taskar N, Cao W, O'Donoghue J, Philips MD, Carrasquillo J, Konner JA, Old LJ, Larson SM. Preclinical radioimmunotargeting of folate receptor alpha using the monoclonal antibody conjugate DOTA-MORAb-003. Nucl Med Biol 2008; 35:343-51. [PMID: 18355690 DOI: 10.1016/j.nucmedbio.2007.12.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Revised: 12/11/2007] [Accepted: 12/23/2007] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The in vitro and in vivo behavior of the radiolabeled monoclonal antibody MORAb-003 was investigated as a prelude to a clinical trial. METHODS The cellular retention of 111In- and 131I-labeled MORAb-003 was investigated using IGROV1 and SW620 cells. Biodistribution studies in tumor-bearing mice were performed with the more favorable agent. RESULTS Five 1,4,7,10-tetraazacyclododecane-N,N',N",N'"-tetraacetic acid (DOTA) molecules were conjugated to MORAb-003 with no apparent loss of immunoreactivity. Radiolabeled MORAb-003 had a high affinity for the folate receptor alpha (FRA) expressed by both IGROV1 and SW620 cells and was found to bind to around 8 x 10(5) and 7 x 10(5) sites/cell, respectively. Both cancer cell lines were found to internalize both 131I- and 111In-labeled MORAb-003, but 111In was retained and 131I was released as iodide. In athymic mice, 111In-DOTA-MORAb-003 was cleared from the blood with a single exponential biological clearance rate of 110 h. The uptake in SW620 tumors was 32+/-5%ID/g after 4 days. The clearance rate of activity from normal organs such as liver, kidney and spleen was similar to the blood clearance and was 5.36%ID/g, 4.03%ID/g and 4.36%ID/g at 1 day postinjection and 2.14%ID/g, 1.65%ID/g and 3.74%ID/g after 8 days, respectively. In a pilot clinical study, the biodistribution and tumor targeting of 111In-MORAb-003 was assessed in three patients undergoing treatment with cold MORAb-003. CONCLUSION MORAb-003 is an attractive antibody for radioimmunoscintigraphy and possibly radioimmunotherapy of FRA-expressing cancers in addition to its potential direct therapeutic effects.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Biological Transport, Active
- Carrier Proteins/analysis
- Carrier Proteins/chemistry
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Drug Evaluation, Preclinical
- Female
- Folate Receptors, GPI-Anchored
- Heterocyclic Compounds, 1-Ring/pharmacokinetics
- Humans
- Immunoconjugates/pharmacokinetics
- Indium Radioisotopes/pharmacokinetics
- Iodine Radioisotopes/pharmacokinetics
- Male
- Metabolic Clearance Rate
- Mice
- Mice, Nude
- Neoplasm Recurrence, Local/diagnostic imaging
- Neoplasm Recurrence, Local/drug therapy
- Neoplasms, Glandular and Epithelial/diagnostic imaging
- Neoplasms, Glandular and Epithelial/drug therapy
- Ovarian Neoplasms/diagnostic imaging
- Ovarian Neoplasms/drug therapy
- Pilot Projects
- Radiography
- Radioimmunodetection/methods
- Radiopharmaceuticals/pharmacokinetics
- Receptors, Cell Surface/analysis
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/metabolism
- Tissue Distribution
Collapse
Affiliation(s)
- Peter M Smith-Jones
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, Box 77, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Boswell CA, Brechbiel MW. Development of radioimmunotherapeutic and diagnostic antibodies: an inside-out view. Nucl Med Biol 2007; 34:757-78. [PMID: 17921028 PMCID: PMC2212602 DOI: 10.1016/j.nucmedbio.2007.04.001] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 03/29/2007] [Accepted: 04/04/2007] [Indexed: 11/25/2022]
Abstract
Only a handful of radiolabeled antibodies (Abs) have gained US Food and Drug Administration (FDA) approval for use in clinical oncology, including four immunodiagnostic agents and two targeted radioimmunotherapeutic agents. Despite the advent of nonimmunogenic Abs and the availability of a diverse library of radionuclides, progress beyond early Phase II radioimmunotherapy (RIT) studies in solid tumors has been marginal. Furthermore, [18F]fluorodeoxyglucose continues to dominate the molecular imaging domain, underscored by a decade-long absence of any newly approved Ab-based imaging agent (none since 1996). Why has the development of clinically successful Abs for RIT been limited to lymphoma? What obstacles must be overcome to allow the FDA approval of immuno-positron emission tomography (immuno-PET) imaging agents? How can we address the unique challenges that have thus far prevented the introduction of Ab-based imaging agents and therapeutics for solid tumors? Many poor decisions have been made regarding radiolabeled Abs, but useful insight can be gained from these mistakes. The following review addresses the physical, chemical, biological, clinical, regulatory and financial limitations that impede the progress of this increasingly important class of drugs.
Collapse
Affiliation(s)
- C. Andrew Boswell
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, Building 10 Center Drive, Bethesda, Maryland, 20892-1088
| | - Martin W. Brechbiel
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, Building 10 Center Drive, Bethesda, Maryland, 20892-1088
| |
Collapse
|
25
|
Gunn AJ, Brechbiel MW, Choyke PL. The emerging role of molecular imaging and targeted therapeutics in peritoneal carcinomatosis. Expert Opin Drug Deliv 2007; 4:389-402. [PMID: 17683252 DOI: 10.1517/17425247.4.4.389] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Peritoneal carcinomatosis is a common and often fatal late-stage complication of many gastrointestinal and gynecologic malignancies. This review discusses the ongoing evolution of diagnostic and treatment strategies for peritoneal carcinomatosis and the role that molecular imaging and radioimmunotherapy may play in improving patient survival. An overview of recent developments in targeted imaging and therapeutics for peritoneal carcinomatosis, as well as the authors' opinions as to future developments in this field is also provided.
Collapse
Affiliation(s)
- Andrew J Gunn
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD 20815-6789, USA
| | | | | |
Collapse
|
26
|
Ciavardelli D, D'Anniballe G, Nano G, Martin F, Federici G, Sacchetta P, Di Ilio C, Urbani A. An inductively coupled plasma mass spectrometry method for the quantification of yttrium-antibody based drugs using stable isotope tracing. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2007; 21:2343-50. [PMID: 17590870 DOI: 10.1002/rcm.3094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Targeted radioimmunotherapy has been recently clinically validated and approved for the treatment of cancer by the US Food and Drug Administration. This therapeutic approach employs monoclonal antibodies directed to cancer-related, cell-surface antigens coupled to beta-emitting nuclides. 90Y is one of the most useful radioisotopes in the development of antibody based radioimmunotherapy and evaluation of the pharmacokinetic profile for 90Y-radiopharmaceuticals is usually performed by radiochemical methods. In this work we have developed an alternative radioactive-free approach to evaluate pharmacokinetic profiles based on the inductively coupled plasma mass spectrometric (ICP-MS) quantification of 89Y. A highly sensitive and rapid method for the determination of yttrium in urine is described and applied to evaluate the urinary clearance of antibody-based drugs labeled with the stable isotope of yttrium, 89Y. This approach overcomes some important limitations for pre-clinical radioanalytical methods such as radiation hazards and radioactive waste disposal. Method development was performed by determining detection and quantification limits, and precision as repeatability and trueness. These performance parameters fulfilled the acceptance criteria for bioanalytical methods.
Collapse
|
27
|
Lucignani G. The immune system and cancer: the evolving role of molecular imaging and molecular targeted therapy. Eur J Nucl Med Mol Imaging 2006; 33:503-5. [PMID: 16596377 DOI: 10.1007/s00259-006-0080-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Giovanni Lucignani
- Institute of Radiological Sciences, University of Milan and Unit of Molecular Imaging, Division of Radiation Therapy, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy.
| |
Collapse
|
28
|
Kaden TA. Labelling monoclonal antibodies with macrocyclic radiometal complexes. A challenge for coordination chemists. Dalton Trans 2006:3617-23. [PMID: 16865171 DOI: 10.1039/b606410h] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The technique consisting of labelling antibodies with radionuclides is a promising approach for the diagnosis and therapy of human cancers. In this paper the chemical aspects of radiolabelling with macrocyclic metal complexes are dealt with. The choice of the radioisotope, its coordination chemistry, the design of the bifunctional macrocyclic ligand, and its attachment to the monoclonal antibody are discussed. Finally several newer ideas for a better application are presented.
Collapse
Affiliation(s)
- Thomas A Kaden
- Department of Chemistry, Spitalstrasse 51, 4056 Basel, Switzerland. th.kaden@.unibas.ch
| |
Collapse
|