1
|
Durand-Panteix S, Monteil J, Sage M, Garot A, Clavel M, Saidi A, Torgue J, Cogne M, Quelven I. Preclinical study of 212Pb alpha-radioimmunotherapy targeting CD20 in non-Hodgkin lymphoma. Br J Cancer 2021; 125:1657-1665. [PMID: 34671126 DOI: 10.1038/s41416-021-01585-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Despite therapeutic advances, Non-Hodgkin lymphoma (NHL) relapses can occur. The development of radioimmunotherapy (RIT) with α-emitters is an attractive alternative. In this study, we investigated the potential of α-RIT in conjunction with 212Pb-rituximab for the treatment of NHL. METHODS EL4-hCD20-Luc cells (mouse lymphoma cell line) were used for in vitro and in vivo studies. Biodistribution and efficacy studies were performed on C57BL/6 mice injected intravenously with 25 × 103 cells. RESULTS 212Pb-rituximab (0.925-7.4 kBq/mL) inhibit proliferation of EL4-hCD20-Luc cells in vitro. Biodistribution of 203/212Pb-rituximab in mice showed a significant tumour uptake and suggested that the liver, spleen, and kidneys were the organs at risk. For efficacy studies, mice were treated at either 11 days (early stage) or 20-30 days after injection of tumour cells (late stage). Treatment with 277.5 kBq 212Pb-rituximab significantly prolonged survival. Even at an advanced tumour stage, significant tumour regression occurred, with an increase in the median survival time to 28 days, compared with 9 days in the controls. CONCLUSIONS These results show the efficacy of 212Pb-rituximab in a murine syngeneic lymphoma model, in terms of significant tumour regression and increased survival, thereby highlighting the potency of α-RIT for the treatment of NHL.
Collapse
Affiliation(s)
- Stéphanie Durand-Panteix
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | - Jacques Monteil
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France.,Nuclear Medicine Department, Limoges University Hospital, Limoges, France
| | - Magali Sage
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | - Armand Garot
- Nuclear Medicine Department, Limoges University Hospital, Limoges, France
| | - Marie Clavel
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | | | | | - Michel Cogne
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France.
| | - Isabelle Quelven
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France. .,Nuclear Medicine Department, Limoges University Hospital, Limoges, France. .,ToNIC, Toulouse NeuroImaging Center - INSERM U1214, Toulouse, France.
| |
Collapse
|
2
|
Quelven I, Monteil J, Sage M, Saidi A, Mounier J, Bayout A, Garrier J, Cogne M, Durand-Panteix S. 212Pb α-Radioimmunotherapy Targeting CD38 in Multiple Myeloma: A Preclinical Study. J Nucl Med 2019; 61:1058-1065. [PMID: 31862796 PMCID: PMC7383085 DOI: 10.2967/jnumed.119.239491] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell cancer and represents the second most frequent hematologic malignancy. Despite new treatments and protocols, including high-dose chemotherapy associated with autologous stem cell transplantation, the prognosis of MM patients is still poor. α-radioimmunotherapy (α-RIT) represents an attractive treatment strategy because of the high-linear-energy transfer and short pathlength of α-radiation in tissues, resulting in high tumor cell killing and low toxicity to surrounding tissues. In this study, we investigated the potential of α-RIT with 212Pb-daratumumab (anti-hCD38), in both in vitro and in vivo models, as well as an antimouse CD38 antibody using in vivo models. Methods: Inhibition of cell proliferation after incubation of the RPMI8226 cell line with an increasing activity (0.185-3.7 kBq/mL) of 212Pb-isotypic control or 212Pb-daratumumab was evaluated. Biodistribution was performed in vivo by SPECT/CT imaging and after death. Dose-range-finding and acute toxicity studies were conducted. Because daratumumab does not bind the murine CD38, biodistribution and dose-range finding were also determined using an antimurine CD38 antibody. To evaluate the in vivo efficacy of 212Pb-daratumumab, mice were engrafted subcutaneously with 5 × 106 RPMI8226 cells. Mice were treated 13 d after engraftment with an intravenous injection of 212Pb-daratumumab or control solution. Therapeutic efficacy was monitored by tumor volume measurements and overall survival. Results: Significant inhibition of proliferation of the human myeloma RPMI8226 cell line was observed after 3 d of incubation with 212Pb-daratumumab, compared with 212Pb-isotypic control or cold antibodies. Biodistribution studies showed a specific tumoral accumulation of daratumumab. No toxicity was observed with 212Pb-daratumumab up to 370 kBq because of lack of cross-reactivity. Nevertheless, acute toxicity experiments with 212Pb-anti-mCD38 established a toxic activity of 277.5 kBq. To remain within realistically safe treatment activities for efficacy studies, mice were treated with 185 kBq or 277.5 kBq of 212Pb-daratumumab. Marked tumor growth inhibition compared with controls was observed, with a median survival of 55 d for 277.5 kBq of 212Pb-daratumumab instead of 11 d for phosphate-buffered saline. Conclusion: These results showed 212Pb-daratumumab to have efficacy in xenografted mice, with significant tumor regression and increased survival. This study highlights the potency of α-RIT in MM treatment.
Collapse
Affiliation(s)
- Isabelle Quelven
- Nuclear Medicine Department, Limoges University Hospital, Limoges, France.,CNRS-UMR7276, INSERM U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Limoges University, Limoges, France; and
| | - Jacques Monteil
- Nuclear Medicine Department, Limoges University Hospital, Limoges, France.,CNRS-UMR7276, INSERM U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Limoges University, Limoges, France; and
| | - Magali Sage
- CNRS-UMR7276, INSERM U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Limoges University, Limoges, France; and
| | | | - Jérémy Mounier
- CNRS-UMR7276, INSERM U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Limoges University, Limoges, France; and
| | - Audrey Bayout
- Nuclear Medicine Department, Limoges University Hospital, Limoges, France
| | - Julie Garrier
- CNRS-UMR7276, INSERM U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Limoges University, Limoges, France; and
| | - Michel Cogne
- CNRS-UMR7276, INSERM U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Limoges University, Limoges, France; and
| | - Stéphanie Durand-Panteix
- CNRS-UMR7276, INSERM U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Limoges University, Limoges, France; and
| |
Collapse
|
3
|
Besemer AE, Titz B, Grudzinski JJ, Weichert JP, Kuo JS, Robins HI, Hall LT, Bednarz BP. Impact of PET and MRI threshold-based tumor volume segmentation on patient-specific targeted radionuclide therapy dosimetry using CLR1404. Phys Med Biol 2017; 62:6008-6025. [PMID: 28682793 DOI: 10.1088/1361-6560/aa716d] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Variations in tumor volume segmentation methods in targeted radionuclide therapy (TRT) may lead to dosimetric uncertainties. This work investigates the impact of PET and MRI threshold-based tumor segmentation on TRT dosimetry in patients with primary and metastatic brain tumors. In this study, PET/CT images of five brain cancer patients were acquired at 6, 24, and 48 h post-injection of 124I-CLR1404. The tumor volume was segmented using two standardized uptake value (SUV) threshold levels, two tumor-to-background ratio (TBR) threshold levels, and a T1 Gadolinium-enhanced MRI threshold. The dice similarity coefficient (DSC), jaccard similarity coefficient (JSC), and overlap volume (OV) metrics were calculated to compare differences in the MRI and PET contours. The therapeutic 131I-CLR1404 voxel-level dose distribution was calculated from the 124I-CLR1404 activity distribution using RAPID, a Geant4 Monte Carlo internal dosimetry platform. The TBR, SUV, and MRI tumor volumes ranged from 2.3-63.9 cc, 0.1-34.7 cc, and 0.4-11.8 cc, respectively. The average ± standard deviation (range) was 0.19 ± 0.13 (0.01-0.51), 0.30 ± 0.17 (0.03-0.67), and 0.75 ± 0.29 (0.05-1.00) for the JSC, DSC, and OV, respectively. The DSC and JSC values were small and the OV values were large for both the MRI-SUV and MRI-TBR combinations because the regions of PET uptake were generally larger than the MRI enhancement. Notable differences in the tumor dose volume histograms were observed for each patient. The mean (standard deviation) 131I-CLR1404 tumor doses ranged from 0.28-1.75 Gy GBq-1 (0.07-0.37 Gy GBq-1). The ratio of maximum-to-minimum mean doses for each patient ranged from 1.4-2.0. The tumor volume and the interpretation of the tumor dose is highly sensitive to the imaging modality, PET enhancement metric, and threshold level used for tumor volume segmentation. The large variations in tumor doses clearly demonstrate the need for standard protocols for multimodality tumor segmentation in TRT dosimetry.
Collapse
Affiliation(s)
- Abigail E Besemer
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, United States of America. Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Nuclear model analysis of excitation functions of proton induced reactions on ⁸⁶Sr, ⁸⁸Sr and natZr: Evaluation of production routes of ⁸⁶Y. Appl Radiat Isot 2015. [PMID: 26210800 DOI: 10.1016/j.apradiso.2015.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The proton induced nuclear reactions on (86)Sr, (88)Sr and (nat)Zr were investigated for the production of (86)Y. The literature data were compared with the results of nuclear model calculations using the codes ALICE-IPPE, TALYS 1.6 and EMPIRE 3.2. The thick target yields of (86)Y were calculated from the recommended excitation functions. Analysis of radioyttrium impurities was also performed. A comparison of the various production routes showed that for medical applications of (86)Y, the reaction (86)Sr(p,n)(86)Y is the method of choice, which gives efficient yield with minimum impurities.
Collapse
|
5
|
Goel A, Spitz DR, Weiner GJ. Manipulation of cellular redox parameters for improving therapeutic responses in B-cell lymphoma and multiple myeloma. J Cell Biochem 2012; 113:419-25. [PMID: 21956712 DOI: 10.1002/jcb.23387] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Developing novel combined-modality therapeutic approaches based on understanding of the involvement of redox biology in apoptosis of malignant cells is a promising approach for improving clinical responses in B-cell lymphoma and multiple myeloma. Therapeutic modalities that generate reactive oxygen species (i.e., radiation, photodynamic therapy, and specific chemotherapeutic drugs) have been shown to be selectively cytotoxic to malignant B-cells. In this review, we will discuss agents that induce apoptosis in B-cell tumors by oxidative stress. Subsequently, a novel biochemical rationale (based on fundamental differences in cancer vs. normal cell oxidative metabolism) for combining oxidative stressors with radiotherapy and chemotherapy, that may lead to designing of more effective treatment strategies for B-cell malignancies, will be discussed. Besides providing potential curative benefit, such novel therapies could also selectively target and inhibit the emergence of drug-resistance in tumor cells, which is a major determinant of treatment failure in many B-cell malignancies.
Collapse
Affiliation(s)
- Apollina Goel
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | |
Collapse
|
6
|
Kreitman RJ, Pastan I. Antibody fusion proteins: anti-CD22 recombinant immunotoxin moxetumomab pasudotox. Clin Cancer Res 2011; 17:6398-405. [PMID: 22003067 PMCID: PMC3201735 DOI: 10.1158/1078-0432.ccr-11-0487] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recombinant immunotoxins are fusion proteins that contain the cytotoxic portion of a protein toxin fused to the Fv portion of an antibody. The Fv binds to an antigen on a target cell and brings the toxin into the cell interior, where it arrests protein synthesis and initiates the apoptotic cascade. Moxetumomab pasudotox, previously called HA22 or CAT-8015, is a recombinant immunotoxin composed of the Fv fragment of an anti-CD22 monoclonal antibody fused to a 38-kDa fragment of Pseudomonas exotoxin A, called PE38. Moxetumomab pasudotox is an improved, more active form of a predecessor recombinant immunotoxin, BL22 (also called CAT-3888), which produced complete remission in relapsed/refractory hairy cell leukemia (HCL), but it had a <20% response rate in chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia (ALL), diseases in which the leukemic cells contain much lower numbers of CD22 target sites. Compared with BL22, moxetumomab pasudotox is up to 50-fold more active on lymphoma cell lines and leukemic cells from patients with CLL and HCL. A phase I trial was recently completed in HCL patients, who achieved response rates similar to those obtained with BL22 but without dose-limiting toxicity. In addition to further testing in HCL, moxetumomab pasudotox is being evaluated in phase I trials in patients with CLL, B-cell lymphomas, and childhood ALL. Moreover, protein engineering is being used to increase its activity, decrease nonspecific side effects, and remove B-cell epitopes.
Collapse
Affiliation(s)
- Robert J Kreitman
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, Maryland, USA.
| | | |
Collapse
|
7
|
Goldenberg DM. Radiolabelled monoclonal antibodies in the treatment of metastatic cancer. ACTA ACUST UNITED AC 2010; 14:39-42. [PMID: 17576463 PMCID: PMC1891189 DOI: 10.3747/co.2007.112] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- D M Goldenberg
- GardenState Cancer Center, Belleville, New Jersey, U.S.A.
| |
Collapse
|
8
|
Porrata LF, Markovic SN. Is Absolute Lymphocyte Count Just Another Prognostic Factor in Cancer? ACTA ACUST UNITED AC 2010. [DOI: 10.3814/2010/812304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
9
|
Jain N, Wierda W, Ferrajoli A, Wong F, Lerner S, Keating M, O'Brien S. A phase 2 study of yttrium-90 ibritumomab tiuxetan (Zevalin) in patients with chronic lymphocytic leukemia. Cancer 2009; 115:4533-9. [PMID: 19637351 DOI: 10.1002/cncr.24522] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Radioimmunotherapy (RIT) with radio-labeled monoclonal antibodies to CD20 produce a high response rate in patients with relapsed lymphoma. Use of this modality in patients with chronic lymphocytic leukemia (CLL) has been hampered by the extensive marrow involvement seen in patients with CLL, which would produce a high risk for marrow aplasia after treatment with RIT. Patients with lymphoma and marrow involvement have been treated with RIT if involved marrow was less than 25% of the total marrow. Thus, we adapted this approach as consolidation therapy in patients with CLL responding to chemoimmunotherapy. METHODS Fourteen patients with relapsed CLL either in partial remission or in complete remission but with disease documented by flow cytometry were treated with (90)Y ibritumomab tiuxetan. RESULTS One patient responded and achieved a complete remission but with residual disease detected by flow cytometry. Of note was that grade 3 or 4 hematologic toxicity was seen in 12 of the 13 (92%) evaluable patients, with grade 3 or 4 thrombocytopenia noted in 11 (85%) of the patients. In addition, myelosuppression was prolonged with a median duration of grade 3 or 4 thrombocytopenia of 37 days. Five patients had persistent thrombocytopenia 3 months post-therapy. CONCLUSIONS Even in patients with CLL and limited marrow involvement, the use of RIT results in unacceptable hematologic toxicity.
Collapse
Affiliation(s)
- Nitin Jain
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Shimoni A, Nagler A. Radioimmunotherapy and stem-cell transplantation in the treatment of aggressive B-cell lymphoma. Leuk Lymphoma 2009; 48:2110-20. [PMID: 17891639 DOI: 10.1080/10428190701573273] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
High-dose chemotherapy and autologous stem-cell transplantation (ASCT) have an established therapeutic role in the treatment of chemo-sensitive relapsed aggressive lymphoma, but has limited success in chemo-refractory disease or in heavily pretreated, multiple relapsed patients. Recurrent disease is the major cause of treatment failure in all patient subsets and the majority is not cured. Methods for better eradication of underlying lymphoma are needed to improve outcome. Radioimmunotherapy (RIT) combines radiation delivered by radio-isotopes with the targeting effect of monoclonal antibodies. Two radioimmunoconjugates are currently approved for relapsed/resistant low-grade or transformed lymphoma: iodine-131 tositumomab and yttrium-90 ibritumomab tiuxetan. These agents are also effective in aggressive lymphoma. Radio-labeled antibodies are ideal candidates to combine with high-dose chemotherapy and ASCT. Their major toxicity is myelosuppression, which is easily reversed by ASCT and they can target disease sites with almost no added toxicity to normal tissues. RIT has been used in escalated doses as the sole treatment prior to ASCT or in standard or escalated doses combined with standard high-dose chemotherapy regimens. RIT was also combined with reduced-intensity conditioning and allogeneic SCT. Preliminary results are promising and suggest improved disease control with no added toxicity; however randomized studies are needed to confirm these initial observations.
Collapse
Affiliation(s)
- Avichai Shimoni
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Israel.
| | | |
Collapse
|
11
|
Abstract
Nanotechnology offers many opportunities for enhanced diagnostic and therapeutic medicine against cancer and other diseases. In this review, the special properties that result from the nanoscale size of quantum dots, metal colloids, superparamagnetic iron oxide, and carbon-based nanostructures are reviewed and interpreted against a background of the structural and electronic detail that gives rise to their nanotechnologic behavior. The detection and treatment of cancer is emphasized, with special attention paid to the biologic targeting of the disease. The future of nanotechnology in cancer research and clinical practice is projected to focus on 'theranostic' nanoparticles that are both diagnostic and therapeutic by design.
Collapse
|
12
|
Kapoor P, Greipp PT, Morice WG, Rajkumar SV, Witzig TE, Greipp PR. Anti-CD20 monoclonal antibody therapy in multiple myeloma. Br J Haematol 2008; 141:135-48. [PMID: 18318769 DOI: 10.1111/j.1365-2141.2008.07024.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
CD20 is a particularly appealing target that is expressed on the surface of almost all B cells, with no significant shedding, secretion or internalization. In contrast to the demonstrated efficacy of anti-CD20 strategies in various B-cell lymphoproliferative disorders, the role of such therapy in multiple myeloma is undetermined and controversial. The expression of CD20 by myeloma cells is heterogeneous, and can be detected only in 13-22% of patients. However, there is increasing interest in testing anti-CD20 therapy in myeloma because of recent studies suggesting the existence of clonogenic CD20-positive precursor B cells in the disease. This article reviews the rationale, preclinical and clinical activity of anti-CD20 therapy in myeloma. Clinical trials show that anti-CD20 therapy with rituximab elicits a partial response in approximately 10% of CD20+ patients with multiple myeloma. In addition, there is preliminary evidence of disease stabilization in 50-57% of CD20+ patients for a period of 10-27 months. Further large-scale clinical trials are therefore needed to establish the role of this promising strategy in the treatment of myeloma.
Collapse
Affiliation(s)
- Prashant Kapoor
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
High-dose therapy (HDT) followed by autologous transplantation of hematopoietic stem cells (ASCT) is frequently performed in patients with lymphoma. For many subentities, reliable results from prospective randomized studies are missing. In Hodgkin's disease (HD), HDT/ASCT is a standard indication for patients with chemosensitive first relapse. Patients with indolent or aggressive B-cell lymphoma may benefit from HDT/ASCT if considered as part of first-line therapy or at the time of relapse. However, new randomized studies comparing HDT/ASCT with optimal chemoimmunotherapy are necessary because the introduction of monoclonal antibodies (rituximab) significantly improved the results of conventional chemotherapy. Because data on patients with less frequent entities like mantle cell lymphoma, T-cell lymphoma, Burkitt's lymphoma, or lymphoblastic lymphoma are insufficient, the role of HDT/ASCT needs further study.
Collapse
Affiliation(s)
- Norbert Schmitz
- Department of Hematology and Stem Cell Transplantation, ASKLEPIOS Klinik St. Georg, Hamburg, Germany.
| | | | | |
Collapse
|
14
|
Zinzani PL, d’Amore F, Bombardieri E, Brammer C, Codina JG, Illidge T, Jurczak W, Linkesch W, Morschhauser F, Vandenberghe E, Van Hoof A. Consensus conference: Implementing treatment recommendations on yttrium-90 immunotherapy in clinical practice – Report of a European workshop. Eur J Cancer 2008; 44:366-73. [DOI: 10.1016/j.ejca.2007.12.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Accepted: 12/17/2007] [Indexed: 11/27/2022]
|
15
|
Gisselbrecht C, Bethge W, Duarte RF, Gianni AM, Glass B, Haioun C, Martinelli G, Nagler A, Pettengell R, Sureda A, Tilly H, Wilson K. Current status and future perspectives for yttrium-90 ((90)Y)-ibritumomab tiuxetan in stem cell transplantation for non-Hodgkin's lymphoma. Bone Marrow Transplant 2007; 40:1007-17. [PMID: 17922042 DOI: 10.1038/sj.bmt.1705868] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Haematopoietic SCT is currently considered a therapeutic option mainly in relapsed or refractory non-Hodgkin's lymphoma (NHL) owing to high post-transplantation relapse rates and significant toxicity of conventional myeloablative conditioning for allogeneic SCT. Radiolabelled immunotherapy combines the benefits of monoclonal antibody targeting with therapeutic doses of radiation, and is a promising advance in the treatment of malignant lymphomas. It is now under investigation as a component of conditioning prior to SCT, with the aim of improving outcomes following SCT without increasing the toxicity of high-dose chemotherapy pre-transplant conditioning. An expert panel met at a European workshop in November 2006 to review the latest data on radiolabelled immunotherapy in the transplant setting, and its potential future directions, with a focus on (90)Y-ibritumomab tiuxetan. They reviewed data on the combination of standard/high/escalating dose (90)Y-ibritumomab tiuxetan with high-dose chemotherapy, and high/escalating dose (90)Y-ibritumomab tiuxetan as the sole myeloablative agent, prior to autologous SCT, and also (90)Y-ibritumomab tiuxetan as a component of reduced intensity conditioning prior to allogeneic SCT. The preliminary data are highly promising in terms of conditioning tolerability and patient outcomes following transplant; further phase II studies are now needed to consolidate these data and to investigate specific patient populations and NHL subtypes.
Collapse
Affiliation(s)
- C Gisselbrecht
- Institut d'Hématologie, Hôpital Saint-Louis, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Friedberg JW. Overcoming Rituximab Resistance: The US Clinical Experience With Bendamustine. Semin Hematol 2007. [DOI: 10.1053/j.seminhematol.2007.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
Witzig TE. Standard Approaches to Relapsed Indolent Non-Hodgkin’s Lymphoma. Semin Hematol 2007. [DOI: 10.1053/j.seminhematol.2007.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
18
|
Porrata LF, Ristow K, Witzig TE, Tuinistra N, Habermann TM, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Markovic SN. Absolute lymphocyte count predicts therapeutic efficacy and survival at the time of radioimmunotherapy in patients with relapsed follicular lymphomas. Leukemia 2007; 21:2554-6. [PMID: 17581607 DOI: 10.1038/sj.leu.2404819] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
19
|
John Wiley & Sons, Ltd.. Current Awareness in Hematological Oncology. Hematol Oncol 2007. [DOI: 10.1002/hon.796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
20
|
Abstract
This article reviews the development of radioimmunoconjugates as a new class of cancer therapeutics. Numerous conjugates involving different antigen targets, antibody forms, radionuclides and methods of radiochemistry have been studied in the half-century since radioactive antibodies were first used in model systems to selectively target radiation to tumors. Whereas directly conjugated antibodies, fragments and subfragments have shown promise preclinically, the same approaches have not gained success in patients except in radiosensitive hematological neoplasms, or in settings involving minimal or locoregional disease. The separation of tumor targeting from the delivery of the therapeutic radionuclide in a multistep process called pretargeting has the potential to overcome many of the limitations of conventional, or one-step, radioimmunotherapy, with initial preclinical and clinical data showing increased sensitivity, specificity and higher radiation doses delivered. Our particular focus in pretargeting is the use of bispecific, trimeric (three Fab's) constructs made by a new antibody engineering method termed 'dock-and-lock.
Collapse
Affiliation(s)
- D M Goldenberg
- Garden State Cancer Center, Center for Molecular Medicine and Immunology, Belleville, NJ 07109, USA.
| | | |
Collapse
|
21
|
Goldenberg DM, Chatal JF, Barbet J, Boerman O, Sharkey RM. Cancer Imaging and Therapy with Bispecific Antibody Pretargeting. UPDATE ON CANCER THERAPEUTICS 2007; 2:19-31. [PMID: 18311322 PMCID: PMC2034280 DOI: 10.1016/j.uct.2007.04.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This article reviews recent preclinical and clinical advances in the use of pretargeting methods for the radioimmunodetection and radioimmunotherapy of cancer. Whereas directly-labeled antibodies, fragments, and subfragments (minibodies and other constructs) have shown promise in both imaging and therapy applications over the past 25 years, their clinical adoption has not fulfilled the original expectations due to either poor image resolution and contrast in scanning or insufficient radiation doses delivered selectively to tumors for therapy. Pretargeting involves the separation of the localization of tumor with an anticancer antibody from the subsequent delivery of the imaging or therapeutic radionuclide. This has shown improvements in both imaging and therapy by overcoming the limitations of conventional, or 1-step, radioimmunodetection or radioimmunotherapy. We focus herein on the use of bispecific antibodies followed by radiolabeled peptide haptens as a new modality of selective delivery of radionuclides for the imaging and therapy of cancer. Our particular emphasis in pretargeting is the use of bispecific trimeric (3 Fab's) recombinant constructs made by a modular method of antibody and protein engineering of fusion molecules called Dock and Lock (DNL).
Collapse
Affiliation(s)
- David M Goldenberg
- Garden State Cancer Center, Center for Molecular Medicine and Immunology, Belleville, NJ, USA
| | | | | | | | | |
Collapse
|