201
|
Ruggiero A, Holland JP, Lewis JS, Grimm J. Cerenkov luminescence imaging of medical isotopes. J Nucl Med 2010; 51:1123-30. [PMID: 20554722 DOI: 10.2967/jnumed.110.076521] [Citation(s) in RCA: 200] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED The development of novel multimodality imaging agents and techniques represents the current frontier of research in the field of medical imaging science. However, the combination of nuclear tomography with optical techniques has yet to be established. Here, we report the use of the inherent optical emissions from the decay of radiopharmaceuticals for Cerenkov luminescence imaging (CLI) of tumors in vivo and correlate the results with those obtained from concordant immuno-PET studies. METHODS In vitro phantom studies were used to validate the visible light emission observed from a range of radionuclides including the positron emitters (18)F, (64)Cu, (89)Zr, and (124)I; beta-emitter (131)I; and alpha-particle emitter (225)Ac for potential use in CLI. The novel radiolabeled monoclonal antibody (89)Zr-desferrioxamine B [DFO]-J591 for immuno-PET of prostate-specific membrane antigen (PSMA) expression was used to coregister and correlate the CLI signal observed with the immuno-PET images and biodistribution studies. RESULTS Phantom studies confirmed that Cerenkov radiation can be observed from a range of positron-, beta-, and alpha-emitting radionuclides using standard optical imaging devices. The change in light emission intensity versus time was concordant with radionuclide decay and was also found to correlate linearly with both the activity concentration and the measured PET signal (percentage injected dose per gram). In vivo studies conducted in male severe combined immune deficient mice bearing PSMA-positive, subcutaneous LNCaP tumors demonstrated that tumor-specific uptake of (89)Zr-DFO-J591 could be visualized by both immuno-PET and CLI. Optical and immuno-PET signal intensities were found to increase over time from 24 to 96 h, and biodistribution studies were found to correlate well with both imaging modalities. CONCLUSION These studies represent the first, to our knowledge, quantitative assessment of CLI for measuring radiotracer uptake in vivo. Many radionuclides common to both nuclear tomographic imaging and radiotherapy have the potential to be used in CLI. The value of CLI lies in its ability to image radionuclides that do not emit either positrons or gamma-rays and are, thus, unsuitable for use with current nuclear imaging modalities. Optical imaging of Cerenkov radiation emission shows excellent promise as a potential new imaging modality for the rapid, high-throughput screening of radiopharmaceuticals.
Collapse
Affiliation(s)
- Alessandro Ruggiero
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | |
Collapse
|
202
|
A generator system for production of medical alpha-radionuclides Ac-225 and Bi-213. J Radioanal Nucl Chem 2010. [DOI: 10.1007/s10967-010-0593-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
203
|
Scheinberg DA, Villa CH, Escorcia FE, McDevitt MR. Conscripts of the infinite armada: systemic cancer therapy using nanomaterials. Nat Rev Clin Oncol 2010; 7:266-76. [PMID: 20351700 PMCID: PMC4411965 DOI: 10.1038/nrclinonc.2010.38] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The field of clinical nanomaterials is enlarging steadily, with more than a billion US dollars of funding allocated to research by US government agencies in the past decade. The first generation of anti-cancer agents using novel nanomaterials has successfully entered widespread use. Newer nanomaterials are garnering increasing interest as potential multifunctional therapeutic agents; these drugs are conferred novel properties, by virtue of their size and shape. The new features of these agents could potentially allow increased cancer selectivity, changes in pharmacokinetics, amplification of cytotoxic effects, and simultaneous imaging capabilities. After attachment to cancer target reactive-ligands, which interact with cell-surface antigens or receptors, these new constructs can deliver cytolytic and imaging payloads. The molecules also introduce new challenges for drug development. While nanoscale molecules are of a similar size to proteins, the paradigms for how cells, tissues and organs of the body react to the non-biological materials are not well understood, because most cellular and metabolic processes have evolved to deal with globular, enzyme degradable molecules. We discuss examples of different materials to illustrate interesting principles for development and future applications of these nanomaterial medicines with emphasis on the possible pharmacologic and safety hurdles for accomplishing therapeutic goals.
Collapse
Affiliation(s)
- David A Scheinberg
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
204
|
Johnson S, Burke S, Huang L, Gorlatov S, Li H, Wang W, Zhang W, Tuaillon N, Rainey J, Barat B, Yang Y, Jin L, Ciccarone V, Moore PA, Koenig S, Bonvini E. Effector cell recruitment with novel Fv-based dual-affinity re-targeting protein leads to potent tumor cytolysis and in vivo B-cell depletion. J Mol Biol 2010; 399:436-49. [PMID: 20382161 DOI: 10.1016/j.jmb.2010.04.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 03/29/2010] [Accepted: 04/01/2010] [Indexed: 11/16/2022]
Abstract
Bispecific antibodies capable of redirecting the lytic potential of immune effector cells to kill tumor targets have long been recognized as a potentially potent biological therapeutic intervention. Unfortunately, efforts to produce such molecules have been limited owing to inefficient production and poor stability properties. Here, we describe a novel Fv-derived strategy based on a covalently linked bispecific diabody structure that we term dual-affinity re-targeting (DART). As a model system, we linked an Fv specific for human CD16 (FcgammaRIII) on effector cells to an Fv specific for mouse or human CD32B (FcgammaRIIB), a normal B-cell and tumor target antigen. DART proteins were produced at high levels in mammalian cells, retained the binding activity of the respective parental Fv domains as well as bispecific binding, and showed extended storage and serum stability. Functionally, the DART molecules demonstrated extremely potent, dose-dependent cytotoxicity in retargeting human PBMC against B-lymphoma cell lines as well as in mediating autologous B-cell depletion in culture. In vivo studies in mice demonstrated effective B-cell depletion that was dependent on the transgenic expression of both CD16A on the effector cells and CD32B on the B-cell targets. Furthermore, DART proteins showed potent in vivo protective activity in a human Burkitt's lymphoma cell xenograft model. Thus, DART represents a biologically potent format that provides a versatile platform for generating bispecific antibody fragments for redirected killing and, with the selection of appropriate binding partners, applications outside of tumor cell cytotoxicity.
Collapse
Affiliation(s)
- Syd Johnson
- MacroGenics, Inc., 1500 East Gude Drive, Rockville, MD 20850, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
205
|
Sgouros G, Roeske JC, McDevitt MR, Palm S, Allen BJ, Fisher DR, Brill AB, Song H, Howell RW, Akabani G, Bolch WE, Brill AB, Fisher DR, Howell RW, Meredith RF, Sgouros G, Wessels BW, Zanzonico PB. MIRD Pamphlet No. 22 (abridged): radiobiology and dosimetry of alpha-particle emitters for targeted radionuclide therapy. J Nucl Med 2010; 51:311-28. [PMID: 20080889 PMCID: PMC5680544 DOI: 10.2967/jnumed.108.058651] [Citation(s) in RCA: 322] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The potential of alpha-particle emitters to treat cancer has been recognized since the early 1900s. Advances in the targeted delivery of radionuclides and radionuclide conjugation chemistry, and the increased availability of alpha-emitters appropriate for clinical use, have recently led to patient trials of radiopharmaceuticals labeled with alpha-particle emitters. Although alpha-emitters have been studied for many decades, their current use in humans for targeted therapy is an important milestone. The objective of this work is to review those aspects of the field that are pertinent to targeted alpha-particle emitter therapy and to provide guidance and recommendations for human alpha-particle emitter dosimetry.
Collapse
Affiliation(s)
- George Sgouros
- Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21231, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Affiliation(s)
- Jason P. Holland
- Radiochemistry Service, Department of Radiology, and Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Matthew J. Williamson
- Radiochemistry Service, Department of Radiology, and Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jason S. Lewis
- Radiochemistry Service, Department of Radiology, and Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
207
|
Song H, Hobbs RF, Vajravelu R, Huso DL, Esaias C, Apostolidis C, Morgenstern A, Sgouros G. Radioimmunotherapy of breast cancer metastases with alpha-particle emitter 225Ac: comparing efficacy with 213Bi and 90Y. Cancer Res 2009; 69:8941-8. [PMID: 19920193 PMCID: PMC2789180 DOI: 10.1158/0008-5472.can-09-1828] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
alpha-Particles are suitable to treat cancer micrometastases because of their short range and very high linear energy transfer. alpha-Particle emitter (213)Bi-based radioimmunotherapy has shown efficacy in a variety of metastatic animal cancer models, such as breast, ovarian, and prostate cancers. Its clinical implementation, however, is challenging due to the limited supply of (225)Ac, high technical requirement to prepare radioimmunoconjugate with very short half-life (T(1/2) = 45.6 min) on site, and prohibitive cost. In this study, we investigated the efficacy of the alpha-particle emitter (225)Ac, parent of (213)Bi, in a mouse model of breast cancer metastases. A single administration of (225)Ac (400 nCi)-labeled anti-rat HER-2/neu monoclonal antibody (7.16.4) completely eradicated breast cancer lung micrometastases in approximately 67% of HER-2/neu transgenic mice and led to long-term survival of these mice for up to 1 year. Treatment with (225)Ac-7.16.4 is significantly more effective than (213)Bi-7.16.4 (120 microCi; median survival, 61 days; P = 0.001) and (90)Y-7.16.4 (120 microCi; median survival, 50 days; P < 0.001) as well as untreated control (median survival, 41 days; P < 0.0001). Dosimetric analysis showed that (225)Ac-treated metastases received a total dose of 9.6 Gy, significantly higher than 2.0 Gy from (213)Bi and 2.4 Gy from (90)Y. Biodistribution studies revealed that (225)Ac daughters, (221)Fr and (213)Bi, accumulated in kidneys and probably contributed to the long-term renal toxicity observed in surviving mice. These data suggest (225)Ac-labeled anti-HER-2/neu monoclonal antibody could significantly prolong survival in HER-2/neu-positive metastatic breast cancer patients.
Collapse
Affiliation(s)
- Hong Song
- Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21231
| | - Robert F. Hobbs
- Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21231
| | - Ravy Vajravelu
- Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21231
| | - David L. Huso
- Department of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21231
| | - Caroline Esaias
- Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21231
| | | | | | - George Sgouros
- Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21231
| |
Collapse
|
208
|
Akiyama K, Haba H, Sueki K, Tsukada K, Asai M, Toyoshima A, Nagame Y, Katada M. 225Ac Metallofullerene: Toward225Ac Nanogenerator in Fullerene. CHEM LETT 2009. [DOI: 10.1246/cl.2009.978] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
209
|
Zhang M, Yao Z, Garmestani K, Yu S, Goldman CK, Paik CH, Brechbiel MW, Carrasquillo JA, Waldmann TA. Preclinical evaluation of an anti-CD25 monoclonal antibody, 7G7/B6, armed with the beta-emitter, yttrium-90, as a radioimmunotherapeutic agent for treating lymphoma. Cancer Biother Radiopharm 2009; 24:303-9. [PMID: 19538052 DOI: 10.1089/cbr.2008.0577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Radioimmunotherapy of cancer with radiolabeled antibodies has shown promise. We evaluated an anti-CD25 monoclonal Antibody, 7G7/B6, armed with (90)Y as a potential radioimmunotherapeutic agent for CD25-expressing lymphomas. MATERIALS AND METHODS The lymphoma model was established by subcutaneous injection of 1 x 10(7) SUDHL-1 cells into nude mice. The biodistribution of (111)In-7G7/B6 and therapeutic studies with (90)Y-7G7/B6 were performed in the tumor-bearing mice. RESULTS Therapy using (90)Y-7G7/B6 prolonged survival of the SUDHL-1 lymphoma-bearing mice significantly, as compared with either untreated mice or the mice treated with (90)Y-11F11, a radiolabeled isotype-matched control antibody (p < 0.001). All of the mice in the control and the (90)Y-11F11 treatment groups died by days 18 and 24, respectively. In contrast, 30% of the mice in the low-dose group (75 microCi of (90)Y-7G7/B6/mouse) and 75% in the high-dose group (150 microCi of (90)Y-7G7/B6/mouse) became tumor free and remained healthy for greater than 6 months. CONCLUSIONS Our findings suggested that (90)Y-7G7/B6 is a potentially useful radioimmunotherapeutic agent for the treatment of patients with CD25-expressing lymphomas.
Collapse
Affiliation(s)
- Meili Zhang
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1374, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Wlodzimirska B, Bartoś B, Bilewicz A. Preparation of 225Ac and 228Ac generators using a cryptomelane manganese dioxide sorbent. RADIOCHIM ACTA 2009. [DOI: 10.1524/ract.91.9.553.19996] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Summary
The distribution coefficients of Ra2+ and Ac3+ on cryptomelane MnO2 from acidic aqueous solutions were determined in order to find the best conditions for separation of both cations. Very high affinity of cryptomelane MnO2 for Ra2+ ions make possible to separate 225Ac from 225Ra, and 228Ac from 228Ra in single-step and rapid procedure. The obtained results enable to design simple and effective generators of 225Ac and 228Ac from their mother radium isotopes.
Collapse
|
211
|
Lv Y, Cheung NKV, Fu BM. A pharmacokinetic model for radioimmunotherapy delivered through cerebrospinal fluid for the treatment of leptomeningeal metastases. J Nucl Med 2009; 50:1324-31. [PMID: 19617331 DOI: 10.2967/jnumed.108.060798] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Radioimmunotherapy can effectively treat leptomeningeal metastases when radiolabeled antibodies are administered into the cerebrospinal fluid (CSF). We developed a pharmacokinetic model to evaluate the role of kinetic and transport parameters of radioimmunotherapy in maximizing the therapeutic ratio, the ratio of the area under the curve for the concentration of the bound antibodies versus time (AUC[C(IAR)]), to that for unbound antibodies (AUC[C(IA)]). METHODS We simplified the CSF space as a single compartment and considered the binding of antibodies to antigens on tumor cells lining the surface of the CSF space. Mass conservation was applied to set up the equations for C(IAR), C(IA), and other pharmacokinetic variables. A Runge-Kutta method was used to solve the equations. RESULTS This model agreed with the measured data in 10 of 14 patients in the phase I trial of intra-Ommaya radioimmunotherapy using (131)I-3F8. Using this model, we predicted that increasing the affinity of antibodies to antigens greatly increases AUC(C(IAR)) but not AUC(C(IA)); for the same amount of isotope administered, the smaller antibody dose and the higher specific activity improves therapeutic ratio. When the isotope half-life (t(1/2-I)) was 0.77 h, increasing the antibody association constant enhanced AUC(C(IAR)) much more than did decreasing the dissociation constant, even if overall affinity was unchanged. When t(1/2-I) reached 240 h, decreasing the dissociation constant would slightly enhance AUC(C(IAR)). Other predictions were that decreasing the CSF bulk flow rate would increase AUC(C(IAR)), with 3 mL/h being optimal; at the same amount of antibody administered by continuous infusion and by split administrations, compared with that by the single bolus administration, one could improve AUC(C(IAR)) by up to 1.8- and 1.7-fold, respectively; and for an antibody affinity of 10(-8) M, increasing t(1/2-I) from 0.77 up to 64 h could greatly enhance the therapeutic ratio. CONCLUSION The strong agreement between model predictions and patient data supports the validity of the assumptions and simplifications in our model. The predictions using this model are not intuitive and need to be validated in future clinical trials. The improved therapeutic ratio by optimized kinetic and transport parameters may enhance the clinical efficacy of this new treatment modality.
Collapse
Affiliation(s)
- Yonggang Lv
- Department of Biomedical Engineering, The City College of New York, New York, New York 10031, USA
| | | | | |
Collapse
|
212
|
Li Y, Song E, Abbas Rizvi SM, Power CA, Beretov J, Raja C, Cozzi PJ, Morgenstern A, Apostolidis C, Allen BJ, Russell PJ. Inhibition of micrometastatic prostate cancer cell spread in animal models by 213Bilabeled multiple targeted alpha radioimmunoconjugates. Clin Cancer Res 2009; 15:865-75. [PMID: 19188157 DOI: 10.1158/1078-0432.ccr-08-1203] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the therapeutic potential of 213Bilabeled multiple targeted alpha-radioimmunoconjugates for treating prostate cancer (CaP) micrometastases in mouse models. EXPERIMENTAL DESIGN PC-3 CaP cells were implanted s.c., in the prostate, and intratibially in NODSCID mice. The expression of multiple tumor-associated antigens on tumor xenografts and micrometastases was detected by immunohistochemistry. Targeting vectors were two monoclonal antibodies, and a plasminogen activator inhibitor type 2 that binds to cell surface urokinase plasminogen activator, labeled with 213Bi using standard methodology. In vivo efficacy of multiple alpha conjugates (MTAT) at different activities was evaluated in these mouse models. Tumor growth was monitored during observations and local regional lymph node metastases were assessed at the end of experiments. RESULTS The take rate of PC-3 cells was 100% for each route of injection. The tumor-associated antigens (MUC1, urokinase plasminogen activator, and BLCA-38) were heterogeneously expressed on primary tumors and metastatic cancer clusters at transit. A single i.p. injection of MTAT (test) at high and low doses caused regression of the growth of primary tumors and prevented local lymph node metastases in a concentration-dependent fashion; it also caused cancer cells to undergo necrosis and apoptosis. CONCLUSIONS Our results suggest that MTAT can impede primary PC-3 CaP growth at three different sites in vivo through induction of apoptosis, and can prevent the spread of cancer cells and target lymph node micrometastases in a concentration-dependent manner. MTAT, by targeting multiple antigens, can overcome heterogeneous antigen expression to kill small CaP cell clusters, thus providing a potent therapy for micrometastases.
Collapse
Affiliation(s)
- Yong Li
- Cancer Care Centre, St George Hospital, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
A tandem generator system for production of 223Ra and 211Pb/211Bi in DTPA solutions suitable for potential application in radiotherapy. J Radioanal Nucl Chem 2009. [DOI: 10.1007/s10967-009-0044-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
214
|
Drecoll E, Gaertner FC, Miederer M, Blechert B, Vallon M, Müller JM, Alke A, Seidl C, Bruchertseifer F, Morgenstern A, Senekowitsch-Schmidtke R, Essler M. Treatment of peritoneal carcinomatosis by targeted delivery of the radio-labeled tumor homing peptide bi-DTPA-[F3]2 into the nucleus of tumor cells. PLoS One 2009; 4:e5715. [PMID: 19479088 PMCID: PMC2682652 DOI: 10.1371/journal.pone.0005715] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2009] [Accepted: 04/24/2009] [Indexed: 12/02/2022] Open
Abstract
Background α-particle emitting isotopes are effective novel tools in cancer therapy, but targeted delivery into tumors is a prerequisite of their application to avoid toxic side effects. Peritoneal carcinomatosis is a widespread dissemination of tumors throughout the peritoneal cavity. As peritoneal carcinomatosis is fatal in most cases, novel therapies are needed. F3 is a tumor homing peptide which is internalized into the nucleus of tumor cells upon binding to nucleolin on the cell surface. Therefore, F3 may be an appropriate carrier for α-particle emitting isotopes facilitating selective tumor therapies. Principal Findings A dimer of the vascular tumor homing peptide F3 was chemically coupled to the α-emitter 213Bi (213Bi-DTPA-[F3]2). We found 213Bi-DTPA-[F3]2 to accumulate in the nucleus of tumor cells in vitro and in intraperitoneally growing tumors in vivo. To study the anti-tumor activity of 213Bi-DTPA-[F3]2 we treated mice bearing intraperitoneally growing xenograft tumors with 213Bi-DTPA-[F3]2. In a tumor prevention study between the days 4–14 after inoculation of tumor cells 6×1.85 MBq (50 µCi) of 213Bi-DTPA-[F3]2 were injected. In a tumor reduction study between the days 16–26 after inoculation of tumor cells 6×1.85 MBq of 213Bi-DTPA-[F3]2 were injected. The survival time of the animals was increased from 51 to 93.5 days in the prevention study and from 57 days to 78 days in the tumor reduction study. No toxicity of the treatment was observed. In bio-distribution studies we found 213Bi-DTPA-[F3]2 to accumulate in tumors but only low activities were found in control organs except for the kidneys, where 213Bi-DTPA-[F3]2 is found due to renal excretion. Conclusions/Significance In conclusion we report that 213Bi-DTPA-[F3]2 is a novel tool for the targeted delivery of α-emitters into the nucleus of tumor cells that effectively controls peritoneal carcinomatosis in preclinical models and may also be useful in oncology.
Collapse
Affiliation(s)
- Enken Drecoll
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
| | | | - Matthias Miederer
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
| | - Birgit Blechert
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
| | - Mario Vallon
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
| | - Jan M. Müller
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
| | - Andrea Alke
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
| | - Christof Seidl
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Institute for Transuranium Elements, Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Institute for Transuranium Elements, Karlsruhe, Germany
| | | | - Markus Essler
- Department of Nuclear Medicine, Klinikum-rechts-der-Isar, München, Germany
- * E-mail:
| |
Collapse
|
215
|
Akiyama K, Haba H, Tsukada K, Asai M, Toyoshima A, Sueki K, Nagame Y, Katada M. A metallofullerene that encapsulates 225Ac. J Radioanal Nucl Chem 2009. [DOI: 10.1007/s10967-009-0522-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
216
|
Villa CH, McDevitt MR, Escorcia FE, Rey DA, Bergkvist M, Batt CA, Scheinberg DA. Synthesis and biodistribution of oligonucleotide-functionalized, tumor-targetable carbon nanotubes. NANO LETTERS 2008; 8:4221-8. [PMID: 19367842 PMCID: PMC4059415 DOI: 10.1021/nl801878d] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Single-wall carbon nanotubes (SWNT) show promise as nanoscale vehicles for targeted therapies. We have functionalized SWNT using regioselective chemistries to confer capabilities of selective targeting using RGD ligands, radiotracing using radiometal chelates, and self-assembly using oligonucleotides. The constructs contained approximately 2-7 phosphorothioate oligonucleotide chains and 50-75 amines per 100 nm length of SWNT, based on a loading of 0.01-0.05 mmol/g and 0.3-0.6 mmol/g, respectively. Dynamic light scattering suggested the functionalized SWNT were well dispersed, without formation of large aggregates in physiologic solutions. The SWNT-oligonucleotide conjugate annealed with a complementary oligonucleotide sequence had a melting temperature of 54 degrees C. Biodistribution in mice was quantified using radiolabeled SWNT-oligonucleotide conjugates. Appended RGD ligands allowed for specific binding to tumor cells in a flow cytometric assay. The techniques employed should enable the synthesis of multifunctional SWNT capable of self-assembly in biological settings.
Collapse
Affiliation(s)
- Carlos H. Villa
- Molecular Pharmacology and Chemistry Program and Departments of Medicine and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021
| | - Michael R. McDevitt
- Molecular Pharmacology and Chemistry Program and Departments of Medicine and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021
| | - Freddy E. Escorcia
- Molecular Pharmacology and Chemistry Program and Departments of Medicine and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021
| | - Diego A. Rey
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853
| | - Magnus Bergkvist
- Department of Nanobioscience, College of Nanoscale Science and Engineering, University at Albany, Albany, New York 12203
| | - Carl A. Batt
- Department of Food Science, Cornell University, Ithaca, New York 14853
| | - David A. Scheinberg
- Molecular Pharmacology and Chemistry Program and Departments of Medicine and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021
- Corresponding author. . Phone: (646) 888-2190
| |
Collapse
|
217
|
Barbet J, Kraeber-Bodéré F, Chatal JF. What can be expected from nuclear medicine tomorrow? Cancer Biother Radiopharm 2008; 23:483-504. [PMID: 18771353 DOI: 10.1089/cbr.2008.010-u] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Imaging can take advantage of developments in "omics" approaches and go from routine individual biomarkers to multiple-scale biomarker profiles. Imaging structural, functional, metabolic, cellular, and molecular changes will be made possible by multimodality hybrid techniques, such as positron emission tomography-magnetic resonance imaging. Imaging should predict treatment response, look at stratification for specific treatment modalities, and look at the "omic" characterization of an individual patient or a specific tumor. This should lead to the development of "personalized" medicine. In cancer radiotherapy, patient responses should be accurately predicted. In specific cases, proton and hadrontherapy will be further enhanced by the irradiation dose delivered to the tumors. For disseminated or metastatic disease, targeted radionuclide therapy is an effective addition to the arsenal against cancer. The clinical efficacy of radiolabeled antibodies has been clearly demonstrated in lymphoma as well as that of radiolabeled peptides derived from somatostatin in the treatment of neuroendocrine tumors. Preliminary studies now show interesting results in solid tumors, too. Even if the number of objective clinical responses based on tumor shrinkage is small, targeted radionuclide therapy increases progression-free survival or overall survival in some specific cases where tumor burden is small. Avenues for further improvement are multiple and include combination with other therapeutic modalities, development of new approaches (e.g., small molecules, pretargeting, and antibody alternatives). Using alpha-emitting radionuclides is another possibility for specific diseases, such as leukemias, multiple myeloma, or brain tumor remnants.
Collapse
Affiliation(s)
- Jacques Barbet
- Centre de Recherche en Cancérologie Nantes-Angers, INSERM, Université de Nantes, Nantes, France
| | | | | |
Collapse
|
218
|
Abstract
Although the general radiobiologic principles underlying external beam therapy and radionuclide therapy are the same, there are significant differences in the biophysical and radiobiologic effects between the 2 types of radiation. In addition to the emission of particulate radiation, targeted radionuclide therapy is characterized by (1) extended exposures and, usually, declining dose rates; (2) nonuniformities in the distribution of radioactivity and, thus, absorbed dose; and (3) particles of varying ionization density and, hence, quality. This review explores the special features that distinguish the biologic effects consequent to the traversal of charged particles through mammalian cells. It also highlights what has been learned when these radionuclides and radiotargeting pharmaceuticals are used to treat cancers.
Collapse
Affiliation(s)
- Amin I Kassis
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
219
|
Sgouros G. Update:Molecular Radiotherapy: Survey and Current Status. Cancer Biother Radiopharm 2008; 23:531-40. [DOI: 10.1089/cbr.2008.0502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- George Sgouros
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD
| |
Collapse
|
220
|
Miederer M, Scheinberg DA, McDevitt MR. Realizing the potential of the Actinium-225 radionuclide generator in targeted alpha particle therapy applications. Adv Drug Deliv Rev 2008; 60:1371-82. [PMID: 18514364 DOI: 10.1016/j.addr.2008.04.009] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Accepted: 04/16/2008] [Indexed: 01/28/2023]
Abstract
Alpha particle-emitting isotopes have been proposed as novel cytotoxic agents for augmenting targeted therapy. Properties of alpha particle radiation such as their limited range in tissue of a few cell diameters and their high linear energy transfer leading to dense radiation damage along each alpha track are promising in the treatment of cancer, especially when single cells or clusters of tumor cells are targeted. Actinium-225 (225 Ac) is an alpha particle-emitting radionuclide that generates 4 net alpha particle isotopes in a short decay chain to stable 209 Bi, and as such can be described as an alpha particle nanogenerator. This article reviews the literature pertaining to the research, development, and utilization of targeted 225 Ac to potently and specifically affect cancer.
Collapse
|
221
|
Miao Y, Quinn TP. Peptide-targeted radionuclide therapy for melanoma. Crit Rev Oncol Hematol 2008; 67:213-28. [PMID: 18387816 PMCID: PMC3197246 DOI: 10.1016/j.critrevonc.2008.02.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 02/06/2008] [Accepted: 02/20/2008] [Indexed: 12/23/2022] Open
Abstract
Melanocortin-1 receptor (MC1-R) and melanin are two attractive melanoma-specific targets for peptide-targeted radionuclide therapy for melanoma. Radiolabeled peptides targeting MC1-R/melanin can selectively and specifically target cytotoxic radiation generated from therapeutic radionuclides to melanoma cells for cell killing, while sparing the normal tissues and organs. This review highlights the recent advances of peptide-targeted radionuclide therapy of melanoma targeting MC1-R and melanin. The promising therapeutic efficacies of 188Re-(Arg(11))CCMSH (188Re-[Cys(3,4,10), D-Phe(7),Arg(11)]-alpha-MSH(3-13)), 177Lu- and 212Pb-labeled DOTA-Re(Arg(11))CCMSH (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-[ReO-(Cys(3,4,10), D-Phe(7), Arg(11))]-alpha-MSH(3-13)) and 188Re-HYNIC-4B4 (188Re-hydrazinonicotinamide-Tyr-Glu-Arg-Lys-Phe-Trp-His-Gly-Arg-His) in preclinical melanoma-bearing models demonstrate an optimistic outlook for peptide-targeted radionuclide therapy for melanoma. Peptide-targeted radionuclide therapy for melanoma will likely contribute in an adjuvant setting, once the primary tumor has been surgically removed, to treat metastatic deposits and for treatment of end-stage disease. The lack of effective treatments for metastatic melanoma and end-stage disease underscores the necessity to develop and implement new treatment strategies, such as peptide-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Yubin Miao
- College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
- Cancer Research and Treatment Center, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Dermatology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Thomas P. Quinn
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
- Department of Radiology, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veteran Hospital, Columbia, MO 65201, USA
| |
Collapse
|
222
|
Sgouros G. Alpha-particles for targeted therapy. Adv Drug Deliv Rev 2008; 60:1402-6. [PMID: 18541332 DOI: 10.1016/j.addr.2008.04.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Accepted: 04/16/2008] [Indexed: 11/24/2022]
Abstract
Alpha-particles are helium nuclei that deposit DNA damaging energy along their track that is 100 to 1000 times greater than that of conventionally used beta-particle emitting radionuclides for targeted therapy; the damage caused by alpha-particles is predominately double-stranded DNA breaks severe enough so as to be almost completely irreparable. This means that a small number of tracks through a cell nucleus can sterilize a cell and that, because the damage is largely irreparable, alpha-particle radiation is not susceptible to resistance as seen with external radiotherapy (e.g., in hypoxic tissue). The ability of a single track to influence biological outcome and the stochastic nature of alpha-particle decay require statistical or microdosimetric techniques to properly reflect likely biological outcome when the biologically relevant target is small or when a low number of radionuclide decays have occurred. In therapeutic implementations, microdosimetry is typically not required and the average absorbed dose over a target volume is typically calculated. Animal and cell culture studies have shown that, per unit absorbed dose, the acute biological effects of alpha-particles are 3 to 7 times greater than the damage caused by external beam or beta-particle radiation. Over the past ten to 15 years, alpha-particle emitting radionuclides have been investigated as a possible new class of radionuclides for targeted therapy. Results from the small number of clinical trials reported to date have shown efficacy without significant toxicity.
Collapse
|
223
|
Miederer M, Henriksen G, Alke A, Mossbrugger I, Quintanilla-Martinez L, Senekowitsch-Schmidtke R, Essler M. Preclinical evaluation of the alpha-particle generator nuclide 225Ac for somatostatin receptor radiotherapy of neuroendocrine tumors. Clin Cancer Res 2008; 14:3555-61. [PMID: 18519789 DOI: 10.1158/1078-0432.ccr-07-4647] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Peptide receptor radionuclide therapy (PRRT) using somatostatin analogues labeled with beta-particle-emitting isotopes such as 90Y or 177Lu has been a promising treatment strategy for metastasized neuroendocrine tumors. Although remission can be accomplished in a high percentage of neuroendocrine tumors, some tumors do not respond to this treatment. alpha-Emitting isotopes-such as the 10-day half-life alpha-emitting generator nuclide Actinum-225 (225Ac)-are characterized by extremely high cytotoxic activity on the cellular level, and may be superior in the treatment of neuroendocrine tumors not responding to PRRT using beta-emitting isotopes. EXPERIMENTAL DESIGN Radiolabeling of 225Ac 1,4,7,10-tetra-azacylododecane N,N',N'',N'''-J-tetraacetic acid-Tyr3-octreotide (DOTATOC) was done at pH 5 (60 minutes at 70 degrees C) without further purification. Biodistribution in nude mice bearing AR42J rat pancreas neuroendocrine tumor xenografts were measured for up to 24 hours. Toxicity was tested by weight changes, retention variables (blood urea nitrogen and creatine), and histopathology in mice 7 months after treatment with 10 to 130 kBq (n = 4-5). Therapeutic efficacy was assessed by tumor weighing in animals treated 4 days after xenotransplantation and compared with 177Lu-DOTATOC as a reference. RESULTS Activities up to 20 kBq had no significant toxic effects in mice. In contrast, activities higher than 30 kBq induced tubular necrosis. Biodistribution studies revealed that 225Ac-DOTATOC effectively accumulated in neuroendocrine xenograft tumors. 225Ac-DOTATOC activities were shown to be nontoxic (12-20 kBq), reduced the growth of neuroendocrine tumors, and showed improved efficacy compared with 177Lu-DOTATOC. CONCLUSIONS 225Ac might be suitable to improve PRRT in neuroendocrine tumors.
Collapse
Affiliation(s)
- Matthias Miederer
- Nuklearmedizinische Klinik und Poliklinik, Klinikum-rechts-der-Isar, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
224
|
Chong HS, Song HA, Ma X, Milenic DE, Brady ED, Lim S, Lee H, Baidoo K, Cheng D, Brechbiel MW. Novel bimodal bifunctional ligands for radioimmunotherapy and targeted MRI. Bioconjug Chem 2008; 19:1439-47. [PMID: 18564868 DOI: 10.1021/bc800050x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The structurally novel bifunctional ligands C-NETA and C-NE3TA, each possessing both acyclic and macrocyclic moieties, were prepared and evaluated as potential chelates for radioimmunotherapy (RIT) and targeted magnetic resonance imaging (MRI). Heptadentate C-NE3TA was fortuitously discovered during the preparation of C-NETA. An optimized synthetic method to C-NETA and C-NE3TA including purification of the polar and tailing reaction intermediates, tert-butyl C-NETA (2) and tert-butyl C-NE3TA (3) using semiprep HPLC was developed. The new Gd(III) complexes of C-NETA and C-NE3TA were prepared as contrast enhancement agents for use in targeted MRI. The T 1 relaxivity data indicate that Gd(C-NETA) and Gd(C-NE3TA) possess higher relaxivity than Gd(C-DOTA), a bifunctional version of a commercially available MRI contrast agent; Gd(DOTA). C-NETA and C-NE3TA were radiolabeled with (177)Lu, (90)Y, (203)Pb, (205/6)Bi, and (153)Gd; and in vitro stability of the radiolabeled corresponding complexes was assessed in human serum. The in vitro studies indicate that the evaluated radiolabeled complexes were stable in serum for 11 days with the exception being the (203)Pb complexes of C-NETA and C-NE3TA, which dissociated in serum. C-NETA and C-NE3TA radiolabeled (177)Lu, (90)Y, or (153)Gd complexes were further evaluated for in vivo stability in athymic mice and possess excellent or acceptable in vivo biodistribution profile. (205/6)Bi- C-NE3TA exhibited extremely rapid blood clearance and low radioactivity level at the normal organs, while (205/6)Bi- C-NETA displayed low radioactivity level in the blood and all of the organs except for the kidney where relatively high renal uptake of radioactivity is observed. C-NETA and C-NE3TA were further modified for conjugation to the monoclonal antibody Trastuzumab.
Collapse
Affiliation(s)
- Hyun-Soon Chong
- Chemistry Division, Biological, Chemical, and Physical Sciences Department, Illinois Institute of Technology, Chicago, Illinois 60616, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Robinson MK, Shaller C, Garmestani K, Plascjak PS, Hodge KM, Yuan QA, Marks JD, Waldmann TA, Brechbiel MW, Adams GP. Effective treatment of established human breast tumor xenografts in immunodeficient mice with a single dose of the alpha-emitting radioisotope astatine-211 conjugated to anti-HER2/neu diabodies. Clin Cancer Res 2008; 14:875-82. [PMID: 18245551 DOI: 10.1158/1078-0432.ccr-07-1250] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Successful radioimmunotherapy strategies depend on selecting radioisotopes with physical properties complementary to the biological properties of the targeting vehicle. Small, engineered antitumor antibody fragments are capable of rapid, highly specific tumor targeting in immunodeficient mouse models. We hypothesized that the C6.5 diabody, a noncovalent anti-HER2 single-chain Fv dimer, would be an ideal radioisotope carrier for the radioimmunotherapy of established tumors using the short-lived alpha-emitting radioisotope (211)At. EXPERIMENTAL DESIGN Immunodeficient nude mice bearing established HER2/neu-positive MDA-MB-361/DYT2 tumors treated with N-succinimidyl N-(4-[(211)At]astatophenethyl)succinamate ((211)At-SAPS)-C6.5 diabody. Additional cohorts of mice were treated with (211)At-SAPS T84.66 diabody targeting the carcinoembryonic antigen or (211)At-SAPS on a diabody specific for the Müllerian inhibiting substance type II receptor, which is minimally expressed on this tumor cell line. RESULTS A single i.v. injection of (211)At-SAPS C6.5 diabody led to a 30-day delay in tumor growth when a 20 muCi dose was administered and a 57-day delay in tumor growth (60% tumor-free after 1 year) when a 45 muCi dose was used. Treatment of mice bearing the same tumors with (211)At-SAPS T84.66 diabody at the same doses led to a delay in tumor growth, but no complete responses, likely due to substantially lower expression of this antigen on the MDA-MB-361/DYT2 tumors. In contrast, a dose of 20 muCi of (211)At-SAPS on the anti-Müllerian-inhibiting substance type II receptor diabody did not affect tumor growth rate, demonstrating specificity of the therapeutic effect. CONCLUSIONS These findings indicate that diabody molecules can be effective agents for targeted radioimmunotherapy of solid tumors using powerful, short-lived alpha-emitting radioisotopes.
Collapse
Affiliation(s)
- Matthew K Robinson
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Sgouros G, Song H. Cancer stem cell targeting using the alpha-particle emitter, 213Bi: mathematical modeling and feasibility analysis. Cancer Biother Radiopharm 2008; 23:74-81. [PMID: 18298331 DOI: 10.1089/cbr.2007.0408] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There is increasing recognition that treatment failure in cancer may be associated with the failure to sterilize a small subpopulation of tumor cells that have been characterized as tumor stem cells. Defined as cells that are able to self-renew and also to replenish a phenotypically diverse tumor-cell population, such cells are also considered resistant to chemotherapy. These characteristics are optimal for targeting by using alpha-particle-emitting radionuclides. Because of their high-energy deposition density per track, alpha-particles are capable of targeting single cells or small clusters of cells with minimal normal organ toxicity. The DNA damage induced by alpha-particles is largely irreparable and, therefore, alpha-particle-induced damage is minimally susceptible to resistance mechanisms. In this work, theoretical modeling was performed to examine the potential of alpha-emitter targeting of such small clusters of cancer stem cells. Critical parameters influencing efficacy and toxicity were identified and their relationship elucidated. The results identify specific activity, antigen site density, and number of target cells as critical parameters for effective cell killing and demonstrate substantial efficacy gains by targeting a smaller number of stem cells, as opposed to the entire tumor-cell population.
Collapse
Affiliation(s)
- George Sgouros
- Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD 21231, USA.
| | | |
Collapse
|
227
|
Therapy of human carcinoma xenografts with antibodies to EGFr and HER-2 conjugated to radionuclides emitting low-energy electrons. Eur J Nucl Med Mol Imaging 2008; 35:1249-58. [PMID: 18265976 DOI: 10.1007/s00259-008-0731-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 01/15/2008] [Indexed: 12/31/2022]
Abstract
PURPOSE Low-energy electrons (10-50 keV) can be effective and specific cytotoxic agents when delivered to the cell surface by antibodies, because their path length in tissue is comparable to a cell diameter. In this study, we have begun to evaluate the therapeutic potential of antibodies (Abs) conjugated to (111)In against carcinoma xenografts in nude mice. METHODS Abs to EGFr or HER-2 were labeled with (111)In to a high specific activity of approximately 1.48 GBq/mg (40 mCi/mg). They were injected into nude mice 5-6 days after inoculation of human carcinoma cells, either A431 or SK-OV-3, and tumor growth was monitored. In preliminary in vitro experiments, we calculated the cumulative decays per cell, estimated the centigray dose delivered to the nucleus, and related this to the fraction surviving. RESULTS Abs to both antigens provided significant protection in nude mouse xenograft models (p values ranging from <0.05 to <0.001). Some mice appeared to be cured, but most had delayed tumor growth. The specificity of the effect was demonstrated by testing non-reactive Abs labeled in the same way. The radioactivity was required, because unconjugated Abs had no therapeutic effect. The maximum tolerated dose was required in order for therapy to be effective, but most of the treated mice had no significant weight loss or other overt signs of toxicity. CONCLUSION Abs labeled with nuclides emitting low-energy electrons, such as (111)In, can be effective therapeutic agents against microscopic s.c. tumors. This strategy should be considered for clinical applications.
Collapse
|
228
|
Escorcia FE, McDevitt MR, Villa CH, Scheinberg DA. Targeted nanomaterials for radiotherapy. Nanomedicine (Lond) 2008; 2:805-15. [PMID: 18095847 DOI: 10.2217/17435889.2.6.805] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nanomaterials have garnered increasing interest recently as potential therapeutic drug-delivery vehicles. Among the existing nanomaterials are the pure carbon-based particles, such as fullerenes and nanotubes, various organic dendrimers, liposomes and other polymeric compounds. These vehicles have been decorated with a wide spectrum of target-reactive ligands, such as antibodies and peptides, which interact with cell-surface tumor antigens or vascular epitopes. Once targeted, these new nanomaterials can then deliver radioisotopes or isotope generators to the cancer cells. Here, we will review some of the more common nanomaterials under investigation and their current and future applications as drug-delivery scaffolds with particular emphasis on targeted cancer radiotherapy.
Collapse
Affiliation(s)
- Freddy E Escorcia
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology and Chemistry Program, New York, NY 10021, USA
| | | | | | | |
Collapse
|
229
|
Radioimmunotherapy. Clin Nucl Med 2008. [DOI: 10.1007/978-3-540-28026-2_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
230
|
Abstract
This review describes strategies for the delivery of therapeutic radionuclides to tumor sites. Therapeutic approaches are summarized in terms of tumor location in the body, and tumor morphology. These determine the radionuclides of choice for suggested targeting ligands, and the type of delivery carriers. This review is not exhaustive in examples of radionuclide carriers for targeted cancer therapy. Our purpose is two-fold: to give an integrated picture of the general strategies and molecular constructs currently explored for the delivery of therapeutic radionuclides, and to identify challenges that need to be addressed. Internal radiotherapies for targeting of cancer are at a very exciting and creative stage. It is expected that the current emphasis on multidisciplinary approaches for exploring such therapeutic directions should enable internal radiotherapy to reach its full potential.
Collapse
Affiliation(s)
- Stavroula Sofou
- Laboratory for Drug Delivery Systems, Othmer-Jacobs Department of Chemical and Biological Engineering, Polytechnic University, Brooklyn, NY 11201, USA.
| |
Collapse
|
231
|
Pagel J. Radioimmunotherapeutic approaches for leukemia: the past, present and future. Cytotherapy 2008; 10:13-20. [DOI: 10.1080/14653240701679881] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
232
|
Brumlik MJ, Daniel BJ, Waehler R, Curiel DT, Giles FJ, Curiel TJ. Trends in immunoconjugate and ligand-receptor based targeting development for cancer therapy. Expert Opin Drug Deliv 2007; 5:87-103. [DOI: 10.1517/17425247.5.1.87] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
233
|
Stein R, Mattes MJ, Cardillo TM, Hansen HJ, Chang CH, Burton J, Govindan S, Goldenberg DM. CD74: a new candidate target for the immunotherapy of B-cell neoplasms. Clin Cancer Res 2007; 13:5556s-5563s. [PMID: 17875789 DOI: 10.1158/1078-0432.ccr-07-1167] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CD74 is an integral membrane protein that functions as a MHC class II chaperone. Moreover, it has recently been shown to have a role as an accessory-signaling molecule and has been implicated in malignant B-cell proliferation and survival. These biological functions combined with expression of CD74 on malignant B cells and limited expression on normal tissues implicate CD74 as a potential therapeutic target. The anti-CD74 monoclonal antibody LL1 has been humanized (hLL1 milatuzumab or IMMU-115) and can provide the basis for novel therapeutic approaches to B-cell malignancies, particularly because this antibody shows rapid internalization into CD74+ malignant cells. This article reviews the preclinical evaluations of LL1, its humanized form, and isotope, drug, and toxin conjugates. These studies show that unconjugated hLL1 and conjugates of hLL1 constructs with radioisotopes, doxorubicin, and frog RNase have high antitumor activity in non-Hodgkin's lymphoma and multiple myeloma in vitro and in tumor xenograft models. Single-dose studies of hLL1 in monkeys showed no adverse effects but did decrease circulating B and T lymphocytes and natural killer cells. When evaluated in combination with rituximab, either equivalent or improved efficacy, compared with either antibody alone, was observed. CD74 is a new candidate target for the immunotherapy of neoplasms expressing this antigen, which can be exploited using either a naked antibody or conjugated to isotopes, drugs, or toxins.
Collapse
Affiliation(s)
- Rhona Stein
- Garden State Cancer Center, Center for Molecular Medicine and Immunology, Belleville, New Jersey 07109, USA.
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Chong HS, Ma X, Le T, Kwamena B, Milenic DE, Brady ED, Song HA, Brechbiel MW. Rational design and generation of a bimodal bifunctional ligand for antibody-targeted radiation cancer therapy. J Med Chem 2007; 51:118-25. [PMID: 18062661 DOI: 10.1021/jm070401q] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An antibody-targeted radiation therapy (radioimmunotherapy, RIT) employs a bifunctional ligand that can effectively hold a cytotoxic metal with clinically acceptable complexation kinetics and stability while being attached to a tumor-specific antibody. Clinical exploration of the therapeutic potential of RIT has been challenged by the absence of adequate ligand, a critical component for enhancing the efficacy of the cancer therapy. To address this deficiency, the bifunctional ligand C-NETA in a unique structural class possessing both a macrocyclic cavity and a flexible acyclic moiety was designed. The practical, reproducible, and readily scalable synthetic route to C-NETA was developed, and its potential as the chelator of (212)Bi, (213)Bi, and (177)Lu for RIT was evaluated in vitro and in vivo. C-NETA rapidly binds both Lu(III) and Bi(III), and the respective metal complexes remain extremely stable in serum for 14 days. (177)Lu -C-NETA and (205/6)Bi -C-NETA possess an excellent or acceptable in vivo biodistribution profile.
Collapse
Affiliation(s)
- Hyun-Soon Chong
- Chemistry Division, Biological, Chemical, and Physical Sciences Department, Illinois Institute of Technology, Chicago, Illinois 60616, USA.
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Abstract
Monoclonal antibodies have become a viable strategy for the delivery of therapeutic, particle emitting radionuclides specifically to tumor cells to either augment anti-tumor action of the native antibodies or to solely take advantage of their action as targeting vectors. Proper and rational selection of radionuclide and antibody combinations is critical to making radioimmunotherapy (RIT) a standard therapeutic modality due to the fundamental and significant differences in the emission of either alpha- and beta-particles. The alpha-particle has a short path length (50-80 microm) that is characterized by high linear energy transfer (100 keV microm(-1)). Actively targeted alpha-therapy potentially offers a more specific tumor cell killing action with less collateral damage to the surrounding normal tissues than beta-emitters. These properties make targeted alpha-therapy an appropriate therapy to eliminate minimal residual or micrometastatic disease. RIT using alpha-emitters such as (213)Bi, (211)At, (225)Ac, and others has demonstrated significant activity in both in vitro and in vivo model systems. Limited numbers of clinical trials have progressed to demonstrate safety, feasibility, and therapeutic activity of targeted alpha-therapy, despite having to traverse complex obstacles. Further advances may require more potent isotopes, additional sources and more efficient means of isotope production. Refinements in chelation and/or radiolabeling chemistry combined with rational improvements of isotope delivery, targeting vectors, molecular targets, and identification of appropriate clinical applications remain as active areas of research. Ultimately, randomized trials comparing targeted alpha-therapy combined with integration into existing standards of care treatment regimens will determine the clinical utility of this modality.
Collapse
Affiliation(s)
- Martin W Brechbiel
- Radioimmune & Inorganic Chemistry Section Radiation Oncology Branch, NCI, NIH Building 10, Room 1B40 10 Center Drive Bethesda, MD 20892-1088, USA.
| |
Collapse
|
236
|
Oyen WJG, Bodei L, Giammarile F, Maecke HR, Tennvall J, Luster M, Brans B. Targeted therapy in nuclear medicine—current status and future prospects. Ann Oncol 2007; 18:1782-92. [PMID: 17434893 DOI: 10.1093/annonc/mdm111] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In recent years, a number of new developments in targeted therapies using radiolabeled compounds have emerged. New developments and insights in radioiodine treatment of thyroid cancer, treatment of lymphoma and solid tumors with radiolabeled monoclonal antibodies (mAbs), the developments in the application of radiolabeled small receptor-specific molecules such as meta-iodobenzylguanidine and peptides and the position of locoregional treatment in malignant involvement of the liver are reviewed. The introduction of recombinant human thyroid-stimulating hormone and the possibility to enhance iodine uptake with retinoids has changed the radioiodine treatment protocol of patients with thyroid cancer. Introduction of radiolabeled mAbs has provided additional treatment options in patients with malignant lymphoma, while a similar approach proves to be cumbersome in patients with solid tumors. With radiolabeled small molecules that target specific receptors on tumor cells, high radiation doses can be directed to tumors in patients with disseminated disease. Radiolabeled somatostatin derivatives for the treatment of neuroendocrine tumors are the role model for this approach. Locoregional treatment with radiopharmaceuticals of patients with hepatocellular carcinoma or metastases to the liver may be used in inoperable cases, but may also be of benefit in a neo-adjuvant or adjuvant setting. Significant developments in the application of targeted radionuclide therapy have taken place. New treatment modalities have been introduced in the clinic. The concept of combining therapeutic radiopharmaceuticals with other treatment modalities is more extensively explored.
Collapse
Affiliation(s)
- W J G Oyen
- Therapy Committee of the European Association of Nuclear Medicine, Hollandstrasse 14 / Mezzanine, A-1020 Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
237
|
Melhus KB, Larsen RH, Stokke T, Kaalhus O, Selbo PK, Dahle J. Evaluation of the binding of radiolabeled rituximab to CD20-positive lymphoma cells: an in vitro feasibility study concerning low-dose-rate radioimmunotherapy with the alpha-emitter 227Th. Cancer Biother Radiopharm 2007; 22:469-79. [PMID: 17803441 DOI: 10.1089/cbr.2007.371] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Radioimmunotherapy (RIT) with the alpha-emitter 227Th is currently under evaluation. 227Th is conjugated to the chimeric anti-CD20 monoclonal antibody rituximab, using the chelator p-isothiocyanato-benzyl-DOTA. In this study, the binding of 227Th-DOTA-p-benzyl-rituximab to three different CD-20-positive lymphoma cell lines, Raji, Rael, and Daudi, were evaluated. Equilibrium and kinetic binding experiments were used to determine binding parameters, including the association and dissociation rate constants, the equilibrium dissociation constants, and the total number of antigens for Raji, Rael, and Daudi cells. There were significant differences between the cell lines with respect to both Kd and the total number of antigens. Rael cells had more than three times as many antigens as the other two cell lines, and the functional Kd found for Rael cells was significantly higher than that found for Raji and Daudi cells. These results were confirmed using flow cytometry. Rituximab was found to be localized in patches on the cell membrane. The findings indicated that 227Th-labeled rituximab has relevant antigen-targeting properties for radioimmunotherapy.
Collapse
Affiliation(s)
- Katrine B Melhus
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
238
|
Sofou S, Kappel BJ, Jaggi JS, McDevitt MR, Scheinberg DA, Sgouros G. Enhanced retention of the alpha-particle-emitting daughters of Actinium-225 by liposome carriers. Bioconjug Chem 2007; 18:2061-7. [PMID: 17935286 DOI: 10.1021/bc070075t] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Targeted alpha-particle emitters hold great promise as therapeutics for micrometastatic disease. Because of their high energy deposition and short range, tumor targeted alpha-particles can result in high cancer-cell killing with minimal normal-tissue irradiation. Actinium-225 is a potential generator for alpha-particle therapy: it decays with a 10-day half-life and generates three alpha-particle-emitting daughters. Retention of (225)Ac daughters at the target increases efficacy; escape and distribution throughout the body increases toxicity. During circulation, molecular carriers conjugated to (225)Ac cannot retain any of the daughters. We previously proposed liposomal encapsulation of (225)Ac to retain the daughters, whose retention was shown to be liposome-size dependent. However, daughter retention was lower than expected: 22% of theoretical maximum decreasing to 14%, partially due to the binding of (225)Ac to the phospholipid membrane. In this study, Multivesicular liposomes (MUVELs) composed of different phospholipids were developed to increase daughter retention. MUVELs are large liposomes with entrapped smaller lipid-vesicles containing (225)Ac. PEGylated MUVELs stably retained over time 98% of encapsulated (225)Ac. Retention of (213)Bi, the last daughter, was 31% of the theoretical maximum retention of (213)Bi for the liposome sizes studied. MUVELs were conjugated to an anti-HER2/neu antibody (immunolabeled MUVELs) and were evaluated in vitro with SKOV3-NMP2 ovarian cancer cells, exhibiting significant cellular internalization (83%). This work demonstrates that immunolabeled MUVELs might be able to deliver higher fractions of generated alpha-particles per targeted (225)Ac compared to the relative fractions of alpha-particles delivered by (225)Ac-labeled molecular carriers.
Collapse
Affiliation(s)
- Stavroula Sofou
- Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | | | | | | | | | |
Collapse
|
239
|
McDevitt MR, Chattopadhyay D, Jaggi JS, Finn RD, Zanzonico PB, Villa C, Rey D, Mendenhall J, Batt CA, Njardarson JT, Scheinberg DA. PET imaging of soluble yttrium-86-labeled carbon nanotubes in mice. PLoS One 2007; 2:e907. [PMID: 17878942 PMCID: PMC1975469 DOI: 10.1371/journal.pone.0000907] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Accepted: 08/20/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The potential medical applications of nanomaterials are shaping the landscape of the nanobiotechnology field and driving it forward. A key factor in determining the suitability of these nanomaterials must be how they interface with biological systems. Single walled carbon nanotubes (CNT) are being investigated as platforms for the delivery of biological, radiological, and chemical payloads to target tissues. CNT are mechanically robust graphene cylinders comprised of sp(2)-bonded carbon atoms and possessing highly regular structures with defined periodicity. CNT exhibit unique mechanochemical properties that can be exploited for the development of novel drug delivery platforms. In order to evaluate the potential usefulness of this CNT scaffold, we undertook an imaging study to determine the tissue biodistribution and pharmacokinetics of prototypical DOTA-functionalized CNT labeled with yttrium-86 and indium-111 ((86)Y-CNT and (111)In-CNT, respectively) in a mouse model. METHODOLOGY AND PRINCIPAL FINDINGS The (86)Y-CNT construct was synthesized from amine-functionalized, water-soluble CNT by covalently attaching multiple copies of DOTA chelates and then radiolabeling with the positron-emitting metal-ion, yttrium-86. A gamma-emitting (111)In-CNT construct was similarly prepared and purified. The constructs were characterized spectroscopically, microscopically, and chromatographically. The whole-body distribution and clearance of yttrium-86 was characterized at 3 and 24 hours post-injection using positron emission tomography (PET). The yttrium-86 cleared the blood within 3 hours and distributed predominantly to the kidneys, liver, spleen and bone. Although the activity that accumulated in the kidney cleared with time, the whole-body clearance was slow. Differential uptake in these target tissues was observed following intravenous or intraperitoneal injection. CONCLUSIONS The whole-body PET images indicated that the major sites of accumulation of activity resulting from the administration of (86)Y-CNT were the kidney, liver, spleen, and to a much less extent the bone. Blood clearance was rapid and could be beneficial in the use of short-lived radionuclides in diagnostic applications.
Collapse
Affiliation(s)
- Michael R. McDevitt
- Molecular Pharmacology and Chemistry Department, Departments of Medicine, Radiology, and Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * To whom correspondence should be addressed. E-mail: (MM); (DS)
| | - Debjit Chattopadhyay
- Molecular Pharmacology and Chemistry Department, Departments of Medicine, Radiology, and Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Jaspreet S. Jaggi
- Molecular Pharmacology and Chemistry Department, Departments of Medicine, Radiology, and Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ronald D. Finn
- Molecular Pharmacology and Chemistry Department, Departments of Medicine, Radiology, and Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Pat B. Zanzonico
- Molecular Pharmacology and Chemistry Department, Departments of Medicine, Radiology, and Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Carlos Villa
- Molecular Pharmacology and Chemistry Department, Departments of Medicine, Radiology, and Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Diego Rey
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Juana Mendenhall
- Department of Food Science, Cornell University, Ithaca, New York, United States of America
| | - Carl A. Batt
- Department of Food Science, Cornell University, Ithaca, New York, United States of America
| | - Jon T. Njardarson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - David A. Scheinberg
- Molecular Pharmacology and Chemistry Department, Departments of Medicine, Radiology, and Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * To whom correspondence should be addressed. E-mail: (MM); (DS)
| |
Collapse
|
240
|
Pope-Harman A, Cheng MMC, Robertson F, Sakamoto J, Ferrari M. Biomedical nanotechnology for cancer. Med Clin North Am 2007; 91:899-927. [PMID: 17826110 DOI: 10.1016/j.mcna.2007.05.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nanotechnology may hold the key to controlling many devastating diseases. In the fight against the pain, suffering, and death due to cancer, nanotechnology will allow earlier diagnosis and even prevention of malignancy at premalignant stages, in addition to providing multimodality treatment not possible with current conventional techniques. This review discusses nanotechnology already used in diagnostic and therapeutic applications for cancer. Also addressed are theoretic and evolving uses of nanotechnology, including multifunctional nanoparticles for imaging and therapy, nanochannel implants for controlled release of drugs, nanoscale devices for evaluation of proteomics and genomics, and diagnostic techniques that take advantage of physical changes in diseased tissue.
Collapse
Affiliation(s)
- Amy Pope-Harman
- Dorothy M. Davis Heart and Lung Research Institute, Department of Internal Medicine, The Ohio State University College of Medicine and Public Health, Columbus, OH 43210, USA.
| | | | | | | | | |
Collapse
|
241
|
Nolan DJ, Ciarrocchi A, Mellick AS, Jaggi JS, Bambino K, Gupta S, Heikamp E, McDevitt MR, Scheinberg DA, Benezra R, Mittal V. Bone marrow-derived endothelial progenitor cells are a major determinant of nascent tumor neovascularization. Genes Dev 2007; 21:1546-58. [PMID: 17575055 PMCID: PMC1891431 DOI: 10.1101/gad.436307] [Citation(s) in RCA: 297] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tumors build vessels by cooption of pre-existing vasculature and de novo recruitment of bone marrow (BM)-derived endothelial progenitor cells (EPCs). However, the contribution and the functional role of EPCs in tumor neoangiogenesis are controversial. Therefore, by using genetically marked BM progenitor cells, we demonstrate the precise spatial and temporal contribution of EPCs to the neovascularization of three transplanted and one spontaneous breast tumor in vivo using high-resolution microscopy and flow cytometry. We show that early tumors recruit BM-derived EPCs that differentiate into mature BM-derived endothelial cells (ECs) and luminally incorporate into a subset of sprouting tumor neovessels. Notably, in later tumors, these BM-derived vessels are diluted with non-BM-derived vessels from the periphery, which accounts for purported differences in previously published reports. Furthermore, we show that specific ablation of BM-derived EPCs with alpha-particle-emitting anti-VE-cadherin antibody markedly impaired tumor growth associated with reduced vascularization. Our results demonstrate that BM-derived EPCs are critical components of the earliest phases of tumor neoangiogenesis.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Line, Tumor
- Endothelial Cells/pathology
- Female
- Hematopoietic Stem Cells/pathology
- Male
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neovascularization, Pathologic
Collapse
Affiliation(s)
- Daniel J. Nolan
- Cancer Genome Research Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, New York 11794, USA
| | - Alessia Ciarrocchi
- Program in Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | - Albert S. Mellick
- Cancer Genome Research Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
| | - Jaspreet S. Jaggi
- Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | - Kathryn Bambino
- Cancer Genome Research Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
| | - Sunita Gupta
- Cancer Genome Research Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
| | - Emily Heikamp
- Cancer Genome Research Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
| | - Michael R. McDevitt
- Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | - David A. Scheinberg
- Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | - Robert Benezra
- Program in Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | - Vivek Mittal
- Cancer Genome Research Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
- Corresponding author.E-MAIL ; FAX (516) 422-4109
| |
Collapse
|
242
|
Dahle J, Borrebaek J, Jonasdottir TJ, Hjelmerud AK, Melhus KB, Bruland ØS, Press OW, Larsen RH. Targeted cancer therapy with a novel low-dose rate alpha-emitting radioimmunoconjugate. Blood 2007; 110:2049-56. [PMID: 17536011 DOI: 10.1182/blood-2007-01-066803] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alpha-emitting radionuclides are highly cytotoxic and are of considerable interest in the treatment of cancer. A particularly interesting approach is in radioimmunotherapy. However, alpha-emitting antibody conjugates have been difficult to exploit clinically due to the short half-life of the radionuclides, low production capability, or limited source materials. We have developed a novel technology based on the low-dose rate alpha-particle-emitting nuclide (227)Th, exemplified here using the monoclonal antibody rituximab. In vitro, this radioimmunoconjugate killed lymphoma cells at Becquerel per milliliter (Bq/mL) levels. A single injection of (227)Th-rituximab induced complete tumor regression in up to 60% of nude mice bearing macroscopic (32-256 mm(3)) human B-lymphoma xenografts at Becquerel per gram (Bq/g) levels without apparent toxicity. Therapy with (227)Th-rituximab was significantly more effective than the control radioimmunoconjugate (227)Th-trastuzumab and the standard beta-emitting radioimmunoconjugate for CD20(+) lymphoma(90)Y-tiuxetan-ibritumomab. Thorium-227 based constructs may provide a novel approach for targeted therapy against a wide variety of cancers.
Collapse
MESH Headings
- Alpha Particles
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Antigens, CD20/immunology
- Cell Proliferation
- Female
- Humans
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/therapeutic use
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Organometallic Compounds/chemistry
- Organometallic Compounds/pharmacokinetics
- Organometallic Compounds/therapeutic use
- Radioimmunotherapy
- Rituximab
- Survival Rate
- Thallium Radioisotopes/pharmacokinetics
- Tissue Distribution
- Tumor Cells, Cultured
- Yttrium Radioisotopes/pharmacokinetics
Collapse
Affiliation(s)
- Jostein Dahle
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
243
|
Zhang M, Yao Z, Patel H, Garmestani K, Zhang Z, Talanov VS, Plascjak PS, Goldman CK, Janik JE, Brechbiel MW, Waldmann TA. Effective therapy of murine models of human leukemia and lymphoma with radiolabeled anti-CD30 antibody, HeFi-1. Proc Natl Acad Sci U S A 2007; 104:8444-8. [PMID: 17488826 PMCID: PMC1895969 DOI: 10.1073/pnas.0702496104] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
CD30 is a member of the TNF receptor superfamily. Overexpression of CD30 on some neoplasms versus limited expression on normal tissues makes this receptor a promising target for antibody-based therapy. Radioimmunotherapy of cancer with radiolabeled antibodies has shown promise. In this study, we evaluated the therapeutic efficacy of an anti-CD30 antibody, HeFi-1, armed with (211)At in a leukemia (karpas299) model and with (90)Y in a lymphoma (SUDHL-1) model. Furthermore, we investigated the combination therapy of (211)At-HeFi-1 with unmodified HeFi-1 in the leukemia model. Treatment with unmodified HeFi-1 significantly prolonged the survival of the karpas299-bearing mice compared with the controls (P < 0.001). Treatment with (211)At-HeFi-1 showed greater therapeutic efficacy than that with unmodified HeFi-1 as shown by survival of the mice (P < 0.001). Combining these two agents further improved the survival of the mice compared with the groups treated with either (211)At-HeFi-1 (P < 0.05) or unmodified HeFi-1 (P < 0.001) alone. In the lymphoma model, the survival of the SUDHL-1-bearing mice was significantly prolonged by the treatment with (90)Y-HeFi-1 compared with the controls (P < 0.001). In summary, radiolabeled HeFi-1 is very promising for the treatment of CD30-expressing leukemias and lymphomas, and the combination regimen of (211)At-HeFi-1 with unmodified HeFi-1 enhanced the therapeutic efficacy.
Collapse
Affiliation(s)
- Meili Zhang
- *Metabolism Branch and
- Laboratory Animal Science Program and
| | | | | | - Kayhan Garmestani
- *Metabolism Branch and
- Applied/Developmental Research Support Program, Science Applications International Corporation–Frederick, National Cancer Institute–Frederick, Frederick, MD 21702
| | | | - Vladimir S. Talanov
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute
| | - Paul S. Plascjak
- PET Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892; and
| | | | | | - Martin W. Brechbiel
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute
| | - Thomas A. Waldmann
- *Metabolism Branch and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
244
|
Abstract
Nanotechnology is increasingly finding use in the management of cancer. Nanoscale devices have impacted cancer biology at three levels: early detection using, for example, nanocantilevers or nanoparticles; tumour imaging using radiocontrast nanoparticles or quantum dots; and drug delivery using nanovectors and hybrid nanoparticles. This review addresses some of the major milestones in the integration of nanotechnology and cancer biology, and the future of nanoscale approaches for cancer management.
Collapse
Affiliation(s)
- S Sengupta
- BWH-HST Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- E-mail:
| | - R Sasisekharan
- Biological Engineering Division, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- E-mail:
| |
Collapse
|
245
|
Diener MD, Alford JM, Kennel SJ, Mirzadeh S. 212Pb@C60 and Its Water-Soluble Derivatives: Synthesis, Stability, and Suitability for Radioimmunotherapy. J Am Chem Soc 2007; 129:5131-8. [PMID: 17394315 DOI: 10.1021/ja068639b] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fullerenes could potentially play a valuable role in radioimmunotherapy by more stably encapsulating radionuclides, especially where conventional chelation chemistry is inadequate due to the physical and/or chemical properties of the radionuclide. One of the therapeutically useful radionuclides that requires improved containment in vivo is 212Pb (tau1/2 = 10.6 h), the beta-emitting parent to alpha-emitting 212Bi (tau1/2 = 60.6 min). Myelotoxicity resulting from the accumulation of 212Pb in the bone marrow has limited the use of this radionuclide despite its favorable decay characteristics. In this work, 212Pb@C60 and its malonic ester derivatives were prepared for the first time by allowing the 212Pb to recoil into C60 following alpha-decay from its parent, 0.15-s 216Po, generated in situ from the decay of 224Ra (tau1/2 = 15 days). Repeated washing of the organic phase containing the 212Pb@C60 malonic esters with challenge solutions containing cold Pb2+ ions demonstrated that some of the 212Pb could not be exchanged and was apparently inside of the fullerenes. Malonic esters of endohedral alpha-emitting 213Bi (tau1/2 = 45 min) fullerenes were prepared by an analogous procedure. Following acidification of the esters, a preliminary biodistribution study in mice was performed with the untargeted water-soluble radiofullerenes. It was found that 212Pb did not accumulate in bone after being administered as an endohedral fullerene, in contrast to results with polyhydroxylated radiofullerenes and conventional polyaminocarboxylate chelators for 212Pb. The results indicate that 212Pb is held more tightly in the fullerene than in other methods and suggest that fullerenes may have an important role in the targeted delivery of 212Pb.
Collapse
Affiliation(s)
- Michael D Diener
- TDA Research Incorporated, 12345 West 52nd Avenue, Wheat Ridge, Colorado 80033 and Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831-6229, USA.
| | | | | | | |
Collapse
|
246
|
Humblet V, Misra P, Frangioni JV. An HPLC/mass spectrometry platform for the development of multimodality contrast agents and targeted therapeutics: prostate-specific membrane antigen small molecule derivatives. CONTRAST MEDIA & MOLECULAR IMAGING 2007; 1:196-211. [PMID: 17193697 PMCID: PMC2734331 DOI: 10.1002/cmmi.106] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The production of disease-targeted agents requires the covalent conjugation of a targeting molecule with a contrast agent or therapeutic, followed by purification of the product to homogeneity. Typical targeting molecules, such as small molecules and peptides, often have high charge-to-mass ratios and/or hydrophobicity. Contrast agents and therapeutics themselves are also diverse, and include lanthanide chelates for MRI, (99m)Tc chelates for SPECT, (90)Y chelates for radiotherapy, (18)F derivatives for PET, and heptamethine indocyanines for near-infrared fluorescent optical imaging. We have constructed a general-purpose HPLC/mass spectrometry platform capable of purifying virtually any targeted agent for any modality. The analytical sub-system is composed of a single dual-head pump that directs mobile phase to either a hot cell for the purification of radioactive agents or to an ES-TOF MS for the purification of nonradioactive agents. Nonradioactive agents are also monitored during purification by ELSD, absorbance and fluorescence. The preparative sub-system is composed of columns and procedures that permit rapid scaling from the analytical system. To demonstrate the platform's utility, we describe the preparation of five small molecule derivatives specific for prostate-specific membrane antigen (PSMA): a gadolinium derivative for MRI, indium, rhenium and technetium derivatives for SPECT, and an yttrium derivative for radiotherapy. All five compounds are derived from a highly anionic targeting ligand engineered to have a single nucleophile for N-hydroxysuccinimide-based conjugation. We also describe optimized column/mobile phase combinations and mass spectrometry settings for each class of agent, and discuss strategies for purifying molecules with extreme charge and/or hydrophobicity. Taken together, our study should expedite the development of disease-targeted, multimodality diagnostic and therapeutic agents.
Collapse
Affiliation(s)
- Valerie Humblet
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Preeti Misra
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - John V. Frangioni
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, MA 02215
- To whom correspondence should be addressed: John V. Frangioni, M.D., Ph.D., Beth Israel Deaconess Medical Center, Room SL-B05, 330 Brookline Avenue, Boston, MA 02215, 617-667-0692 FAX: 617-667-0981,
| |
Collapse
|
247
|
Nijhara R, Balakrishnan K. Bringing nanomedicines to market: regulatory challenges, opportunities, and uncertainties. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2007; 2:127-36. [PMID: 17292125 DOI: 10.1016/j.nano.2006.04.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Accepted: 04/08/2006] [Indexed: 11/28/2022]
Abstract
Scientists and entrepreneurs who contemplate developing nanomedicine products face several unique challenges in addition to many of the traditional hurdles of product development. In this review we analyze the major physicochemical, biologic and functional characteristics of several nanomedicine products on the market and explore the question of what made them unique. What made them successful? We also focus on the regulatory challenges faced by nanomedicine product developers. Based on these analyses, we propose the factors that are most likely to contribute to the success of nanomedicine products.
Collapse
Affiliation(s)
- Ruchika Nijhara
- University of Maryland, Office of Research and Development, Baltimore, MD, USA
| | | |
Collapse
|
248
|
Faunce TA. Nanotherapeutics: new challenges for safety and cost-effectiveness regulation in Australia. Med J Aust 2007; 186:189-91. [PMID: 17309421 DOI: 10.5694/j.1326-5377.2007.tb00860.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 10/18/2006] [Indexed: 11/17/2022]
Abstract
Nanotechnology is a revolutionary field of micro-manufacturing involving manipulation, by chemical or physical processes, of individual atoms and molecules. Pharmaceutical and medical device manufacturers, both in Australia and internationally, have significant investments in nanotechnology research and development. It is important that safety regulation of nanotherapeutics keep pace with this growing level of industry interest. A recent senate inquiry recommended the establishment of a working party, including representatives of the Therapeutic Goods Administration, to consider whether bulk materials classified as safe should be routinely reassessed for use at the nanoscale level by a permanent, distinct nanotechnology regulator. Safety regulation of nanotherapeutics may present unique risk assessment challenges, given the novelty and variety of products, high mobility and reactivity of engineered nanoparticles, and blurring of the diagnostic and therapeutic classifications of "medicines" and "medical devices". Nanotherapeutics is likely to make increasing claims on a particular area of Australian health care regulatory strength: scientific cost-effectiveness assessment of innovation in medical products. Any review of Australian regulation of nanotechnology should include a critical analysis of both safety issues and cost-effectiveness assessment systems for nanotherapeutics.
Collapse
Affiliation(s)
- Thomas A Faunce
- Medical School and Faculty of Law, Australian National University, Canberra, ACT.
| |
Collapse
|
249
|
Jaggi JS, Henke E, Seshan SV, Kappel BJ, Chattopadhyay D, May C, McDevitt MR, Nolan D, Mittal V, Benezra R, Scheinberg DA. Selective alpha-particle mediated depletion of tumor vasculature with vascular normalization. PLoS One 2007; 2:e267. [PMID: 17342201 PMCID: PMC1801076 DOI: 10.1371/journal.pone.0000267] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Accepted: 02/12/2007] [Indexed: 12/21/2022] Open
Abstract
Background Abnormal regulation of angiogenesis in tumors results in the formation of vessels that are necessary for tumor growth, but compromised in structure and function. Abnormal tumor vasculature impairs oxygen and drug delivery and results in radiotherapy and chemotherapy resistance, respectively. Alpha particles are extraordinarily potent, short-ranged radiations with geometry uniquely suitable for selectively killing neovasculature. Methodology and Principal Findings Actinium-225 (225Ac)-E4G10, an alpha-emitting antibody construct reactive with the unengaged form of vascular endothelial cadherin, is capable of potent, selective killing of tumor neovascular endothelium and late endothelial progenitors in bone-marrow and blood. No specific normal-tissue uptake of E4G10 was seen by imaging or post-mortem biodistribution studies in mice. In a mouse-model of prostatic carcinoma, 225Ac-E4G10 treatment resulted in inhibition of tumor growth, lower serum prostate specific antigen level and markedly prolonged survival, which was further enhanced by subsequent administration of paclitaxel. Immunohistochemistry revealed lower vessel density and enhanced tumor cell apoptosis in 225Ac-E4G10 treated tumors. Additionally, the residual tumor vasculature appeared normalized as evident by enhanced pericyte coverage following 225Ac-E4G10 therapy. However, no toxicity was observed in vascularized normal organs following 225Ac-E4G10 therapy. Conclusions The data suggest that alpha-particle immunotherapy to neovasculature, alone or in combination with sequential chemotherapy, is an effective approach to cancer therapy.
Collapse
Affiliation(s)
- Jaspreet Singh Jaggi
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Erik Henke
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Surya V. Seshan
- Department of Pathology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Barry J. Kappel
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Debjit Chattopadhyay
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Chad May
- ImClone Systems Incorporated, New York, New York, United States of America
| | - Michael R. McDevitt
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Daniel Nolan
- Cold Spring Harbor Laboratory, Woodbury, New York, United States of America
| | - Vivek Mittal
- Cold Spring Harbor Laboratory, Woodbury, New York, United States of America
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - David A. Scheinberg
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
250
|
Yordanov AT, Hens M, Pegram C, Bigner DD, Zalutsky MR. Antitenascin antibody 81C6 armed with 177Lu: in vivo comparison of macrocyclic and acyclic ligands. Nucl Med Biol 2007; 34:173-83. [PMID: 17307125 DOI: 10.1016/j.nucmedbio.2006.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 10/26/2006] [Accepted: 11/29/2006] [Indexed: 11/17/2022]
Abstract
INTRODUCTION When labeled with iodine-131, the antitenascin monoclonal antibody (mAb) 81C6 has shown promise as a targeted radiotherapeutic in patients with brain tumors. Because of its more favorable gamma-ray properties, lutetium-177 might be a better low-energy beta-emitter for this type of therapy. MATERIALS AND METHODS Chimeric 81C6 (ch81C6) was labeled with (177)Lu using the acyclic 1B4M ligand and the macrocyclic ligands NHS-DOTA and MeO-DOTA and evaluated for binding to tenascin. Three paired-label tissue distribution experiments were performed in normal mice receiving one of the (177)Lu-labeled immunoconjugates plus (125)I-labeled ch81C6 labeled using Iodogen. Paired-label experiments in athymic mice bearing subcutaneous D54 MG human glioma xenografts were done to directly compare the biodistribution of ch81C6-1B4M-(177)Lu and (125)I-labeled ch81C6, and ch81C6-MeO-DOTA-(177)Lu and (125)I-labeled ch81C6. Similar comparisons were done using murine (mu) instead of ch81C6. The primary parameter utilized for evaluation was the (177)Lu/(125)I uptake ratio in each tissue. RESULTS In the studies performed in normal mice, the NHS-DOTA ligand yielded the highest (177)Lu/(125)I uptake ratios in tissues indicative of loss of label from the chelate; for this reason, only 1B4M and MeO-DOTA were evaluated further. The (177)Lu/(125)I ratio in bone increased gradually with time for the chimeric conjugates; however, there were no significant differences between ch81C6-1B4M-DTPA-(177)Lu and ch81C6-MeO-DOTA-(177)Lu. In contrast, mu81C6-1B4M-DTPA-(177)Lu and mu81C6-MeO-DOTA-(177)Lu showed a more dramatic increase in the (177)Lu/(125)I ratio in bone - from 2.4+/-0.3 and 1.7+/-0.2 at Day 1 to 8.5+/-1.1 and 4.2+/-0.5 at Day 7, respectively. CONCLUSION With these antitenascin constructs, the nature of the mAb had a profound influence on the relative degree of loss of (177)Lu from these immunoconjugates. MeO-DOTA shows promise as a bifunctional chelate for labeling 81C6 mAbs with (177)Lu.
Collapse
|