51
|
Anti-CD45 pretargeted radioimmunotherapy using bismuth-213: high rates of complete remission and long-term survival in a mouse myeloid leukemia xenograft model. Blood 2011; 118:703-11. [PMID: 21613259 DOI: 10.1182/blood-2011-04-347039] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pretargeted radioimmunotherapy (PRIT) using an anti-CD45 antibody (Ab)-streptavidin (SA) conjugate and DOTA-biotin labeled with β-emitting radionuclides has been explored as a strategy to decrease relapse and toxicity. α-emitting radionuclides exhibit high cytotoxicity coupled with a short path length, potentially increasing the therapeutic index and making them an attractive alternative to β-emitting radionuclides for patients with acute myeloid leukemia. Accordingly, we have used (213)Bi in mice with human leukemia xenografts. Results demonstrated excellent localization of (213)Bi-DOTA-biotin to tumors with minimal uptake into normal organs. After 10 minutes, 4.5% ± 1.1% of the injected dose of (213)Bi was delivered per gram of tumor. α-imaging demonstrated uniform radionuclide distribution within tumor tissue 45 minutes after (213)Bi-DOTA-biotin injection. Radiation absorbed doses were similar to those observed using a β-emitting radionuclide ((90)Y) in the same model. We conducted therapy experiments in a xenograft model using a single-dose of (213)Bi-DOTA-biotin given 24 hours after anti-CD45 Ab-SA conjugate. Among mice treated with anti-CD45 Ab-SA conjugate followed by 800 μCi of (213)Bi- or (90)Y-DOTA-biotin, 80% and 20%, respectively, survived leukemia-free for more than 100 days with minimal toxicity. These data suggest that anti-CD45 PRIT using an α-emitting radionuclide may be highly effective and minimally toxic for treatment of acute myeloid leukemia.
Collapse
|
52
|
Walter RB, Press OW, Pagel JM. Pretargeted radioimmunotherapy for hematologic and other malignancies. Cancer Biother Radiopharm 2010; 25:125-42. [PMID: 20423225 DOI: 10.1089/cbr.2010.0759] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Radioimmunotherapy (RIT) has emerged as one of the most promising treatment options, particularly for hematologic malignancies. However, this approach has generally been limited by a suboptimal therapeutic index (target-to-nontarget ratio) and an inability to deliver sufficient radiation doses to tumors selectively. Pretargeted RIT (PRIT) circumvents these limitations by separating the targeting vehicle from the subsequently administered therapeutic radioisotope, which binds to the tumor-localized antibody or is quickly excreted if unbound. A growing number of preclinical proof-of-principle studies demonstrate that PRIT is feasible and safe and provides improved directed radionuclide delivery to malignant cells compared with conventional RIT while sparing normal cells from nonspecific radiotoxicity. Early phase clinical studies corroborate these preclinical findings and suggest better efficacy and lesser toxicities in patients with hematologic and other malignancies. With continued research, PRIT-based treatment strategies promise to become cornerstones to improved outcomes for cancer patients despite their complexities.
Collapse
Affiliation(s)
- Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.
| | | | | |
Collapse
|
53
|
Alyea EP, DeAngelo DJ, Moldrem J, Pagel JM, Przepiorka D, Sadelin M, Young JW, Giralt S, Bishop M, Riddell S. NCI First International Workshop on The Biology, Prevention and Treatment of Relapse after Allogeneic Hematopoietic Cell Transplantation: report from the committee on prevention of relapse following allogeneic cell transplantation for hematologic malignancies. Biol Blood Marrow Transplant 2010; 16:1037-69. [PMID: 20580849 PMCID: PMC3235046 DOI: 10.1016/j.bbmt.2010.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Accepted: 05/14/2010] [Indexed: 10/19/2022]
Abstract
Prevention of relapse after allogeneic hematopoietic stem cell transplantation is the most likely approach to improve survival of patients treated for hematologic malignancies. Herein we review the limits of currently available transplant therapies and the innovative strategies being developed to overcome resistance to therapy or to fill therapeutic modalities not currently available. These novel strategies include nonimmunologic therapies, such as targeted preparative regimens and posttransplant drug therapy, as well as immunologic interventions, including graft engineering, donor lymphocyte infusions, T cell engineering, vaccination, and dendritic cell-based approaches. Several aspects of the biology of the malignant cells as well as the host have been identified that obviate success of even these newer strategies. To maximize the potential for success, we recommend pursuing research to develop additional targeted therapies to be used in the preparative regimen or as maintenance posttransplant, better characterize the T cell and dendritic cells subsets involved in graft-versus-host disease and the graft-versus-leukemia/tumor effect, identify strategies for timing immunologic or nonimmunologic therapies to eliminate the noncycling cancer stem cell, identify more targets for immunotherapies, develop new vaccines that will not be limited by HLA, and develop methods to identify populations at very high risk for relapse to accelerate clinical development and avoid toxicity in patients not at risk for relapse.
Collapse
Affiliation(s)
- Edwin P Alyea
- Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Pilot study of a (213)bismuth-labeled anti-CD45 mAb as a novel nonmyeloablative conditioning for DLA-haploidentical littermate hematopoietic transplantation. Transplantation 2010; 89:1336-40. [PMID: 20351629 DOI: 10.1097/tp.0b013e3181d98c3d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND A pilot study was conducted to determine whether conditioning using selective targeting of hematopoietic cells with an alpha-particle emitter, bismuth-213 ((213)Bi)-labeled anti-CD45 monoclonal antibody (mAb) is sufficient to overcome the major histocompatibility barrier in a canine model of dog leukocyte antigen-haploidentical hematopoietic cell transplantation (HCT). METHODS Six dogs were administered 0.5 mg/kg (213)Bi-labeled anti-CD45 mAb (dose (213)Bi=2.26-4.9 mCi/kg) in six to eight injections. For postgrafting immunosuppression, all dogs received cyclosporine and mycophenolate mofetil. RESULTS All dogs had initial donor engraftment, with three of six dogs having sustained engraftment to last point of follow-up. Two dogs receiving 2.26 and 3.25 mCi/kg of (213)Bi rejected their grafts at day +127 and +125, respectively, whereas dogs receiving (213)Bi doses of 3.3 mCi/kg or greater achieved high level donor chimerism. CONCLUSION The results suggest that nonmyeloablative conditioning with (213)Bi-labeled anti-CD45 mAb could be applicable to major histocompatibility haploidentical HCT without excessive nonhematologic regimen-related toxicity.
Collapse
|
55
|
Appelbaum FR. Optimising the conditioning regimen for acute myeloid leukaemia. Best Pract Res Clin Haematol 2010; 22:543-50. [PMID: 19959106 DOI: 10.1016/j.beha.2009.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The conditioning regimen administered prior to allogeneic transplantation for acute myeloid leukaemia (AML) must be sufficiently immunosuppressive to ensure engraftment, and contributes to the anti-leukaemic impact of the procedure. A broad spectrum of regimens have been studied, varying in their intensity, whether high-dose or reduced intensity, and in the agents used, containing total body irradiation (TBI) plus cyclophosphamide, fludarabine, busulfan and/or anti-thymocyte globulin. Over the past 2 decades, research has influenced the way conditioning regimens are applied. Newer research shows that targeted radiotherapy using an anti-CD45 antibody should be able to reduce toxicity, improve tumour cell kill and thereby improve results.
Collapse
|
56
|
Recent advances in novel radioimmunotherapeutic approaches for allogeneic hematopoietic cell transplantation. Curr Opin Oncol 2010; 22:143-9. [DOI: 10.1097/cco.0b013e328335ccc2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
57
|
Forman SJ. Allogeneic Hematopoietic Cell Transplantation for Acute Lymphoblastic Leukemia in Adults. Hematol Oncol Clin North Am 2009; 23:1011-31, vi. [DOI: 10.1016/j.hoc.2009.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
58
|
Abstract
Over the past decade, monoclonal antibodies have dramatically impacted the treatment of haematological malignancies, as evidenced by the effect of rituximab on the response rate and survival of patients with follicular and diffuse large B cell non-Hodgkin's lymphoma. Currently, only two monoclonal antibodies - the anti-CD33 immunotoxin gemtuzumab ozogamicin and the CD52-directed antibody alemtuzumab - are approved for treatment of relapsed acute myeloid leukaemia in older patients and B cell chronic lymphocytic leukaemia, respectively. Although not approved for such treatment, alemtuzumab is also active against T cell prolymphocytic leukaemia, cutaneous T cell lymphoma and Sézary syndrome, and adult T cell leukaemia and lymphoma. In addition, rituximab has demonstrated activity against B cell chronic lymphocytic and hairy cell leukaemia. Monoclonal antibodies targeting CD4, CD19, CD20, CD22, CD23, CD25, CD45, CD66 and CD122 are now being studied in the clinic for the treatment of leukaemia. Here, we discuss how these new antibodies have been engineered to reduce immunogenicity and improve antibody targeting and binding. Improved interactions with Fc receptors on immune effector cells can enhance destruction of target cells through antibody-dependent cellular cytotoxicity and complement-mediated cell lysis. The antibodies can also be armed with cellular toxins or radionuclides to enhance the destruction of leukaemia cells.
Collapse
Affiliation(s)
- John C Morris
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1457, USA.
| | | |
Collapse
|
59
|
Allogeneic hematopoietic cell transplantation after conditioning with 131I-anti-CD45 antibody plus fludarabine and low-dose total body irradiation for elderly patients with advanced acute myeloid leukemia or high-risk myelodysplastic syndrome. Blood 2009; 114:5444-53. [PMID: 19786617 DOI: 10.1182/blood-2009-03-213298] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We conducted a study to estimate the maximum tolerated dose (MTD) of (131)I-anti-CD45 antibody (Ab; BC8) that can be combined with a standard reduced-intensity conditioning regimen before allogeneic hematopoietic cell transplantation. Fifty-eight patients older than 50 years with advanced acute myeloid leukemia (AML) or high-risk myelodysplastic syndrome (MDS) were treated with (131)I-BC8 Ab and fludarabine plus 2 Gy total body irradiation. Eighty-six percent of patients had AML or MDS with greater than 5% marrow blasts at the time of transplantation. Treatment produced a complete remission in all patients, and all had 100% donor-derived CD3(+) and CD33(+) cells in the blood by day 28 after the transplantation. The MTD of (131)I-BC8 Ab delivered to liver was estimated to be 24 Gy. Seven patients (12%) died of nonrelapse causes by day 100. The estimated probability of recurrent malignancy at 1 year is 40%, and the 1-year survival estimate is 41%. These results show that CD45-targeted radiotherapy can be safely combined with a reduced-intensity conditioning regimen to yield encouraging overall survival for older, high-risk patients with AML or MDS. This study was registered at www.clinicaltrials.gov as #NCT00008177.
Collapse
|
60
|
Straathof KC, Rao K, Eyrich M, Hale G, Bird P, Berrie E, Brown L, Adams S, Schlegel PG, Goulden N, Gaspar HB, Gennery AR, Landais P, Davies EG, Brenner MK, Veys PA, Amrolia PJ. Haemopoietic stem-cell transplantation with antibody-based minimal-intensity conditioning: a phase 1/2 study. Lancet 2009; 374:912-20. [PMID: 19729196 DOI: 10.1016/s0140-6736(09)60945-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Stem-cell transplantation can cure primary immunodeficiencies. However, in patients with pre-existing organ toxicity, patients younger than 1 year, and those with DNA or telomere repair disorders, chemotherapy-based conditioning is poorly tolerated and results in major morbidity and mortality. We tested a novel antibody-based minimal-intensity conditioning (MIC) regimen to assess whether this approach allowed curative donor stem-cell engraftment without non-haemopoietic toxicity. METHODS 16 high-risk patients underwent stem-cell transplantation for primary immunodeficiencies with an MIC regimen consisting of two rat anti-CD45 monoclonal antibodies YTH 24.5 and YTH 54.12 for myelosuppression, and alemtuzumab (anti-CD52) and fludarabine, and low dose cyclophosphamide for immunosuppression. Donors were matched siblings (n=5), and matched (9) and mismatched (2) unrelated donors. FINDINGS Antibody-based conditioning was well tolerated, with only two cases of grade 3 and no grade 4 toxicity. Rates of clinically significant acute (n=6, 36%) and chronic graft-versus-host disease (GVHD) (n=5, 31%) were acceptable. 15 of 16 patients (94%) engrafted, of whom 11 (69%) achieved full or high-level mixed chimerism in both lymphoid and myeloid lineages, and three achieved engraftment in the T-lymphoid lineage only. One patient needed retransplantation. At a median of 40 months post-transplant, 13 of 16 patients (81%) in this high-risk cohort were alive and cured from their underlying disease. INTERPRETATION Monoclonal antibody-based conditioning seems well tolerated and can achieve curative engraftment even in patients with severe organ toxicity or DNA repair defects, or both. This novel approach represents a shift from the paradigm that intensive chemotherapy or radiotherapy, or both, is needed for donor stem-cell engraftment. This antibody-based conditioning regimen may reduce toxicity and late effects and enable SCT in virtually any primary immunodeficiency patient with a matched donor. FUNDING None.
Collapse
|
61
|
Dahlke MH, Larsen SR, Rasko JEJ, Schlitt HJ. The Biology of CD45 and its Use as a Therapeutic Target. Leuk Lymphoma 2009; 45:229-36. [PMID: 15101706 DOI: 10.1080/1042819031000151932] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
All mature hemopoietic lineage cells, with exclusion of platelets and mature erythrocytes, share the surface expression of a transmembrane phosphatase, the CD45 molecule. It is also present on hemopoietic stem cells and most leukemic clones and therefore presents as an appropriate target for immunotherapy with anti-CD45 antibodies. This short review details the biology of CD45 and its recent targeting for both treatment of malignant disorders and tolerance induction. In particular, the question of potential stem cell depletion for induction of central tolerance or depletion of malignant hemopoietic cells is addressed. Mechanisms underlying the effects downstream of CD45 binding to the cell surface are discussed.
Collapse
Affiliation(s)
- Marc H Dahlke
- Centenary Institute of Cancer Medicine and Cell Biology & University of Sydney, Sydney, Australia.
| | | | | | | |
Collapse
|
62
|
Rodriguez V, Anderson PM, Litzow MR, Erlandson L, Trotz BA, Arndt CAS, Khan SP, Wiseman GA. Marrow irradiation with high-dose 153Samarium-EDTMP followed by chemotherapy and hematopoietic stem cell infusion for acute myelogenous leukemia. Leuk Lymphoma 2009; 47:1583-92. [PMID: 16966270 DOI: 10.1080/10428190600580817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In four patients, aged 15 - 20 years, with high-risk acute myeloid leukemia (AML), high-dose samarium 153-labelled ethylenediaminetetramethylenephosphonate (153Sm-EDTMP) was used for targeted marrow irradiation before preparative chemotherapy conditioning regimens and allogeneic (three patients) or autologous (one patient) hematopoietic stem cell transplantation. The dose of 153Sm-EDTMP was 703 MBq/kg (n = 1) or 1110 MBq/kg (n = 3). No side-effects occurred during the 30-min infusion of 153Sm-EDTMP. Samarium - melphalan regimens were given to three patients; one had 153Sm-EDTMP - busulfan + cyclophosphamide. Total body radioactivity was below the 133 MBq safe limit before infusion of stem cells (day 14 after 153Sm-EDTMP). No hemorrhagic cystitis, nephrotoxicity or serious infections occurred. Leukocyte engraftment (white blood cell count >0.5 x 10(9)/l) occurred between 12 and 23 days after stem cell infusion (mean of 17 days). Complete cytogenetic and morphologic remission of AML was evident on follow-up marrow aspirate and biopsy specimens from all patients. In two of the four study patients, the disease remains in complete remission and the patients have an excellent quality of life (Eastern Cooperative Oncology Group performance status 0; no medications) and no organ toxicity more than 2 years and more than 4 years, respectively, after their blood and bone marrow transplantations. Thus, in adolescents and adults, 153Sm-EDTMP may provide a relatively simple and effective means for using irradiation to eliminate AML within the marrow.
Collapse
Affiliation(s)
- Vilmarie Rodriguez
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Pretargeting CD45 enhances the selective delivery of radiation to hematolymphoid tissues in nonhuman primates. Blood 2009; 114:1226-35. [PMID: 19515724 DOI: 10.1182/blood-2009-03-210344] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pretargeted radioimmunotherapy (PRIT) is designed to enhance the directed delivery of radionuclides to malignant cells. Through a series of studies in 19 nonhuman primates (Macaca fascicularis), the potential therapeutic advantage of anti-CD45 PRIT was evaluated. Anti-CD45 PRIT demonstrated a significant improvement in target-to-normal organ ratios of absorbed radiation compared with directly radiolabeled bivalent antibody (conventional radioimmunotherapy [RIT]). Radio-DOTA-biotin administered 48 hours after anti-CD45 streptavidin fusion protein (FP) [BC8 (scFv)(4)SA] produced markedly lower concentrations of radiation in nontarget tissues compared with conventional RIT. PRIT generated superior target:normal organ ratios in the blood, lung, and liver (10.3:1, 18.9:1, and 9.9:1, respectively) compared with the conventional RIT controls (2.6:1, 6.4:1, and 2.9:1, respectively). The FP demonstrated superior retention in target tissues relative to comparable directly radiolabeled bivalent anti-CD45 RIT. The time point of administration of the second step radiolabeled ligand (radio-DOTA-biotin) significantly impacted the biodistribution of radioactivity in target tissues. Rapid clearance of the FP from the circulation rendered unnecessary the addition of a synthetic clearing agent in this model. These results support proceeding to anti-CD45 PRIT clinical trials for patients with both leukemia and lymphoma.
Collapse
|
64
|
Jones CV, Copelan EA. Treatment of acute myeloid leukemia with hematopoietic stem cell transplantation. Future Oncol 2009; 5:559-68. [DOI: 10.2217/fon.09.20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation provides the most powerful antileukemic effect in the treatment of acute myeloid leukemia. Due to its significant morbidity and mortality, it should be used in first remission patients whose relapse risk is substantial. Reduced intensity transplantation is safer and extends the application of early transplantation to older patients and those with comorbidities. In patients with advanced disease, allotransplantation provides a lower chance for cure, but is often the only curative treatment available. Advances in histocompatibility typing and supportive care have improved results of allogeneic transplantation in acute myeloid leukemia.
Collapse
Affiliation(s)
- Cortney V Jones
- Department of Hematologic Oncology & Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Ave, R35, Cleveland, OH 44195, USA
| | - Edward A Copelan
- Director, Acute Leukemia Program, Department of Hematologic Oncology & Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Ave, R35, Cleveland, OH 44195, USA
| |
Collapse
|
65
|
Lin T, Jones RJ, Matsui W. Cancer stem cells: relevance to SCT. Bone Marrow Transplant 2009; 43:517-23. [PMID: 19234507 PMCID: PMC2950943 DOI: 10.1038/bmt.2009.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Accepted: 12/31/2008] [Indexed: 12/26/2022]
Abstract
The cancer stem cell (CSC) hypothesis suggests that clonogenic growth potential within an individual tumor is restricted to a specific and phenotypically defined cell population. Evidence for CSC in human tumors initially arose from studies of AML, but functionally similar cell populations have been identified in an increasing number of malignancies. Despite these findings, controversy surrounds the CSC hypothesis, especially the generalization that clonogenic tumor cells are rare. Nevertheless, efforts to define the cellular processes regulating self-renewal and resistance to anticancer therapeutics, two of the major properties ascribed to CSC, are likely to provide useful insights into tumor biology as a whole. BMT has been at the forefront of clinically translating basic stem cell concepts starting with the original hypothesis that normal hematopoietic precursors could rescue patients from myeloablative doses of radiation or chemotherapy. Even today, a better understanding of CSC may enhance ongoing efforts to induce specific and effective anti-tumor immune responses in both the allogeneic and autologous setting. It is also likely that new clinical research approaches will be required to accurately evaluate novel CSC-targeting strategies. Owing to the capacity to produce remissions in most diseases, SCT may provide the ideal clinical platform to carry out these investigations by studying the ability of anti-CSC agents to prolong relapse free and overall survival.
Collapse
Affiliation(s)
- T Lin
- Section of Hematology and Oncology, Department of Internal Medicine, LSU School of Medicine, New Orleans, LA, USA
| | - RJ Jones
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - W Matsui
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| |
Collapse
|
66
|
Kletting P, Bunjes D, Reske SN, Glatting G. Improving Anti-CD45 Antibody Radioimmunotherapy Using a Physiologically Based Pharmacokinetic Model. J Nucl Med 2009; 50:296-302. [DOI: 10.2967/jnumed.108.054189] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
67
|
Myeloablative radioimmunotherapy in conditioning prior to haematological stem cell transplantation: closing the gap between benefit and toxicity? Eur J Nucl Med Mol Imaging 2009; 36:484-98. [PMID: 19130053 DOI: 10.1007/s00259-008-0996-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 10/16/2008] [Indexed: 12/22/2022]
Abstract
High-dose radio-/chemotherapy in the context of autologous and allogeneic haematopoietic stem cell transplantation is a double-edged sword. The requirement for dose intensification is linked to an increase in toxicity to noninvolved organs. Particularly for older patients and patients with comorbidities, efficient but toxicity-reduced schemes are needed. Myeloablative radioimmunotherapy is a targeted, internal radiotherapy that uses radiolabelled monoclonal antibodies (mAb) with affinity to the bone marrow. It involves the administration of high radiation doses (up to 30 Gy) to the bone marrow and spleen but without exposing radiosensitive organs to doses higher than 1-7 Gy. Added to conventional or intensity-reduced conditioning, myeloablative radioimmunotherapy may achieve a pronounced antileukaemic effect with tolerable toxicities. A rational and individual design of the ideal nuclide-antibody combination optimizes therapy. The anti-CD33, anti-CD45 and anti-CD66 mAbs appear to be ideal tracers so far. The beta-emitter (90)Y is coupled by DTPA and is the best nuclide for myeloablation. Approval trials for DTPA anti-CD66 mAb are underway in Europe, and in the near future these therapies may become applicable in practice. This review gives an overview of current myeloablative conditioning radioimmunotherapy. We discuss the selection of the optimal radioimmunoconjugate and discuss how radioimmunotherapy might be optimized in the future by individualization of therapy protocols. We also highlight the potential advantages of combination therapies.
Collapse
|
68
|
Affiliation(s)
- Ajay K Gopal
- Division of Medical Oncology, Department of Medicine, University of Washington and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | |
Collapse
|
69
|
Weisdorf D, Forman S. Allogeneic hematopoietic stem cell transplantation for adult acute lymphoblastic leukemia. Cancer Treat Res 2009; 144:441-54. [PMID: 19779879 DOI: 10.1007/978-0-387-78580-6_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Daniel Weisdorf
- University of Minnesota, Mayo Mail Code 480, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
70
|
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
|
71
|
Abstract
Due to the high rate of relapse in younger patients and the overall poor outcome in older patients, novel therapies are needed for the treatment of acute myeloid leukemia (AML). Monoclonal antibodies have become an important treatment modality in cancer therapy. Gemtuzumab ozogamicin (GO), an anti-CD33 immunoconjugate, was approved by the US Food and Drug Administration (FDA) for the treatment of elderly patients with relapsed AML who are not candidates for standard chemotherapy. Single-agent GO and combinations with standard chemotherapeutics have been explored extensively in this disease. Hepatotoxicity and delayed myelosuppression have been dose-limiting. Its toxicity profile is reduced with decreased doses of GO and even by administering only a single infusion. In patients with acute promyelocytic leukemia (APL), the addition of GO can produce molecular remissions and is well tolerated. Targeted immunotherapy with GO for treatment of AML has produced remissions. In order to reduce toxicity and improve efficacy, its optimal dose and schedule and pairing with other standard chemotherapeutic agents need to be defined better in large clinical trials.
Collapse
Affiliation(s)
- Deborah Mulford
- Department of Medicine, Leukemia Program, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
72
|
Sharkey RM, Goldenberg DM. Use of antibodies and immunoconjugates for the therapy of more accessible cancers. Adv Drug Deliv Rev 2008; 60:1407-20. [PMID: 18508155 DOI: 10.1016/j.addr.2008.04.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 04/16/2008] [Indexed: 02/06/2023]
Abstract
There are currently 6 unconjugated antibodies and 3 immunoconjugates approved for use in the United States in a variety of cancers, with a considerable number of new agents in clinical testing and preclinical development. Unconjugated antibodies alone can be effective, but more often, antibodies need to be combined with chemotherapy, which enhances the efficacy of the standard treatment. Immunoconjugates tend to be more effective than their unconjugated counterparts, but their increased toxicity often restricts when and how they are used. In order to improve efficacy, a number of immunoconjugates are being examined in settings where the disease is more easily accessible, such as leukemias, or within compartments that allow easier and more direct access to the tumor, such as in the peritoneal cavity or brain, or both locally and systemically, in adjuvant situations, where the disease burden has been reduced by some other means, and with the main goal of these treatments being to kill residual disease.
Collapse
|
73
|
|
74
|
Abstract
Allogeneic transplantation is an effective treatment for adult patients with high-risk ALL, including patients in first or second remission. Although only a few studies have evaluated the optimal transplant regimens, the data would suggest that a TBI-based regimen results in better disease control. Although not as potent as it is in other hematologic malignancies, the GVL effect is an important component of achieving cure of ALL. Because of the toxicity of the fully ablative regimen, reduced-intensity transplants are being explored in older patients with ALL when the prognosis is especially poor with standard chemotherapy.
Collapse
|
75
|
Integration of monoclonal antibodies and immunoconjugates into the treatment of acute myeloid leukemia. Curr Opin Hematol 2008; 15:95-100. [PMID: 18300754 DOI: 10.1097/moh.0b013e3282f3de84] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review addresses use of monoclonal antibodies and immunoconjugates to treat acute myeloid leukemia. RECENT FINDINGS Monoclonal antibodies used in acute myeloid leukemia have been directed against the antigens CD33, CD45, and CD66. Unconjugated monoclonal antibodies such as lintuzumab have modest activity against overt acute myeloid leukemia but can eliminate minimal residual disease in acute promyelocytic leukemia. Most experience with immunoconjugates is with gemtuzumab ozogamicin, an anti-CD33 monoclonal antibody linked to the potent antitumor antibiotic calicheamicin. Gemtuzumab ozogamicin has shown activity both singly, particularly in acute promyelocytic leukemia, and combined with conventional cytotoxic chemotherapy. Radiolabeled monoclonal antibodies against CD45 and CD66 have also been used to intensify the conditioning regimen before stem cell transplantation. The most promising results were obtained with radiolabeled anti-CD45 antibodies. Antibodies reactive with CD66 have been used to deliver targeted radiation to hematopoietic tissues in patients with advanced myeloid malignancies. SUMMARY Both unlabeled monoclonal antibodies and immunoconjugates appear to have a limited role if used as single agents to treat acute myeloid leukemia. These agents hold promise as potentially useful additions to conventional therapy, but the optimal dosing and timing remain to be defined.
Collapse
|
76
|
Nemecek ER, Green DJ, Fisher DR, Pagel JM, Lin Y, Gopal AK, Durack LD, Rajendran JG, Wilbur DS, Nilsson R, Sandberg B, Press OW. Extracorporeal Adsorption Therapy: A Method to Improve Targeted Radiation Delivered by Radiometal-Labeled Monoclonal Antibodies. Cancer Biother Radiopharm 2008; 23:181-91. [DOI: 10.1089/cbr.2007.0433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Eneida R. Nemecek
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Damian J. Green
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | - John M. Pagel
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Yukang Lin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| | - Ajay K. Gopal
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | | | - D. Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | | | | | - Oliver W. Press
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
77
|
Abstract
Radioimmunotherapy (RIT) combines the mechanism of action and targeting capability of monoclonal antibodies with the tumoricidal effect of radiation and has shown promising results in the treatment of various hematologic malignancies. Based on RIT's efficacy and safety profile, many investigators have evaluated its use in transplant conditioning regimens with the goal of improving long-term disease control with limited toxicity. In lymphoma, two basic transplant approaches targeting CD20 have emerged: (1) myeloablative doses of RIT with or without chemotherapy, and (2) standard nonmyeloablative doses of RIT combined with high-dose chemotherapy. Myeloablative RIT has been shown to be feasible and efficacious using escalated doses of iodine 131-tositumomab, yttrium 90-ibritumomab tiuxetan, and (131)I-rituximab with or without chemotherapy followed by autologous stem cell transplant (ASCT). The second approach predominantly has used standard doses of (90)Y-ibritumomab tiuxetan or (131)I-tositumomab plus BEAM chemotherapy (carmustine [BCNU], etoposide, cytarabine, melphalan) followed by ASCT. RIT targeting CD45, CD33, and CD66 prior to allogeneic transplantation also has been evaluated for the treatment of acute leukemia. Overall RIT-based transplant conditioning for lymphoma and leukemia has been shown to be safe, effective, and feasible with ongoing randomized trials currently underway to definitively establish its place in the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Michelle M Zhang
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
78
|
Simultaneously targeting CD45 significantly increases cytotoxicity of the anti-CD33 immunoconjugate, gemtuzumab ozogamicin, against acute myeloid leukemia (AML) cells and improves survival of mice bearing human AML xenografts. Blood 2008; 111:4813-6. [PMID: 18326813 DOI: 10.1182/blood-2008-01-133785] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Targeting CD33 or CD45 is currently exploited for immunotherapy of acute myeloid leukemia (AML). Gemtuzumab ozogamicin (GO), an immunoconjugate of an anti-CD33 antibody that facilitates cellular uptake of a toxic calicheamicin-gamma(1) derivative, induces complete remissions in a subset of patients with AML. We herein tested whether simultaneous targeting of CD45 could improve GO cytotoxicity against AML cell lines and primary AML cells. We found that the anti-CD45 antibody, BC8, dose-dependently increased cytotoxicity induced by GO, and, to a lesser degree, free calicheamicin-gamma(1). BC8 promoted CD33 endocytosis, suggesting that its effect on GO cytotoxicity may be, at least partly, due to increased uptake and intracellular GO availability. Finally, compared with either agent alone, BC8 combined with GO resulted in marked tumor growth inhibition and superior survival rates of mice bearing human AML xenografts. These data suggest that further study of this antibody combination for clinical use in AML is warranted.
Collapse
|
79
|
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]
|
80
|
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]
|
81
|
|
82
|
Eradication of disseminated leukemia in a syngeneic murine leukemia model using pretargeted anti-CD45 radioimmunotherapy. Blood 2007; 111:2261-8. [PMID: 18042793 DOI: 10.1182/blood-2007-06-097451] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We describe the use of pretargeted radioimmunotherapy (PRIT) using an anti-murine CD45 antibody-streptavidin (SA) conjugate followed by radiobiotin to deliver radiation selectively to murine hematolymphoid tissues, which may potentially augment the efficacy and decrease the toxicity of radioimmunotherapy for disseminated murine leukemia. Biodistribution and therapeutic results demonstrated high target organ to nontarget organ ratios of radioactivity and significant long-term survival in leukemic mice using PRIT. These data suggest that anti-CD45 PRIT using an anti-CD45-SA conjugate in a syngeneic murine model of disseminated leukemia may be more effective and less toxic than directly labeled monoclonal antibodies.
Collapse
|
83
|
Park SI, Press OW. Radioimmunotherapy for treatment of B-cell lymphomas and other hematologic malignancies. Curr Opin Hematol 2007; 14:632-8. [PMID: 17898567 DOI: 10.1097/moh.0b013e3282efb17c] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Radioimmunotherapy has emerged as one of the most promising treatment options for hematologic malignancies. This review will present the latest information on radioimmunotherapy for treatment of hematologic malignancies in various clinical settings and assess its long-term safety profile. RECENT FINDINGS Recent data suggest that radioimmunotherapy with 131I-tositumomab or 90Y-ibritumomab tiuxetan not only induces high response rates but also results in durable remissions in patients with relapsed or refractory indolent non-Hodgkin's lymphomas. Even more notable response rates have been observed when radioimmunotherapy is used as front-line treatment in patients with indolent non-Hodgkin's lymphomas. The use of radioimmunotherapy has been evaluated in the treatment of aggressive lymphomas with promising results, but it remains investigational. Standard doses of radioimmunotherapy given as a conditioning regimen for hematopoietic stem-cell transplant or myeloablative doses of radioimmunotherapy given in conjunction with stem-cell support have yielded encouraging outcomes with durable remissions and a low incidence of treatment-related mortality. SUMMARY The safety and efficacy of radioimmunotherapy has been demonstrated for patients with B-cell lymphomas and other hematologic malignancies in various clinical settings. A number of randomized phase III clinical trials are currently underway to further define radioimmunotherapy's role in the treatment of lymphomas.
Collapse
Affiliation(s)
- Steven I Park
- Department of Medicine, Division of Hematology, University of Washington, USA
| | | |
Collapse
|
84
|
Vallera DA, Sicheneder AR, Taras EP, Brechbiel MW, Vallera JA, Panoskaltsis-Mortari A, Burns LJ. Radiotherapy of CD45-Expressing Daudi Tumors in Nude Mice with Yttrium-90-Labeled, PEGylated Anti-CD45 Antibody. Cancer Biother Radiopharm 2007; 22:488-500. [PMID: 17803443 DOI: 10.1089/cbr.2007.366] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Studies were performed to determine the suitability of using the polyethylene glycol (PEG)-labeled AHN-12 anti-CD45 monoclonal antibody to deliver the high-energy beta-particle-emitting isotope 90Y to a CD45+ B-cell Daudi lymphoma grown as flank tumors in athymic nude mice. The PEGylated radiolabeled antibody displayed a significantly better antitumor effect in the mouse tumor flank model (p<0.03) and significantly better blood pharmacokinetics in normal rats (p<0.05) than the non-PEGylated radiolabeled antibody. Studies of two different sizes of PEG showed that rats given 43 kDa of PEGylated AHN-12, but not 5 kDa of PEGylated AHN-12, had significantly higher radiolabeled antibody blood levels and, therefore, improved pharmacokinetics, as compared to rodents given non-PEGylated radiolabeled AHN-12 (p<0.05). Surviving mice revealed no signs of kidney, liver, or gastrointestinal damage by histology study. Notably, in vitro studies indicated that PEGylation did not have a major effect on labeling efficiency and the binding of labeled antibody. These findings indicate that PEGylation of radiolabeled anti-CD45 antibody may be a useful and desirable means of extending blood half-life and enhancing efficacy. Also, the final outcome may be impacted by the size of the PEG molecule used for the modification of the blood half-life.
Collapse
Affiliation(s)
- Daniel A Vallera
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Cancer Center, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | |
Collapse
|
85
|
Dearling JLJ, Pedley RB. Technological advances in radioimmunotherapy. Clin Oncol (R Coll Radiol) 2007; 19:457-69. [PMID: 17537620 DOI: 10.1016/j.clon.2007.03.016] [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] [Received: 03/08/2007] [Revised: 03/13/2007] [Accepted: 03/28/2007] [Indexed: 10/23/2022]
Abstract
Radioimmunotherapy (RIT) is a method of selectively delivering radionuclides with toxic emissions to cancer cells, while reducing the dose to normal tissues. Although primary tumours can often be treated successfully with external beam radiotherapy or surgery, metastases often escape detection and treatment, leading to therapy failure, and these can be treated with systemic targeted therapies such as RIT. This review describes more recent developments in the field, including both technological developments from the laboratory and increasingly encouraging findings from clinical studies.
Collapse
Affiliation(s)
- J L J Dearling
- Cancer Research UK Targeting & Imaging Group, Department of Oncology, University College London (Hampstead Campus), London, UK.
| | | |
Collapse
|
86
|
Appelbaum FR. Hematopoietic cell transplantation from unrelated donors for treatment of patients with acute myeloid leukemia in first complete remission. Best Pract Res Clin Haematol 2007; 20:67-75. [PMID: 17336256 DOI: 10.1016/j.beha.2006.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hematopoietic cell transplantation from a histocompatible sibling is generally recommended for patients with acute myeloid leukemia in first remission with intermediate or high risk disease. Two-thirds of patients lack a matched sibling raising the question of the utility of matched unrelated transplantation for such patients. Retrospective studies from single institutions and registry data report 44-50% disease-free survival at 5-years following ablative unrelated donor transplantation for adults. The German AML 01/99 is the only prospective study evaluating the utility of matched related and unrelated transplantation for AML patients in first remission with high risk disease and reported 4-year survival of 68% with matched related transplants, 56% with matched unrelated transplants and 23% with autografting. Thus, results suggest that for patients with AML in first remission with high risk features (as determined by cytogenetics or >5% blasts on day 15 of induction) who lack matched siblings, unrelated donor transplantation should be considered. Current challenges are to improve our ability to identify those patients most likely to benefit from early transplantation, to better select donors, and to develop transplant preparative regimens that are safer and more effective.
Collapse
Affiliation(s)
- Frederick R Appelbaum
- Fred Hutchinson Cancer Research Center and University of Washington School of Medicine, Seattle, WA 98109-1024, USA.
| |
Collapse
|
87
|
Small TN, Young JW, Castro-Malaspina H, Prockop S, Wilton A, Heller G, Boulad F, Chiu M, Hsu K, Jakubowski A, Kernan NA, Perales MA, O'Reilly RJ, Papadopoulos EB. Intravenous Busulfan and Melphalan, Tacrolimus, and Short-Course Methotrexate Followed by Unmodified HLA-Matched Related or Unrelated Hematopoietic Stem Cell Transplantation for the Treatment of Advanced Hematologic Malignancies. Biol Blood Marrow Transplant 2007; 13:235-44. [PMID: 17241929 DOI: 10.1016/j.bbmt.2006.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Accepted: 10/10/2006] [Indexed: 12/31/2022]
Abstract
Results of allogeneic hematopoietic stem cell transplantation (HCT) to treat advanced leukemia or myelodysplastic syndrome (MDS) remain poor due to excessive relapse and transplant-related mortality. To improve transplant outcome in this patient population, 43 patients (median age, 46.1 years) with high-risk or advanced lymphoid (n = 5) or myeloid malignancy (n = 38) were prospectively enrolled on a pilot trial of cytoreduction with intravenous busulfan and melphalan followed by an unmodified HLA-A, -B, and -DRbeta1-matched related (n = 18) or unrelated (n = 25) HCT. Graft-versus-host disease (GVHD) prophylaxis consisted of tacrolimus and methotrexate. Thirty-four patients had > or = 5% blasts at the time of HCT; 12 of these had > 20% blasts. Seventeen patients had unfavorable cytogenetics, 8 patients underwent transplantation for secondary MDS or acute myelogenous leukemia, and 4 patients had relapsed after a previous allogeneic transplantation. Although mucositis was the most significant regimen-related toxicity, requiring the addition of folinic acid rescue and failure to receive all 4 doses of methotrexate in 23 patients, the nonrelapse mortality at 30 and 100 days was low at 0% and 16%, respectively. The cumulative incidence of grade II-IV acute GVHD was 24%, and that of extensive chronic GVHD was 7%. With a minimum follow-up of 18 months, the estimated 3-year overall survival is 37% and the estimated disease-free survival (DFS) is 33%. For 18 patients with MDS (< or = RAEB-2) or high-risk myeloproliferative disorder, the estimated 3 year DFS is 61%. These data demonstrate the curative potential of this regimen in patients with high-risk myeloid malignancies.
Collapse
Affiliation(s)
- Trudy N Small
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Bethge WA, Wilbur DS, Sandmaier BM. Radioimmunotherapy as non-myeloablative conditioning for allogeneic marrow transplantation. Leuk Lymphoma 2007; 47:1205-14. [PMID: 16923548 DOI: 10.1080/00423110500485822] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Radioimmunotherapy (RIT) combines the advantages of specific immunotherapy with the cytotoxicity of radionuclides resulting in targeted radiation therapy. In the setting of allogeneic hematopoietic cell transplantation (HCT), RIT can be used to both target tumor cells and to suppress immunocompetent recipient cells. RIT with beta-emitters has been successfully used for further dose intensification of myeloablative conditioning regimens for HCT. Alpha-emitters with their short path length and high linear energy transfer bear certain advantages over beta-emitters if used to target hematopoietic cells. Using a canine model of non-myeloablative HCT, one could demonstrate that pre-transplant RIT with the alpha-emitter bismuth-213 (213Bi) coupled to anti-CD45 or anti-TCRalphabeta monoclonal antibodies (mAb) together with post-grafting immunosuppression with mycophenolate mofetil and cyclosporine can achieve stable engraftment and long-term mixed chimerism. The two mAbs may allow a tailored approach to RIT in non-myeloablative conditioning with 213Bi-anti-CD45 used for patients with hematologic malignancies and 213Bi-anti-TCRalphabeta for non-malignant diseases. Extensive studies of biodistribution, dose de-escalation and toxicity provide the basis for the conception of clinical trials using RIT for non-myeloablative HCT.
Collapse
Affiliation(s)
- Wolfgang A Bethge
- Medical Center, University of Tuebingen, Department of Hematology and Oncology, Tuebingen, Germany
| | | | | |
Collapse
|
89
|
Amadori S, Stasi R. Monoclonal antibodies and immunoconjugates in acute myeloid leukemia. Best Pract Res Clin Haematol 2006; 19:715-36. [PMID: 16997179 DOI: 10.1016/j.beha.2006.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The use of monoclonal antibodies for patients with acute myeloid leukemia is based on targeting cell-surface antigens preferentially expressed on leukemic blasts while sparing normal cells and tissues. The majority of studies performed to date have used antibodies reactive with the CD33 antigen. Phase II studies have demonstrated antileukemic responses with all agents, although less so with unlabeled antibodies. The most promising results have been obtained in the treatment of minimal residual disease in patients with acute promyelocytc leukemia. Antibody-targeted chemotherapy with gemtuzumab ozogamicin has also shown significant activity in patients with relapsed acute myeloid leukemia. Radioimmunotherapy with beta-particle emitters may be most effective for the treatment of bulky disease or as part of a conditioning regimen for hematopoietic stem-cell transplantation, whereas radioimmunotherapy with alpha-particle emitters may be better suited to the treatment of small-volume or minimal residual leukemia. Whether or not monoclonal antibody therapy will improve disease outcome compared with conventional treatment regimens remains to be demonstrated by well-designed clinical trials.
Collapse
Affiliation(s)
- Sergio Amadori
- Department of Hematology, Tor Vergata University Hospital, Viale Oxford 81, 00133 Rome, Italy.
| | | |
Collapse
|
90
|
Quintas-Cardama A, Aribi A, Cortes J, Giles FJ, Kantarjian H, Verstovsek S. Novel approaches in the treatment of systemic mastocytosis. Cancer 2006; 107:1429-39. [PMID: 16948123 DOI: 10.1002/cncr.22187] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the absence of curative options, therapy for aggressive forms of systemic mastocytosis (SM) has relied in the use of cytoreductive agents, mainly interferon-alpha (IFN-alpha) and cladribine. However, responses are transient and only occur in a subset of patients. Gain-of-function mutations at codon 816 of the KIT protooncogene lead to constitutively active Kit receptor molecules, which are central to the pathogenesis of SM. Recent advances in the understanding of the molecular underpinnings of SM have led to the development of small molecules targeting mutant Kit tyrosine kinase isoforms that significantly have widened the range of therapeutic options for patients with SM. Some of these promising agents, such as dasatinib, AMN107, and PKC412, currently are under investigation in clinical trials whereas, others are at different stages of preclinical development. In addition, monoclonal antibodies directed to neoplastic mast cell-restricted surface antigens constitute a viable option for the treatment of SM that warrants further investigation.
Collapse
Affiliation(s)
- Alfonso Quintas-Cardama
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
91
|
Wong JYC. Systemic targeted radionuclide therapy: potential new areas. Int J Radiat Oncol Biol Phys 2006; 66:S74-82. [PMID: 16979445 DOI: 10.1016/j.ijrobp.2005.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 05/31/2005] [Accepted: 05/31/2005] [Indexed: 10/24/2022]
Abstract
Radiation oncology is entering an exciting new era with therapies being delivered in a targeted fashion through an increasing number of novel approaches. External beam radiotherapy now integrates functional and anatomic tumor imaging to guide delivery of conformal radiation to the tumor target. Systemic targeted radionuclide therapy (STaRT) adds an important new dimension by making available to the radiation oncologist biologically targeted radiation therapy. Impressive clinical results with antibody-targeted radiotherapy, leading to the Food and Drug Administration's approval of two anti-CD20 radiolabeled antibodies, highlight the potential of STaRT. Optimization strategies will further improve the efficacy of STaRT by improving delivery systems, modifying the tumor microenvironment to increase targeted dose, and maximizing dose effect. Ultimately, the greatest potential for STaRT will not be as monotherapy, but as therapy integrated into established multimodality regimens and used as adjuvant or consolidative therapy in patients with minimal or micrometastatic disease.
Collapse
Affiliation(s)
- Jeffrey Y C Wong
- Division of Radiation Oncology and Radiation Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA.
| |
Collapse
|
92
|
Gopal AK, Pagel JM, Rajendran JG, Maloney DG, Appelbaum FR, Sorror ML, Sandmaier BM, Storb R, Press OW. Improving the Efficacy of Reduced Intensity Allogeneic Transplantation for Lymphoma using Radioimmunotherapy. Biol Blood Marrow Transplant 2006; 12:697-702. [PMID: 16785058 DOI: 10.1016/j.bbmt.2006.03.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Accepted: 03/28/2006] [Indexed: 11/23/2022]
Abstract
Nonmyeloablative allogeneic transplantation provides a valuable therapeutic option for patients with relapsed non-Hodgkin lymphomas, particularly those that have recurred after autologous transplantation. However, the absence of an intensive conditioning regimen renders this approach less effective for patients with aggressive or bulky lymphoma because rapid tumor growth may outpace the evolution of the graft-versus-lymphoma effect. Radioimmunotherapy provides an attractive, minimally toxic modality to safely prevent early progression of B-cell lymphomas and induce remissions without incurring the risks of traditional intensive therapy. This approach provides a time window during which a robust graft-versus-lymphoma effect may be established before tumor progression, thereby providing more effective long-term disease control. The rationale for incorporation of radioimmunotherapy into reduced intensity allogeneic transplantation regimens for non-Hodgkin lymphoma is discussed, as are current study designs, preliminary results, and future directions.
Collapse
Affiliation(s)
- Ajay K Gopal
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Affiliation(s)
- Edward A Copelan
- Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, the Ohio State University, Columbus, USA.
| |
Collapse
|
94
|
Kotzerke J, Bunjes D, Scheinberg DA. Radioimmunoconjugates in acute leukemia treatment: the future is radiant. Bone Marrow Transplant 2006; 36:1021-6. [PMID: 16247432 DOI: 10.1038/sj.bmt.1705182] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Targeted radiotherapy of the bone marrow using radiolabeled monoclonal antibodies is a therapeutic approach of considerable potential for the treatment of acute leukemia in addition to or as a substitute for total body irradiation. The data currently available, of about 300 patients, suggest that radioimmunotherapy (RIT) with beta-emitters in acute leukemia is feasible and safe using a variety of antibodies (anti-CD33, anti-CD45, anti-CD66) and radionuclides (131I, 90Y, 188Re). It appears to reduce the risk of relapse in high-risk acute myelogenous leukemia (AML) patients transplanted early in the course of their disease (<15% blasts) to 20-30%. Furthermore, it has shown the potential to safely intensify reduced-intensity conditioning regimens (nonrelapse mortality of 25% compared to relapse rate of 55% within 2 years). Significant improvements in the results of refractory patients will probably depend on the successful further development of RIT with alpha-emitters or the use of a cocktail of antibodies labeled with alpha- and beta-emitters, in a first dose escalation study of 213Bi-labeled anti-CD33 in refractory AML (partial) remission could be achieved in 5/18 patients. Randomized trials to evaluate the therapeutic efficacy of RIT in the context of stem cell transplantation have been initiated and the results are keenly anticipated.
Collapse
Affiliation(s)
- J Kotzerke
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | | | | |
Collapse
|
95
|
Schulenburg A, Ulrich-Pur H, Thurnher D, Erovic B, Florian S, Sperr WR, Kalhs P, Marian B, Wrba F, Zielinski CC, Valent P. Neoplastic stem cells: A novel therapeutic target in clinical oncology. Cancer 2006; 107:2512-20. [PMID: 17039500 DOI: 10.1002/cncr.22277] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cancer is among the leading causes of morbidity and mortality in the Western world. Despite recent advances, most therapeutic approaches fail to eradicate the entire neoplastic clone. The remaining cells often develop metastasis and/or recurrences and therefore may represent attractive targets of therapy. A new exciting concept in this regard suggests that each neoplasm represents a heterogeneous population of cells that pertain to long-term tumor growth both in vivo in the natural host and in experimental animals. This concept postulates the existence of small fractions of 'tumor stem cells' that exhibit a capacity for self-renewal and unlimited growth and therefore are distinct from their progeny. Based on these hypotheses, the targeting of neoplastic stem cells is considered indispensable for eradication of the entire clone and for the development of curative treatment approaches. However, tumor stem cells often may be quiescent cells and may express a different profile of targets compared with 'more mature' tumor cells. Therefore, current efforts have attempted to characterize target expression profiles in cancer stem cells in various malignancies. In the this review, the authors have provided a brief summary of the current knowledge of neoplastic stem cells and the application of respective concepts in translational oncology with the ultimate objective of improving anticancer therapy.
Collapse
Affiliation(s)
- Axel Schulenburg
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
Immunotherapeutic strategies have become part of standard cancer treatment. Chimeric and humanized antibodies have demonstrated activity against a variety of tumors. Although the humanized anti-CD33 antibody HuM195 has only modest activity against overt acute myeloid leukemia (AML), it can eliminate minimal residual disease in acute promyelocytic leukemia. High-dose radioimmunotherapy with b-particle-emitting isotopes targeting CD33, CD45, and CD66 can potentially allow intensification of antileukemic therapy before hematopoietic stem cell transplantation. Conversely, a-particle immunotherapy with isotopes such as bismuth-213 or actinium-225 offers the possibility of selective tumor cell kill while sparing surrounding normal tissues. Targeted chemotherapy with the anti-CD33- calicheamicin construct gemtuzumab ozogamicin has produced remissions in relapsed AML and appears promising when used in combination with standard chemotherapy for newly diagnosed AML. T-cell recognition of peptide antigens presented on the cell surface in combination with major histocompatibility complex antigen provides another potentially promising approach for the treatment of AML.
Collapse
Affiliation(s)
- Joseph G Jurcic
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, Box 458, New York, NY 10021, USA.
| |
Collapse
|
97
|
Abstract
Paul Erhlich conceived of antibody-based immunotherapy in the nineteenth century. Rituximab, which is a chimeric monoclonal antibody produced by recombinant technology, became the first monoclonal antibody to be approved for haematological malignancies by the US Food and Drug Administration. Subsequently, radiolabelling technologies have made it possible to chelate radioactive isotopes to monoclonal antibodies, which retain their specificity and take advantage of targeted delivery of localised radiation. Radioimmunoconjugates are an attractive therapeutic option for lymphomas due to the inherent sensitivity to radiotherapy, the fact that the local emission of ionising radiation by radiolabelled antibodies may kill cells with or without the target antigen in close proximity to the bound antibody, and penetrating radiation may obviate the problem of limited antibody penetration into bulky, poorly vascularised tumours. This paper reviews rituximab, alemtuzumab and gemtuzumab ozogamicin as monoclonal antibody therapies for leukaemias and lymphomas.
Collapse
MESH Headings
- Alemtuzumab
- Aminoglycosides/therapeutic use
- Animals
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/therapeutic use
- Antigens, CD/immunology
- Antigens, CD20/immunology
- Antigens, Differentiation, Myelomonocytic/immunology
- Antigens, Neoplasm/immunology
- Antineoplastic Agents/therapeutic use
- CD52 Antigen
- Gemtuzumab
- Glycoproteins/immunology
- Humans
- Leukemia/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, T-Cell/drug therapy
- Lymphoma/drug therapy
- Lymphoma, B-Cell/drug therapy
- Lymphoma, Follicular/drug therapy
- Lymphoma, Mantle-Cell/drug therapy
- Randomized Controlled Trials as Topic
- Rituximab
- Sialic Acid Binding Ig-like Lectin 3
Collapse
Affiliation(s)
- Samuel A Jacobs
- University of Pittsburgh School of Medicine, 5150 Center Avenue, Suite 510, Pittsburgh, PA 15232, USA.
| | | |
Collapse
|
98
|
Sui X, Bensinger W, Press O. Improved conditioning regimens for autologous transplantation using targeted radiotherapy. Acta Haematol 2005; 114:230-8. [PMID: 16269863 DOI: 10.1159/000088414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Most patients undergoing autologous hematopoietic stem cell transplantation for malignant diseases suffer recurrences of their neoplasms and die due to the inability of conventional high-dose conditioning regimens to eradicate their malignancies. As a result, intensive efforts to develop more effective conditioning regimens are currently under way at many institutions. Encouraging results have been obtained using targeted radiotherapy with radiolabeled antibodies or bone-seeking isotopes as components of novel conditioning regimens for autologous transplantation of patients with lymphomas, multiple myeloma and bone metastases. Results with radiolabeled antibodies targeting epithelial antigens on solid tumors, however, have been less encouraging. This report reviews the status of clinical studies using myeloablative doses of targeted radiotherapy in patients undergoing autologous stem cell transplantation for hematological malignancies or solid tumors.
Collapse
Affiliation(s)
- Xingwei Sui
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | |
Collapse
|
99
|
Pagel JM, Appelbaum FR, Eary JF, Rajendran J, Fisher DR, Gooley T, Ruffner K, Nemecek E, Sickle E, Durack L, Carreras J, Horowitz MM, Press OW, Gopal AK, Martin PJ, Bernstein ID, Matthews DC. 131I-anti-CD45 antibody plus busulfan and cyclophosphamide before allogeneic hematopoietic cell transplantation for treatment of acute myeloid leukemia in first remission. Blood 2005; 107:2184-91. [PMID: 16254140 PMCID: PMC1895719 DOI: 10.1182/blood-2005-06-2317] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In an attempt to improve outcomes for patients with acute myeloid leukemia (AML) after allogeneic hematopoietic cell transplantation (HCT), we conducted a phase 1/2 study in which targeted irradiation delivered by 131I-anti-CD45 antibody was combined with targeted busulfan (BU; area-under-curve, 600-900 ng/mL) and cyclophosphamide (CY; 120 mg/kg). Fifty-two (88%) of 59 patients receiving a trace 131I-labeled dose of 0.5 mg/kg anti-CD45 murine antibody had higher estimated absorbed radiation in bone marrow and spleen than in any other organ. Forty-six patients were treated with 102 to 298 mCi (3774-11 026 MBq) 131I, delivering an estimated 5.3 to 19 (mean, 11.3) Gy to marrow, 17-72 (mean, 29.7) Gy to spleen, and 3.5 Gy (n = 4) to 5.25 Gy (n = 42) to the liver. The estimated 3-year nonrelapse mortality and disease-free survival (DFS) were 21% and 61%, respectively. These results were compared with those from 509 similar International Bone Marrow Transplant Registry patients who underwent transplantation using BU/CY alone. After adjusting for differences in age and cytogenetics risk, the hazard of mortality among all antibody-treated patients was 0.65 times that of the Registry patients (95% CI 0.39-1.08; P = .09). The addition of targeted hematopoietic irradiation to conventional BU/CY is feasible and well tolerated, and phase 2 results are sufficiently encouraging to warrant further study.
Collapse
Affiliation(s)
- John M Pagel
- Division of Clinical Research, Fred Hutchinson Cancer Research Center D5-380, 1100 Fairview Ave N, PO Box 19024, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Disease relapse remains the major cause of treatment failure in adults with acute myeloid leukaemia (AML). This reflects both the failure of current salvage regimens and the absence of effective strategies to secure long-term disease-free survival in those patients who achieve a second remission. Recent progress in understanding the pathogenesis of relapsed disease has enabled the identification of a variety of dysregulated molecular pathways and these now provide a rational basis for the design of novel targeted therapies. At the same time, advances in allogeneic stem-cell transplantation have permitted the extension of the curative potential of allografting to patients in whom it was previously contraindicated. As a result, a range of novel drug and transplant therapies has become available in patients with relapsed AML, and it is realistic to anticipate that a co-ordinated assessment of their clinical and biological impact will provide the basis for the design of future, more effective treatments in relapsed disease.
Collapse
Affiliation(s)
- Charles Craddock
- Leukaemia Unit, Department of Haematology, Queen Elizabeth Hospital, Birmingham, UK.
| | | | | | | |
Collapse
|