1
|
Orchard K, Langford J, Guy M, Lewis G, Michopoulou S, Cooper M, Zvavamwe C, Richardson D, Lewington V. Efficient bone marrow irradiation and low uptake by non-haematological organs with an yttrium-90-anti-CD66 antibody prior to haematopoietic stem cell transplantation. Bone Marrow Transplant 2024; 59:1247-1257. [PMID: 38867006 PMCID: PMC11368815 DOI: 10.1038/s41409-024-02317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 06/14/2024]
Abstract
We report the results of a Phase I radiation dose escalation study using an yttrium-90 (90Y) labelled anti-CD66 monoclonal antibody given with standard conditioning regimen for patients receiving haematopoietic stem cell transplants for myeloid leukaemia or myeloma. The 90Y-labelled anti-CD66 was infused prior to standard conditioning. In total, 30 patients entered the trial and 29 received 90Y-labelled mAb, at infused radiation activity levels of 5, 10, 25, or 37.5 megaBequerel (MBq)/kg lean body weight. A prerequisite for receiving the 90Y-labelled mAb was favourable dosimetry determined by single-photon emission computerised tomography (SPECT) dosimetry following administration of indium-111 (111In) anti-CD66. Estimated absorbed radiation doses delivered to the red marrow demonstrated a linear relationship with the infused activity of 90Y-labelled mAb. At the highest activity level of 37.5 MBq/kg, mean estimated radiation doses for red marrow, liver, spleen, kidneys and lungs were 24.6 ± 5.6 Gy, 5.8 ± 2.7 Gy, 19.1 ± 8.0 Gy, 2.1 ± 1.1 and 2.2 ± 0.9, respectively. All patients engrafted, treatment-related mortality 1-year post-transplant was zero. Toxicities were no greater than those anticipated for similar conditioning regimens without targeted radiation. The ability to substantially intensify conditioning prior to haematopoietic stem cell transplantation without increasing toxicity warrants further testing to determine efficacy. clinicaltrials.gov identifier: NCT01521611.
Collapse
Affiliation(s)
- Kim Orchard
- Wessex Blood and Marrow Transplantation Programme, Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
- NIHR/CRUK Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK.
| | - Jonathan Langford
- NIHR/CRUK Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK
| | - Matthew Guy
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Gemma Lewis
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Sofia Michopoulou
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Margaret Cooper
- Department of Nuclear Medicine, Bart's and the London NHS Trust, London, UK
- PET Imaging Centre Facility, King's College London, London, UK
| | - Clint Zvavamwe
- Radiopharmacy, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Deborah Richardson
- Wessex Blood and Marrow Transplantation Programme, Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Valerie Lewington
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
2
|
Latgé A, Krim M, Vija L, Ysebaert L, Zerdoud S. Diffuse Bone Uptake of 131 I and Effect on Blood Counts in a Patient With Papillary Thyroid Carcinoma and Chronic Lymphocytic Leukemia. Clin Nucl Med 2024; 49:340-341. [PMID: 38377372 DOI: 10.1097/rlu.0000000000005119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
ABSTRACT A 57-year-old woman with history of chronic lymphocytic leukemia was referred to our center for adjuvant 131 I therapy following complete thyroidectomy for differentiated thyroid cancer. Posttherapeutic scintigraphy revealed atypical diffuse osteomedullar uptake. A major drop in lymphocyte count was observed, from 117.7 g/L to 4.8 g/L 8 weeks after 131 I therapy. Bone marrow uptake is presumed to be related to tracer sequestration in leukemic cells. White blood cell count normalization suggests a high sensitivity of leukemic cells to beta emission. This scintigraphic pattern may act as a pitfall for nuclear medicine physician.
Collapse
Affiliation(s)
- Adrien Latgé
- From the Nuclear Medicine Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse
| | - Mehdi Krim
- From the Nuclear Medicine Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse
| | | | | | - Slimane Zerdoud
- From the Nuclear Medicine Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse
| |
Collapse
|
3
|
Crossley M, Christakopoulos GE, Weiss MJ. Effective therapies for sickle cell disease: are we there yet? Trends Genet 2022; 38:1284-1298. [PMID: 35934593 PMCID: PMC9837857 DOI: 10.1016/j.tig.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 01/24/2023]
Abstract
Sickle cell disease (SCD) is a common genetic blood disorder associated with acute and chronic pain, progressive multiorgan damage, and early mortality. Recent advances in technologies to manipulate the human genome, a century of research and the development of techniques enabling the isolation, efficient genetic modification, and reimplantation of autologous patient hematopoietic stem cells (HSCs), mean that curing most patients with SCD could soon be a reality in wealthy countries. In parallel, ongoing research is pursuing more facile treatments, such as in-vivo-delivered genetic therapies and new drugs that can eventually be administered in low- and middle-income countries where most SCD patients reside.
Collapse
Affiliation(s)
- Merlin Crossley
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia 2052.
| | | | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
4
|
Wong JY, Liu A, Han C, Dandapani S, Schultheiss T, Palmer J, Yang D, Somlo G, Salhotra A, Hui S, Al Malki MM, Rosenthal J, Stein A. Total marrow irradiation (TMI): Addressing an unmet need in hematopoietic cell transplantation - a single institution experience review. Front Oncol 2022; 12:1003908. [PMID: 36263219 PMCID: PMC9574324 DOI: 10.3389/fonc.2022.1003908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose TMI utilizes IMRT to deliver organ sparing targeted radiotherapy in patients undergoing hematopoietic cell transplantation (HCT). TMI addresses an unmet need, specifically patients with refractory or relapsed (R/R) hematologic malignancies who have poor outcomes with standard HCT regimens and where attempts to improve outcomes by adding or dose escalating TBI are not possible due to increased toxicities. Over 500 patients have received TMI at this center. This review summarizes this experience including planning and delivery, clinical results, and future directions. Methods Patients were treated on prospective allogeneic HCT trials using helical tomographic or VMAT IMRT delivery. Target structures included the bone/marrow only (TMI), or the addition of lymph nodes, and spleen (total marrow and lymphoid irradiation, TMLI). Total dose ranged from 12 to 20 Gy at 1.5-2.0 Gy fractions twice daily. Results Trials demonstrate engraftment in all patients and a low incidence of radiation related toxicities and extramedullary relapses. In R/R acute leukemia TMLI 20 Gy, etoposide, and cyclophosphamide (Cy) results in a 1-year non-relapse mortality (NRM) rate of 6% and 2-year overall survival (OS) of 48%; TMLI 12 Gy added to fludarabine (flu) and melphalan (mel) in older patients (≥ 60 years old) results in a NRM rate of 33% comparable to flu/mel alone, and 5-year OS of 42%; and TMLI 20 Gy/flu/Cy and post-transplant Cy (PTCy) in haplo-identical HCT results in a 2-year NRM rate of 13% and 1-year OS of 83%. In AML in complete remission, TMLI 20 Gy and PTCy results in 2-year NRM, OS, and GVHD free/relapse-free survival (GRFS) rates of 0%, 86·7%, and 59.3%, respectively. Conclusion TMI/TMLI shows significant promise, low NRM rates, the ability to offer myeloablative radiation containing regimens to older patients, the ability to dose escalate, and response and survival rates that compare favorably to published results. Collaboration between radiation oncology and hematology is key to successful implementation. TMI/TMLI represents a paradigm shift from TBI towards novel strategies to integrate a safer and more effective target-specific radiation therapy into HCT conditioning beyond what is possible with TBI and will help expand and redefine the role of radiotherapy in HCT.
Collapse
Affiliation(s)
- Jeffrey Y.C. Wong
- Departments of Radiation Oncology, City of Hope, Duarte, CA, United States
| | - An Liu
- Departments of Radiation Oncology, City of Hope, Duarte, CA, United States
| | - Chunhui Han
- Departments of Radiation Oncology, City of Hope, Duarte, CA, United States
| | - Savita Dandapani
- Departments of Radiation Oncology, City of Hope, Duarte, CA, United States
| | | | - Joycelynne Palmer
- Department Computational and Quantitative Medicine, City of Hope, Duarte, CA, United States
| | - Dongyun Yang
- Department Computational and Quantitative Medicine, City of Hope, Duarte, CA, United States
| | - George Somlo
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Amandeep Salhotra
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Susanta Hui
- Departments of Radiation Oncology, City of Hope, Duarte, CA, United States
| | - Monzr M. Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Joseph Rosenthal
- Department of Pediatrics, City of Hope, Duarte, CA, United States
| | - Anthony Stein
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, United States
| |
Collapse
|
5
|
Thomas X, Elhamri M, Deloire A, Heiblig M. Antibody-based therapy for acute myeloid leukemia: a review of phase 2 and 3 trials. Expert Opin Emerg Drugs 2022; 27:169-185. [PMID: 35749672 DOI: 10.1080/14728214.2022.2094365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite recent advances in the treatment of adult acute myeloid leukemia (AML), the clinical outcome of patients continues to be unsatisfactory especially among older patients, those with a high-risk profile, and in the relapsed/refractory setting. For this reason, recent clinical trials have explored novel therapeutic agents either used alone or in combination with intensive chemotherapy or low-intensity treatments. AREAS COVERED The current paper reviews the clinical development of monoclonal antibody-based therapies in AML, their current status and phases 2 and 3 prospective trials. EXPERT OPINION Monoclonal antibody-based therapies demonstrated efficacy and tolerability in several clinical trials, especially when used in combination either with '3+7' chemotherapy or with low-intensity treatments. Additional studies are needed to determine new antigens for antibody-based therapies that target leukemia stem cells and spare normal hematopoiesis. Phase 2 and 3 additional clinical trial data are needed to assess the promise of first trials, especially regarding chimeric antigen receptor T cells redirected against myeloid antigens and immune checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Xavier Thomas
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Mohamed Elhamri
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Alexandre Deloire
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Maël Heiblig
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| |
Collapse
|
6
|
Parakh S, Lee ST, Gan HK, Scott AM. Radiolabeled Antibodies for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:1454. [PMID: 35326605 PMCID: PMC8946248 DOI: 10.3390/cancers14061454] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/14/2022] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
Radioimmunoconjugates consist of a monoclonal antibody (mAb) linked to a radionuclide. Radioimmunoconjugates as theranostics tools have been in development with success, particularly in hematological malignancies, leading to approval by the US Food and Drug Administration (FDA) for the treatment of non-Hodgkin's lymphoma. Radioimmunotherapy (RIT) allows for reduced toxicity compared to conventional radiation therapy and enhances the efficacy of mAbs. In addition, using radiolabeled mAbs with imaging methods provides critical information on the pharmacokinetics and pharmacodynamics of therapeutic agents with direct relevance to the optimization of the dose and dosing schedule, real-time antigen quantitation, antigen heterogeneity, and dynamic antigen changes. All of these parameters are critical in predicting treatment responses and identifying patients who are most likely to benefit from treatment. Historically, RITs have been less effective in solid tumors; however, several strategies are being investigated to improve their therapeutic index, including targeting patients with minimal disease burden; using pre-targeting strategies, newer radionuclides, and improved labeling techniques; and using combined modalities and locoregional application. This review provides an overview of the radiolabeled intact antibodies currently in clinical use and those in development.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
| | - Sze Ting Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Hui K. Gan
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| | - Andrew M. Scott
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| |
Collapse
|
7
|
Rosenblat TL, McDevitt MR, Carrasquillo JA, Pandit-Taskar N, Frattini MG, Maslak PG, Park JH, Douer D, Cicic D, Larson SM, Scheinberg DA, Jurcic JG. Treatment of Patients with Acute Myeloid Leukemia with the Targeted Alpha-Particle Nano-Generator Actinium-225-Lintuzumab. Clin Cancer Res 2022; 28:2030-2037. [PMID: 35247915 DOI: 10.1158/1078-0432.ccr-21-3712] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE The anti-CD33 antibody lintuzumab has modest activity against acute myeloid leukemia (AML). To increase its potency, lintuzumab was conjugated to actinium-225 (225Ac), a radionuclide yielding 4 α-particles. This first-in-human, phase I trial was conducted to determine the safety, pharmacology, and biological activity of 225Ac-lintuzumab. PATIENTS AND METHODS Eighteen patients (median age, 64 years; range, 45-80) with relapsed or refractory AML received a single infusion of 225Ac-lintuzumab at activities of 18.5-148 kBq/kg. RESULTS The maximum tolerated dose was 111 kBq/kg. Dose-limiting toxicities included myelosuppression lasting > 35 days in one patient receiving 148 kBq/kg and death from sepsis in two patients treated with 111 and 148 kBq/kg. Myelosuppression was the most common toxicity. Significant extramedullary toxicities were limited to transient grade 3 liver function abnormalities. Pharmacokinetics were determined by gamma counting serial whole blood, plasma, and urine samples at energy windows for the 225Ac daughters, francium-221 and bismuth-213. Two-phase elimination kinetics were seen with mean plasma t1/2-α and t1/2-β of 1.9 and 38 hours, respectively. Peripheral blood blasts were eliminated in 10 of 16 evaluable patients (63%) but only at doses of {greater than or equal to} 37 kBq/kg. Bone marrow blasts were reduced in 10 of 15 evaluable patients (67%), including 3 patients with marrow blasts {less than or equal to} 5% and 1 patient with a morphologic leukemia-free state. CONCLUSIONS Therapy for AML with the targeted α-particle generator 225Ac-lintuzumab was feasible with an acceptable safety profile. Elimination of circulating blasts or reductions in marrow blasts were observed across all dose levels.
Collapse
Affiliation(s)
| | - Michael R McDevitt
- Memorial Sloan Kettering Cancer Center, New York, New York, United States
| | | | | | | | - Peter G Maslak
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jae H Park
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Dan Douer
- University of Southern California, United States
| | - Dragan Cicic
- Sellas Life Sciences Group, New York, NY, United States
| | - Steven M Larson
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Joseph G Jurcic
- Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
8
|
Coltoff AR, Jurcic JG. Targeted radionuclide therapy of hematologic malignancies. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00117-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
9
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
10
|
Suman SK, Subramanian S, Mukherjee A. Combination radionuclide therapy: A new paradigm. Nucl Med Biol 2021; 98-99:40-58. [PMID: 34029984 DOI: 10.1016/j.nucmedbio.2021.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022]
Abstract
Targeted molecular radionuclide therapy (MRT) has shown its potential for the treatment of cancers of multiple origins. A combination therapy strategy employing two or more distinct therapeutic approaches in cancer management is aimed at circumventing tumor resistance by simultaneously targeting compensatory signaling pathways or bypassing survival selection mutations acquired in response to individual monotherapies. Combination radionuclide therapy (CRT) is a newer application of the concept, utilizing a combination of radiolabeled molecular targeting agents with chemotherapy and beam radiation therapy for enhanced therapeutic index. Encouraging results are reported with chemotherapeutic agents in combination with radiolabeled targeting molecules for cancer therapy. With increasing awareness of the various survival and stress response pathways activated after radionuclide therapy, different holistic combinations of MRT agents with radiosensitizers targeting such pathways have also been explored. MRT has also been studied in combination with beam radiotherapy modalities such as external beam radiation therapy and carbon ion radiation therapy to enhance the anti-tumor response. Nanotechnology aids in CRT by bringing together multiple monotherapies on a single nanostructure platform for treating cancers in a more precise or personalized way. CRT will be a key player in managing cancers if correctly tailored to the individual patient profile. The success of CRT lies in an in-depth understanding of the radiobiological principles and pathways activated in response.
Collapse
Affiliation(s)
- Shishu Kant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Suresh Subramanian
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India.
| |
Collapse
|
11
|
Doerfler PA, Sharma A, Porter JS, Zheng Y, Tisdale JF, Weiss MJ. Genetic therapies for the first molecular disease. J Clin Invest 2021; 131:146394. [PMID: 33855970 PMCID: PMC8262557 DOI: 10.1172/jci146394] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Sickle cell disease (SCD) is a monogenic disorder characterized by recurrent episodes of severe bone pain, multi-organ failure, and early mortality. Although medical progress over the past several decades has improved clinical outcomes and offered cures for many affected individuals living in high-income countries, most SCD patients still experience substantial morbidity and premature death. Emerging technologies to manipulate somatic cell genomes and insights into the mechanisms of developmental globin gene regulation are generating potentially transformative approaches to cure SCD by autologous hematopoietic stem cell (HSC) transplantation. Key components of current approaches include ethical informed consent, isolation of patient HSCs, in vitro genetic modification of HSCs to correct the SCD mutation or circumvent its damaging effects, and reinfusion of the modified HSCs following myelotoxic bone marrow conditioning. Successful integration of these components into effective therapies requires interdisciplinary collaborations between laboratory researchers, clinical caregivers, and patients. Here we summarize current knowledge and research challenges for each key component, emphasizing that the best approaches have yet to be developed.
Collapse
Affiliation(s)
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy
| | | | - Yan Zheng
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - John F. Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
12
|
Mahalleh M, Shabani M, Rayzan E, Rezaei N. Reinforcing the primary immunotherapy modulators against acute leukemia; monoclonal antibodies in AML. Immunotherapy 2019; 11:1583-1600. [PMID: 31841068 DOI: 10.2217/imt-2019-0043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent therapeutic advances in cancer treatment recruit immune system potentiation against malignant cells. Numerous ongoing clinical trials on immunotherapy methods, either monotherapy or combination therapy, are investigating the impeding factors on the way of acute myeloid leukemia (AML) treatment. Due to the genetic diversity in AML progenitors, combining various strategies is more likely to be useful for improving patient outcomes. This review describes the details of applying monoclonal antibodies against AML, focusing on CD33, CD123, FLT3, CD45 and CD66 targeting. Furthermore, it clarifies the importance of immunotoxins, bispecific antibodies, chimeric antigen receptor (CAR)-T cells and T cell receptor-modified cells as reinforcing agents for monoclonal antibodies.
Collapse
Affiliation(s)
- Mehrdad Mahalleh
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsima Shabani
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Elham Rayzan
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Abstract
The short range and high linear energy transfer of α-particles offer the potential for efficient tumor killing while sparing normal bystander cells. Hematologic malignancies are ideally suited to targeted α-particle therapy (TAT) due to easy accessibility of malignant cells in blood, bone marrow, lymph nodes, and spleen as well as their radiosensitivity. Most clinical trials using α-particle therapy for hematologic malignancies have focused on acute myeloid leukemia (AML); however, preclinical studies have shown activity against other diseases such as non-Hodgkin's lymphoma and multiple myeloma. To date, the short-lived radionuclide bismuth-213 (213Bi) and its parent actinium-225 (225Ac) have been used clinically, but trials with astatinie-211 (211At) have recently begun, and thorium-227 (227Th) has shown promising preclinical results. Lintuzumab is a humanized monoclonal antibody that targets the cell surface antigen CD33, which is expressed on the vast majority of AML cells. Initial studies showed that 213Bi-labeled lintuzumab had antileukemic activity and could produce remissions after partial cytoreduction with cytarabine. An initial phase I trial demonstrated that a single infusion of 225Ac-lintuzumab could be given safely at doses upto 111 kBq/kg with antileukemic activity across all dose levels. A second phase I study showed that fractionated-dose 225Ac-lintuzumab could be safely combined with low-dose cytarabine and produced objective responses in 28% of older patients with untreated AML. In a phase II study, treatment with 225Ac-lintuzumab monotherapy for a similar patient population resulted in remission in 69% of patients receiving two fractions of 74 kBq/kg and 22% of patients receiving two 55.5-kBq/kg fractions. Additionally, TAT may be useful in intensifying antileukemic therapy prior to hematopoietic cell transplantation, and pretargeting strategies offer the possibility for improved tumor-to-normal organ dose ratios.
Collapse
Affiliation(s)
- Joseph G Jurcic
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center; Herbert Irving Comprehensive Cancer Center, and New York-Presbyterian Hospital, New York, NY.
| |
Collapse
|
14
|
Srideshikan SM, Brooks J, Zuro D, Kumar B, Sanchez J, Echavarria Parra L, Orellana M, Vishwasrao P, Nair I, Chea J, Poku K, Bowles N, Miller A, Ebner T, Molnar J, Rosenthal J, Vallera DA, Wong JYC, Stein AS, Colcher D, Shively JE, Yazaki PJ, Hui SK. ImmunoPET, [ 64Cu]Cu-DOTA-Anti-CD33 PET-CT, Imaging of an AML Xenograft Model. Clin Cancer Res 2019; 25:7463-7474. [PMID: 31548348 DOI: 10.1158/1078-0432.ccr-19-1106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/13/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Acute myeloid leukemia (AML) is a highly aggressive form of leukemia, which results in poor survival outcomes. Currently, diagnosis and prognosis are based on invasive single-point bone marrow biopsies (iliac crest). There is currently no AML-specific noninvasive imaging method to detect disease, including in extramedullary organs, representing an unmet clinical need. About 85% to 90% of human myeloid leukemia cells express CD33 cell surface receptors, highlighting CD33 as an ideal candidate for AML immunoPET. EXPERIMENTAL DESIGN We evaluated whether [64Cu]Cu-DOTA-anti-CD33 murine mAb can be used for immunoPET imaging of AML in a preclinical model. MicroCT was adjusted to detect spatial/anatomical details of PET activity. For translational purposes, a humanized anti-CD33 antibody was produced; we confirmed its ability to detect disease and its distribution. We reconfirmed/validated CD33 antibody-specific targeting with an antibody-drug conjugate (ADC) and radioimmunotherapy (RIT). RESULTS [64Cu]Cu-DOTA-anti-CD33-based PET-CT imaging detected CD33+ AML in mice with high sensitivity (95.65%) and specificity (100%). The CD33+ PET activity was significantly higher in specific skeletal niches [femur (P < 0.00001), tibia (P = 0.0001), humerus (P = 0.0014), and lumber spine (P < 0.00001)] in AML-bearing mice (over nonleukemic control mice). Interestingly, the hybrid PET-CT imaging showed high disease activity in the epiphysis/metaphysis of the femur, indicating regional spatial heterogeneity. Anti-CD33 therapy using newly developed humanized anti-CD33 mAb as an ADC (P = 0.02) and [225Ac]Ac-anti-CD33-RIT (P < 0.00001) significantly reduced disease burden over that of respective controls. CONCLUSIONS We have successfully developed a novel anti-CD33 immunoPET-CT-based noninvasive modality for AML and its spatial distribution, indicating a preferential skeletal niche.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Cell Line, Tumor
- Copper Radioisotopes/chemistry
- Disease Models, Animal
- Heterocyclic Compounds, 1-Ring/chemistry
- Immunoconjugates/pharmacokinetics
- Leukemia, Myeloid, Acute/diagnostic imaging
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Positron Emission Tomography Computed Tomography/methods
- Radiopharmaceuticals/pharmacokinetics
- Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
- Sialic Acid Binding Ig-like Lectin 3/immunology
- Tissue Distribution
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | - Jamison Brooks
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Darren Zuro
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Bijender Kumar
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - James Sanchez
- Beckman Research Institute of City of Hope, Duarte, California
| | | | - Marvin Orellana
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Paresh Vishwasrao
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Indu Nair
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Junie Chea
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Kofi Poku
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Nicole Bowles
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Aaron Miller
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Todd Ebner
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Justin Molnar
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Joseph Rosenthal
- Department of Pediatrics, City of Hope National Medical Center, Duarte, California
| | - Daniel A Vallera
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey Y C Wong
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Anthony S Stein
- Department of Hematology/HCT, City of Hope National Medical Center, Duarte, California
| | - David Colcher
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - John E Shively
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Paul J Yazaki
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, California
| | - Susanta K Hui
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California.
| |
Collapse
|
15
|
Egorova BV, Fedorova OA, Kalmykov SN. Cationic radionuclides and ligands for targeted therapeutic radiopharmaceuticals. RUSSIAN CHEMICAL REVIEWS 2019. [DOI: 10.1070/rcr4890] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This review considers the already used and potential α- and β-emitting cationic radionuclides for targeted radionuclide therapy. Recent results of laboratory, preclinical and clinical applications of these radionuclides are discussed. As opposed to β-emitters, which are already used in nuclear medicine, α-emitters involved in targeted radiopharmaceuticals were subjected to clinical trials only recently and were found to be therapeutically effective. The review summarizes recent trends in the development of ligands as components of radiopharmaceuticals addressing specific features of short-lived cationic radionuclides applied in medicine. Despite a steadily growing number of chelating ligands, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and diethylenetriaminepentaacetic acid (DTPA) remain the most widely used agents in nuclear medicine. The drawbacks of these compounds restrict the application of radionuclides in medicine. Variations in the macrocycle size, the introduction and modification of substituents can significantly improve the chelating ability of ligands, enhance stability of radionuclide complexes with these ligands and eliminate the influence of ligands on the affinity of biological targeting vectors.
The bibliography includes 189 references.
Collapse
|
16
|
Morsink LM, Walter RB. Novel monoclonal antibody-based therapies for acute myeloid leukemia. Best Pract Res Clin Haematol 2019; 32:116-126. [PMID: 31203993 DOI: 10.1016/j.beha.2019.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/07/2019] [Indexed: 12/21/2022]
Abstract
There has been long-standing interest in using monoclonal antibodies to improve outcomes of people with acute myeloid leukemia (AML). While several candidate therapeutics have failed at various stages of clinical testing, improved survival of some patients receiving the CD33 antibody-drug conjugate gemtuzumab ozogamicin has provided first evidence that monoclonal antibodies have a role in the armamentarium against AML. Over the last several years, work to improve the success of monoclonal antibody-based therapies in AML has focused on the identification and exploration of new antigen targets as much as on the development of novel treatment formats such as use of unconjugated engineered monoclonal antibodies and conjugated antibodies, delivering highly potent small molecule drugs or radionuclides to AML cells. Here, we will provide a brief overview of current efforts with such investigational monoclonal antibody-based therapeutics.
Collapse
Affiliation(s)
- Linde M Morsink
- Department of Hematology, University Medical Center Groningen, Groningen, the Netherlands
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Nawrocki T, Tritt TC, Neti PVSV, Rosen AS, Dondapati AR, Howell RW. Design and testing of a microcontroller that enables alpha particle irradiators to deliver complex dose rate patterns. Phys Med Biol 2018; 63:245022. [PMID: 30524061 PMCID: PMC8528213 DOI: 10.1088/1361-6560/aaf269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
There is increasing interest in using alpha particle emitting radionuclides for cancer therapy because of their unique cytotoxic properties which are advantageous for eradicating tumor cells. The high linear energy transfer (LET) of alpha particles produces a correspondingly high density of ionizations along their track. Alpha particle emitting radiopharmaceuticals deposit this energy in tissues over prolonged periods with complex dose rate patterns that depend on the physical half-life of the radionuclide, and the biological uptake and clearance half-times in tumor and normal tissues. We have previously shown that the dose rate increase half-time that arises as a consequence of these biokinetics can have a profound effect on the radiotoxicity of low-LET radiation. The microcontroller hardware and software described here offer a unique way to deliver these complex dose rate patterns with a broad-beam alpha particle irradiator, thereby enabling experiments to study the radiobiology of complex dose rate patterns of alpha particles. Complex dose rate patterns were created by precise manipulation of the timing of opening and closing of the electromechanical shutters of an α-particle irradiator. An Arduino Uno and custom circuitry was implemented to control the shutters. The software that controls the circuits and shutters has a user-friendly Graphic User Interface (GUI). Alpha particle detectors were used to validate the programmed dose rate profiles. Circuit diagrams and downloadable software are provided to facilitate adoption of this technology by other radiobiology laboratories.
Collapse
Affiliation(s)
- Tomer Nawrocki
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
INTRODUCTION There is long-standing interest in drugs targeting the myeloid differentiation antigen CD33 in acute myeloid leukemia (AML). Positive results from randomized trials with the antibody-drug conjugate (ADC) gemtuzumab ozogamicin (GO) validate this approach. Partly stimulated by the success of GO, several CD33-targeted therapeutics are currently in early phase testing. AREAS COVERED CD33-targeted therapeutics in clinical development include Fc-engineered unconjugated antibodies (BI 836858 [mAb 33.1]), ADCs (SGN-CD33A [vadastuximab talirine], IMGN779), radioimmunoconjugates (225Ac-lintuzumab), bi- and trispecific antibodies (AMG 330, AMG 673, AMV564, 161533 TriKE fusion protein), and chimeric antigen receptor (CAR)-modified immune effector cells. Besides limited data on 225Ac-lintuzumab showing modest single-agent activity, clinical data are so far primarily available for SGN-CD33A. SGN-CD33A has single-agent activity and has shown encouraging results when combined with an azanucleoside or standard chemotherapeutics. However, concerns about toxicity to the liver and normal hematopoietic cells - the latter leading to early termination of a phase 3 trial - have derailed the development of SGN-CD33A, and its future is uncertain. EXPERT OPINION Early results from a new generation of CD33-targeted therapeutics are anticipated in the next 2-3 years. Undoubtedly, re-approval of GO in 2017 has changed the landscape and rendered clinical development for these agents more challenging.
Collapse
Affiliation(s)
- Roland B Walter
- a Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA.,b Department of Medicine, Division of Hematology , University of Washington , Seattle , WA , USA.,c Department of Epidemiology , University of Washington , Seattle , WA , USA
| |
Collapse
|
19
|
Yang D, Zhang X, Zhang X, Xu Y. The progress and current status of immunotherapy in acute myeloid leukemia. Ann Hematol 2017; 96:1965-1982. [PMID: 29080982 DOI: 10.1007/s00277-017-3148-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/02/2017] [Indexed: 02/08/2023]
Abstract
Recently, there has been remarkable progress in basic and preclinical studies of acute myeloid leukemia (AML). The improved outcomes of AML can largely be attributed to advances in supportive care and hematopoietic cell transplantation as opposed to conventional chemotherapy. However, as the 5-year survival rate remains low due to a high incidence of relapse, novel and effective treatments are urgently needed. Increasing attention is focusing on identifying suitable immunotherapeutic strategies for AML. Here, we describe the immunological features, mechanisms of immune escape, and recent progress in immunotherapy for AML. Problems encountered in the clinic will also be discussed. Although current outcomes may be limited, ongoing preclinical or clinical efforts are aimed at improving immunotherapy modalities and designing novel therapies, such as vaccines, monoclonal antibody therapy, chimeric antibody receptor-engineered T cells (CAR-T), TCR-engineered T cells (TCR-T), and checkpoint inhibitors, which may provide promising and effective therapies with higher specificity and efficacy for AML.
Collapse
Affiliation(s)
- Dan Yang
- Department of Hematology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Xiuqun Zhang
- Department of Hematology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Xuezhong Zhang
- Department of Hematology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Yanli Xu
- Department of Hematology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| |
Collapse
|
20
|
Allogeneic hematopoietic cell transplantation in patients with AML not achieving remission: potentially curative therapy. Bone Marrow Transplant 2017; 52:1083-1090. [DOI: 10.1038/bmt.2017.8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/29/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022]
|
21
|
Sinusoidal obstruction syndrome following CD33-targeted therapy in acute myeloid leukemia. Blood 2017; 129:2330-2332. [PMID: 28153826 DOI: 10.1182/blood-2017-01-762419] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
22
|
Aghevlian S, Boyle AJ, Reilly RM. Radioimmunotherapy of cancer with high linear energy transfer (LET) radiation delivered by radionuclides emitting α-particles or Auger electrons. Adv Drug Deliv Rev 2017; 109:102-118. [PMID: 26705852 DOI: 10.1016/j.addr.2015.12.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/26/2015] [Accepted: 12/06/2015] [Indexed: 12/31/2022]
Abstract
Radioimmunotherapy (RIT) aims to selectively deliver radionuclides emitting α-particles, β-particles or Auger electrons to tumors by conjugation to monoclonal antibodies (mAbs) that recognize tumor-associated antigens/receptors. The approach has been most successful for treatment of non-Hodgkin's B-cell lymphoma but challenges have been encountered in extending these promising results to the treatment of solid malignancies. These challenges include the low potency of β-particle emitters such as 131I, 177Lu or 90Y which have been commonly conjugated to the mAbs, due to their low linear energy transfer (LET=0.1-1.0keV/μm). Furthermore, since the β-particles have a 2-10mm range, there has been dose-limiting non-specific toxicity to hematopoietic stem cells in the bone marrow (BM) due to the cross-fire effect. Conjugation of mAbs to α-particle-emitters (e.g. 225Ac, 213Bi, 212Pb or 211At) or Auger electron-emitters (e.g. 111In, 67Ga, 123I or 125I) would increase the potency of RIT due to their high LET (50-230keV/μm and 4 to 26keV/μm, respectively). In addition, α-particles have a range in tissues of 28-100μm and Auger electrons are nanometer in range which greatly reduces or eliminates the cross-fire effect compared to β-particles, potentially reducing their non-specific toxicity to the BM. In this review, we describe the results of preclinical and clinical studies of RIT of cancer using radioimmunoconjugates emitting α-particles or Auger electrons, and discuss the potential of these high LET forms of radiation to improve the outcome of cancer patients.
Collapse
Affiliation(s)
- Sadaf Aghevlian
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Amanda J Boyle
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute and Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
23
|
Masarova L, Kantarjian H, Garcia-Mannero G, Ravandi F, Sharma P, Daver N. Harnessing the Immune System Against Leukemia: Monoclonal Antibodies and Checkpoint Strategies for AML. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 995:73-95. [PMID: 28321813 DOI: 10.1007/978-3-319-53156-4_4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute myeloid leukemia (AML) is the most common leukemia among adults and is associated with a poor prognosis, especially in patients with adverse prognostic factors, older age, or relapsed disease. The last decade has seen a surge in successful immune-based therapies in various solid tumors; however, the role of immune therapies in AML remains poorly defined. This chapter describes the rationale, clinical data, and toxicity profiles of immune-based therapeutic modalities in AML including naked and conjugated monoclonal antibodies, bispecific T-cell engager antibodies, chimeric antigen receptor (CAR)-T cells, and checkpoint blockade via blockade of PD1/PDL1 or CTLA4. Monoclonal antibodies commonly used in AML therapy target highly expressed "leukemia" surface antigens and include (1) naked antibodies against common myeloid markers such as anti-CD33 (e.g., lintuzumab), (2) antibody-drug conjugates linked to either, (a) a highly potent toxin such as calicheamicin, pyrrolobenzodiazepine, maytansine, or others in various anti-CD33 (gemtuzumab ozogamicin, SGN 33A), anti-123 (SL-401), and anti-CD56 (lorvotuzumab mertansine) formulations, or (b) radioactive particles, such as 131I, 213Bi, or 225Ac-labeled anti-CD33 or CD45 antibodies. Novel monoclonal antibodies that recruit and promote proximity-induced cytotoxicity of tumor cells by T cells (bispecific T-cell engager [BiTE] such as anti CD33/CD3, e.g., AMG 330) or block immune checkpoint pathways such as CTLA4 (e.g., ipilimumab) or PD1/PD-L1 (e.g., nivolumab) unleashing the patients T cells to fight leukemic cells are being evaluated in clinical trials in patients with AML. The numerous ongoing clinical trials with immunotherapies in AML will improve our understanding of the biology of AML and allow us to determine the best approaches to immunotherapy in AML.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibody Specificity
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Humans
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Molecular Targeted Therapy
- Signal Transduction/drug effects
- T-Lymphocytes/microbiology
- T-Lymphocytes/transplantation
- Tumor Microenvironment
Collapse
Affiliation(s)
- Lucia Masarova
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Hagop Kantarjian
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | | | - Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Padmanee Sharma
- Immunotherapy Platform, MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
|
25
|
Hagemann UB, Wickstroem K, Wang E, Shea AO, Sponheim K, Karlsson J, Bjerke RM, Ryan OB, Cuthbertson AS. In Vitro and In Vivo Efficacy of a Novel CD33-Targeted Thorium-227 Conjugate for the Treatment of Acute Myeloid Leukemia. Mol Cancer Ther 2016; 15:2422-2431. [PMID: 27535972 DOI: 10.1158/1535-7163.mct-16-0251] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/02/2016] [Indexed: 11/16/2022]
Abstract
The clinical efficacy of the first approved alpha pharmaceutical, Xofigo (radium-223 dichloride, 223RaCl2), has stimulated significant interest in the development of new alpha-particle emitting drugs in oncology. Unlike radium-223 (223Ra), the parent radionuclide thorium-227 (227Th) is able to form highly stable chelator complexes and is therefore amenable to targeted radioimmunotherapy. We describe the preparation and use of a CD33-targeted thorium-227 conjugate (CD33-TTC), which binds to the sialic acid receptor CD33 for the treatment of acute myeloid leukemia (AML). A chelator was conjugated to the CD33-targeting antibody lintuzumab via amide bonds, enabling radiolabeling with the alpha-emitter 227Th. The CD33-TTC induced in vitro cytotoxicity on CD33-positive cells, independent of multiple drug resistance (MDR) phenotype. After exposure to CD33-TTC, cells accumulated DNA double-strand breaks and were arrested in the G2 phase of the cell cycle. In vivo, the CD33-TTC demonstrated antitumor activity in a subcutaneous xenograft mouse model using HL-60 cells at a single dose regimen. Dose-dependent significant survival benefit was further demonstrated in a disseminated mouse tumor model after single dose injection or administered as a fractionated dose. The data presented support the further development of the CD33-TTC as a novel alpha pharmaceutical for the treatment of AML. Mol Cancer Ther; 15(10); 2422-31. ©2016 AACR.
Collapse
Affiliation(s)
| | | | - Ellen Wang
- Thorium Conjugate Research, Bayer AS, Oslo, Norway
| | - Adam O Shea
- Thorium Conjugate Research, Bayer AS, Oslo, Norway
| | | | | | | | - Olav B Ryan
- Thorium Conjugate Research, Bayer AS, Oslo, Norway
| | | |
Collapse
|
26
|
Bethge WA, Sandmaier BM. Targeted Cancer Therapy Using Radiolabeled Monoclonal Antibodies. Technol Cancer Res Treat 2016; 4:393-405. [PMID: 16029058 DOI: 10.1177/153303460500400407] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Radioimmunotherapy (RIT) combines the advantages of targeted radiation therapy and specific immunotherapy using monoclonal antibodies. RIT can be used either to target tumor cells or to specifically suppress immunocompetent host cells in the setting of allogeneic transplantation. The choice of radionuclide used for RIT depends on its distinct radiation characteristics and the type of malignancy or cells targeted. Beta-emitters with their lower energy and longer path length are more suitable to target bulky, solid tumors whereas α-emitters with their high linear energy transfer and short path length are better suited to target hematopoietic cells (normal or malignant). Different approaches of RIT such as the use of stable radioimmunoconjugates or of pretargeting strategies are available. Encouraging results have been obtained with RIT in patients with hematologic malignancies. The results in solid tumors are somewhat less favorable but new strategies for patients with minimal residual disease using adjuvant and locoregional treatment are evolving. This report outlines basic principles of RIT, gives an overview of available radionuclides and radioimmunoconjugates, and discusses clinical results with special emphasis on their use in hematologic malignancies including use in conditioning regimens for bone marrow transplantation.
Collapse
Affiliation(s)
- Wolfgang A Bethge
- Medical Center, University of Tuebingen, Department of Hematology and Oncology, Otfried-Mueller Str. 10, 72076 Tuebingen, Germany
| | | |
Collapse
|
27
|
Ali AM, Dehdashti F, DiPersio JF, Cashen AF. Radioimmunotherapy-based conditioning for hematopoietic stem cell transplantation: Another step forward. Blood Rev 2016; 30:389-99. [PMID: 27174151 DOI: 10.1016/j.blre.2016.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/16/2016] [Accepted: 04/26/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Alaa M Ali
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus 8058, St. Louis, MO 63110, USA.
| | - Farrokh Dehdashti
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA.
| | - John F DiPersio
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus 8058, St. Louis, MO 63110, USA.
| | - Amanda F Cashen
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus 8058, St. Louis, MO 63110, USA.
| |
Collapse
|
28
|
Kumar C, Shetake N, Desai S, Kumar A, Samuel G, Pandey BN. Relevance of radiobiological concepts in radionuclide therapy of cancer. Int J Radiat Biol 2016; 92:173-86. [PMID: 26917443 DOI: 10.3109/09553002.2016.1144944] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Radionuclide therapy (RNT) is a rapidly growing area of clinical nuclear medicine, wherein radionuclides are employed to deliver cytotoxic dose of radiation to the diseased cells/tissues. During RNT, radionuclides are either directly administered or delivered through biomolecules targeting the diseased site. RNT has been clinically used for diverse range of diseases including cancer, which is the focus of the review. CONCLUSIONS The major emphasis in RNT has so far been given towards developing peptides/antibodies and other molecules to conjugate a variety of therapeutic radioisotopes for improved targeting/delivery of radiation dose to the tumor cells. Despite that, many of the RNT approaches have not achieved their desired therapeutic success probably due to poor knowledge about complex and dynamic (i) fate of radiolabeled molecules; (ii) radiation dose delivered; (iii) cellular heterogeneity in tumor mass; and (iv) cellular radiobiological response. Based on understanding gathered during recent years, it may be stated that besides the absorbed dose, the net radiobiological response of tumor/normal cells also determines the clinical response of radiotherapeutic modalities including RNT. The radiosensitivity of tumor/normal cells is governed by radiobiological phenomenon such as radiation-induced bystander effect, genomic instability, adaptive response and low dose hyper-radiosensitivity. These concepts have been well investigated in the context of external beam radiotherapy, but their clinical implications during RNT have received meagre attention. In this direction, a few studies performed using in vitro and in vivo models envisage the possibilities of exploiting the radiobiological knowledge for improved therapeutic outcome of RNT.
Collapse
Affiliation(s)
- Chandan Kumar
- a Radiopharmaceutical Chemistry Section , Bhabha Atomic Research Centre , Mumbai
| | - Neena Shetake
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai
| | - Sejal Desai
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Amit Kumar
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Grace Samuel
- c Isotope Production and Applications Division , Bhabha Atomic Research Centre , Mumbai
| | - Badri N Pandey
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| |
Collapse
|
29
|
Jurcic JG, Rosenblat TL. Targeted alpha-particle immunotherapy for acute myeloid leukemia. Am Soc Clin Oncol Educ Book 2015:e126-31. [PMID: 24857092 DOI: 10.14694/edbook_am.2014.34.e126] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Because alpha-particles have a shorter range and a higher linear energy transfer (LET) compared with beta-particles, targeted alpha-particle immunotherapy offers the potential for more efficient tumor cell killing while sparing surrounding normal cells. To date, clinical studies of alpha-particle immunotherapy for acute myeloid leukemia (AML) have focused on the myeloid cell surface antigen CD33 as a target using the humanized monoclonal antibody lintuzumab. An initial phase I study demonstrated the safety, feasibility, and antileukemic effects of bismuth-213 ((213)Bi)-labeled lintuzumab. In a subsequent study, (213)Bi-lintuzumab produced remissions in some patients with AML after partial cytoreduction with cytarabine, suggesting the utility of targeted alpha-particle therapy for small-volume disease. The widespread use of (213)Bi, however, is limited by its short half-life. Therefore, a second-generation construct containing actinium-225 ((225)Ac), a radiometal that generates four alpha-particle emissions, was developed. A phase I trial demonstrated that (225)Ac-lintuzumab is safe at doses of 3 μCi/kg or less and has antileukemic activity across all dose levels studied. Fractionated-dose (225)Ac-lintuzumab in combination with low-dose cytarabine (LDAC) is now under investigation for the management of older patients with untreated AML in a multicenter trial. Preclinical studies using (213)Bi- and astatine-211 ((211)At)-labeled anti-CD45 antibodies have shown that alpha-particle immunotherapy may be useful as part conditioning before hematopoietic cell transplantation. The use of novel pretargeting strategies may further improve target-to-normal organ dose ratios.
Collapse
Affiliation(s)
- Joseph G Jurcic
- From the Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center; the New York-Presbyterian Hospital, and the Herbert Irving Comprehensive Cancer Center, New York, NY
| | - Todd L Rosenblat
- From the Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center; the New York-Presbyterian Hospital, and the Herbert Irving Comprehensive Cancer Center, New York, NY
| |
Collapse
|
30
|
Grosso DA, Hess RC, Weiss MA. Immunotherapy in acute myeloid leukemia. Cancer 2015; 121:2689-704. [PMID: 26095886 DOI: 10.1002/cncr.29378] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 01/23/2015] [Accepted: 02/09/2015] [Indexed: 11/08/2022]
Abstract
Despite the remarkable progress made in some leukemias such as CML and CLL, cytotoxic treatment for AML remains essentially unchanged over the last 4 decades. Several lines of evidence, including the graft versus leukemia effect associated with allogeneic hematopoietic stem cell transplantation (HSCT), suggest that immunotherapy is an active modality in AML. Given the lack of progress for chemotherapy in this disease, many novel immunologic treatment approaches have been explored. The goals of non-transplant-based immune approaches have largely consisted of the stimulation or restoration of endogenous immune responses or the targeting of specific tumor antigens by immune cells. These strategies have been associated with less toxicity than allogeneic HSCT but typically have inferior efficacy. Allogeneic HSCT exploits major and minor histocompatibility differences between the donor and recipient in order to recognize and eradicate malignancy. With the recognition that the immune system itself provides a basis for treating AML, immunotherapy continues to be an attractive modality to exploit in the treatment of this disease.
Collapse
Affiliation(s)
- Dolores A Grosso
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rosemary C Hess
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mark A Weiss
- Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Teiluf K, Seidl C, Blechert B, Gaertner FC, Gilbertz KP, Fernandez V, Bassermann F, Endell J, Boxhammer R, Leclair S, Vallon M, Aichler M, Feuchtinger A, Bruchertseifer F, Morgenstern A, Essler M. α-Radioimmunotherapy with ²¹³Bi-anti-CD38 immunoconjugates is effective in a mouse model of human multiple myeloma. Oncotarget 2015; 6:4692-703. [PMID: 25576914 PMCID: PMC4467108 DOI: 10.18632/oncotarget.2986] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/09/2014] [Indexed: 11/25/2022] Open
Abstract
In spite of development of molecular therapeutics, multiple myeloma (MM) is fatal in most cases. CD38 is a promising target for selective treatment of MM. We tested radioimmunoconjugates consisting of the α-emitter ²¹³Bi coupled to an anti-CD38 MAb in preclinical treatment of MM. Efficacy of ²¹³Bi-anti-CD38-MAb was assayed towards different MM cell lines with regard to induction of DNA double-strand breaks, induction of apoptosis and initiation of cell cycle arrest. Moreover, mice bearing luciferase-expressing MM xenografts were treated with ²¹³Bi-anti-CD38-MAb. Therapeutic efficacy was monitored by bioluminescence imaging, overall survival and histology. ²¹³Bi-anti-CD38-MAb treatment induced DNA damage which did not result in activation of the G2 DNA-damage-response checkpoint, but instead in mitotic arrest and subsequent mitotic catastrophe. The anti-tumor effect of ²¹³Bi-anti-CD38-MAb correlated with the expression level of CD38 in each MM cell line. In myeloma xenografts, treatment with ²¹³Bi-anti-CD38-MAb suppressed tumor growth via induction of apoptosis in tumor tissue and significantly prolonged survival compared to controls. The major organ systems did not show any signs of ²¹³Bi-induced toxicity. Preclinical treatment of MM with ²¹³Bi-anti-CD38-MAb turned out as an effective therapeutic option.
Collapse
Affiliation(s)
- Katharina Teiluf
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Christof Seidl
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
- Department of Obstetrics and Gynecology, Technische Universität München, Munich, Germany
| | - Birgit Blechert
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Florian C. Gaertner
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
- Department of Nuclear Medicine, Universitätsklinikum Bonn, Bonn, Germany
| | | | - Vanesa Fernandez
- III. Medical Department, Technische Universität München, Munich, Germany
| | - Florian Bassermann
- III. Medical Department, Technische Universität München, Munich, Germany
| | - Jan Endell
- MorphoSys AG, Martinsried/Planegg, Germany
| | | | | | - Mario Vallon
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
- Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michaela Aichler
- Institute of Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Institute for Transuranium Elements, Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Institute for Transuranium Elements, Karlsruhe, Germany
| | - Markus Essler
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
- Department of Nuclear Medicine, Universitätsklinikum Bonn, Bonn, Germany
| |
Collapse
|
32
|
Buckley SA, Walter RB. Antigen-specific immunotherapies for acute myeloid leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2015; 2015:584-595. [PMID: 26637776 DOI: 10.1182/asheducation-2015.1.584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Antigen-specific immunotherapies have emerged as important components of curative treatment algorithms for many cancers. In acute myeloid leukemia (AML), success has been less obvious. Nonetheless, among the few drugs shown to improve survival in recent randomized trials is the CD33 antibody-drug conjugate gemtuzumab ozogamicin. Significant antileukemic activity is also well documented for radioimmunoconjugates targeting CD33, CD45, or CD66. These therapeutics can intensify conditioning before hematopoietic cell transplantation, but their effect on patient outcomes needs clarification. Emerging data now suggest clinical antileukemic activity of several novel antibodies and perhaps some adoptive T-cell immunotherapies and vaccines. In parallel, numerous other agents targeting a wider variety of antigens are currently being explored. However, the antigenic heterogeneity characteristic of AML is a considerable limitation for all these therapeutics, and many important questions related to the ideal target antigen(s), disease situation in which to use these therapies, most suitable patient populations, exact treatment modalities, and details of supportive care needs remain open. Addressing such questions in upcoming studies will be required to ensure that antigen-directed therapies become an effective tool in AML, a disease for which outcomes with standard "3 + 7"-based chemotherapy have remained unsatisfactory in many patients.
Collapse
Affiliation(s)
| | - Roland B Walter
- Department of Medicine, Division of Hematology, and Department of Epidemiology, University of Washington, Seattle, WA; and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
33
|
Orozco JJ, Balkin ER, Gooley TA, Kenoyer A, Hamlin DK, Wilbur DS, Fisher DR, Hylarides MD, Shadman M, Green DJ, Gopal AK, Press OW, Pagel JM. Anti-CD45 radioimmunotherapy with 90Y but not 177Lu is effective treatment in a syngeneic murine leukemia model. PLoS One 2014; 9:e113601. [PMID: 25460570 PMCID: PMC4252056 DOI: 10.1371/journal.pone.0113601] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/24/2014] [Indexed: 11/19/2022] Open
Abstract
Radioimmunotherapy (RIT) for treatment of hematologic malignancies has primarily employed monoclonal antibodies (Ab) labeled with 131I or 90Y which have limitations, and alternative radionuclides are needed to facilitate wider adoption of RIT. We therefore compared the relative therapeutic efficacy and toxicity of anti-CD45 RIT employing 90Y and 177Lu in a syngeneic, disseminated murine myeloid leukemia (B6SJLF1/J) model. Biodistribution studies showed that both 90Y- and 177Lu-anti-murine CD45 Ab conjugates (DOTA-30F11) targeted hematologic tissues, as at 24 hours 48.8 ± 21.2 and 156 ± 14.6% injected dose per gram of tissue (% ID/g) of 90Y-DOTA-30F11 and 54.2 ± 9.5 and 199 ± 11.7% ID/g of 177Lu-DOTA-30F11 accumulated in bone marrow (BM) and spleen, respectively. However, 90Y-DOTA-30F11 RIT demonstrated a dose-dependent survival benefit: 60% of mice treated with 300 µCi 90Y-DOTA-30F11 lived over 180 days after therapy, and mice treated with 100 µCi 90Y-DOTA-30F11 had a median survival 66 days. 90Y-anti-CD45 RIT was associated with transient, mild myelotoxicity without hepatic or renal toxicity. Conversely, 177Lu- anti-CD45 RIT yielded no long-term survivors. Thus, 90Y was more effective than 177Lu for anti-CD45 RIT of AML in this murine leukemia model.
Collapse
Affiliation(s)
- Johnnie J. Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Hematology Division, University of Washington, Seattle, WA, United States of America
| | - Ethan R. Balkin
- Radiation Oncology, University of Washington, Seattle, WA, United States of America
| | - Ted A. Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Aimee Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Donald K. Hamlin
- Radiation Oncology, University of Washington, Seattle, WA, United States of America
| | - D. Scott Wilbur
- Radiation Oncology, University of Washington, Seattle, WA, United States of America
| | | | - Mark D. Hylarides
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Hematology Division, University of Washington, Seattle, WA, United States of America
| | - Damian J. Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Medical Oncology, University of Washington, Seattle, WA, United States of America
| | - Ajay K. Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Medical Oncology, University of Washington, Seattle, WA, United States of America
| | - Oliver W. Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Medical Oncology, University of Washington, Seattle, WA, United States of America
| | - John M. Pagel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Medical Oncology, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
34
|
Visani G, Malagola M, Guiducci B, Lucesole M, Loscocco F, Gabucci E, Paolini S, Piccaluga PP, Isidori A. Conditioning regimens in acute myeloid leukemia. Expert Rev Hematol 2014; 7:465-479. [PMID: 25025371 DOI: 10.1586/17474086.2014.939066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Current intensive consolidation chemotherapy for patients with acute myeloid leukemia (AML) produces median remission duration of 12-18 months, with less than 30% of patients surviving 5 years free of disease. Post-remission therapy is necessary to prevent relapse in most patients with AML; therefore, the aim of post-remission treatment is to eradicate the minimal residual disease. Nevertheless, the optimal form of treatment is still under debate. The choice among the possible approaches (intensive chemotherapy, autologous or allogeneic hematopoietic stem cell transplantation) relies on two main factors: the expected risk of relapse, as determined by biological features, and expected morbidity and mortality associated with a specific option. In this review, we focus on the different preparative regimens before autologous and allogeneic hematopoietic stem cell transplantation in patients with AML, stressing the importance of an adequate conditioning regimen as a mandatory element of a successful AML therapy, in both the allogeneic and the autologous transplant setting.
Collapse
|
35
|
The past and future of CD33 as therapeutic target in acute myeloid leukemia. Blood Rev 2014; 28:143-53. [DOI: 10.1016/j.blre.2014.04.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 02/05/2023]
|
36
|
Abstract
Radioimmunotherapy (RIT) represents an attractive strategy to deliver radiation selectively to tumor and other target organs while minimizing toxicity to normal tissues. RIT with β-particle-emitting isotopes targeting CD33, CD45 and CD66 can potentially allow intensification of conditioning before hematopoietic cell transplantation (HCT) in leukemia. Similarly, RIT directed against CD20 has shown promise in the setting of autologous and allogeneic HCT for B-cell lymphomas. α-particle immunotherapy with isotopes such as bismuth-213, actinium-225 and astatinine-211 offers the possibility of more selective and efficient killing of target cells while sparing the surrounding normal cells. Pretargeting strategies may further improve target:normal organ dose ratios. While RIT has demonstrated significant antitumor activity, ultimately, randomized studies will be required to determine if conditioning regimens that include this therapeutic modality can improve patient outcomes after HCT.
Collapse
Affiliation(s)
- Joseph G Jurcic
- Columbia University Medical Center, 177 Fort Washington Avenue, 6-435, New York, NY 10032, USA.
| |
Collapse
|
37
|
Abstract
CD33, a 67-kDa glycoprotein expressed on the majority of myeloid leukemia cells as well as on normal myeloid and monocytic precursors, has been an attractive target for monoclonal antibody (mAb)-based therapy of acute myeloid leukemia (AML). Lintuzumab, an unconjugated, humanized anti-CD33 mAb, has modest single-agent activity against AML but failed to improve patient outcomes in two randomized trials when combined with conventional chemotherapy. Gemtuzumab ozogamicin, an anti-CD33 mAb conjugated to the antitumor antibiotic calicheamicin, improved survival in a subset of AML patients when combined with standard chemotherapy, but safety concerns led to US marketing withdrawal. The activity of these agents confirms that CD33 remains a viable therapeutic target for AML. Strategies to improve the results of mAb-based therapies for AML include antibody engineering to enhance effector function, use of alternative drugs and chemical linkers to develop safer and more effective drug conjugates, and radioimmunotherapeutic approaches.
Collapse
Affiliation(s)
- Joseph G Jurcic
- Department of Medicine, Leukemia Service, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
38
|
Abstract
Conventional external-beam radiation therapy is dedicated to the treatment of localized disease, whereas radioimmunotherapy represents an innovative tool for the treatment of local or diffuse tumors. Radioimmunotherapy involves the administration of radiolabeled monoclonal antibodies that are directed specifically against tumor-associated antigens or against the tumor microenvironment. Although many tumor-associated antigens have been identified as possible targets for radioimmunotherapy of patients with hematological or solid tumors, clinical success has so far been achieved mostly with radiolabeled antibodies against CD20 ((131)I-tositumomab and (90)Y-ibritumomab tiuxetan) for the treatment of lymphoma. In this Review, we provide an update on the current challenges aimed to improve the efficacy of radioimmunotherapy and discuss the main radiobiological issues associated with clinical radioimmunotherapy.
Collapse
|
39
|
Leyton JV, Hu M, Gao C, Turner PV, Dick JE, Minden M, Reilly RM. Auger electron radioimmunotherapeutic agent specific for the CD123+/CD131- phenotype of the leukemia stem cell population. J Nucl Med 2011; 52:1465-73. [PMID: 21816968 DOI: 10.2967/jnumed.111.087668] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Our aim was to construct and characterize (111)In-nuclear translocation sequence (NLS)-7G3, an Auger electron-emitting radioimmunotherapeutic agent that preferentially recognizes the expression of CD123 (interleukin-3 receptor [IL-3R] α-subchain) in the absence of CD131 (IL-3R β-subchain) displayed by leukemia stem cells. METHODS Monoclonal antibody 7G3 was modified with 13-mer peptides [CGYGPKKKRKVGG] harboring the NLS of SV-40 large T-antigen and with diethylenetriaminepentaacetic acid for labeling with (111)In. Immunoreactivity was evaluated in a competition radioligand binding assay and by flow cytometry. Nuclear localization of (111)In-NLS-7G3 was studied by cell fractionation in CD123(+)/CD131(-) acute myelogenous leukemia (AML)-3, -4, and -5 cells or in primary AML or normal leukocytes. Micro-SPECT was performed in nonobese diabetic (NOD)/severe combined immune deficient (SCID) mice engrafted subcutaneously with Raji-CD123 tumors or with disseminated AML-3 or -5 cells. The cytotoxicity of (111)In-NLS-7G3 on AML-5 cells was studied after 7 d in culture by trypan blue dye exclusion. DNA damage was assessed using the γ-H2AX assay. RESULTS NLS-7G3 exhibited preserved CD123 immunoreactivity (affinity, 4.6 nmol/L). Nuclear importation of (111)In-NLS-7G3 in AML-3, -4, or -5 cells was specific and significantly higher than unmodified (111)In-7G3 and was greater in primary AML cells than in normal leukocytes. Rapid elimination of (111)In-NLS-7G3 in NOD/SCID mice prevented imaging of subcutaneous Raji-CD123 tumors. This phenomenon was Fc-dependent and IgG(2a) isotype-specific and was overcome by the preadministration of excess IgG(2a) or using (111)In-NLS-7G3 F(ab')(2) fragments. AML-3 and -5 cells were engrafted into the bone marrow or spleen or at extramedullary sites in NOD/SCID mice. Micro-SPECT/CT with (111)In-NLS-7G3 F(ab')(2) showed splenic involvement, whereas foci of disease were seen in the spine or femur or at extramedullary sites in the brain and lymph nodes using (111)In-NLS-7G3 IgG(2a). The viability of AML-5 cells was reduced by exposure in vitro to (111)In-NLS-7G3; this reduction was associated with an increase in unrepaired DNA double-strand breaks. CONCLUSION (111)In-NLS-7G3 is a promising novel Auger electron-emitting radioimmunotherapeutic agent for AML aimed at the leukemia stem cell population. Micro-SPECT/CT was useful for visualizing the engraftment of leukemia in NOD/SCID mice.
Collapse
Affiliation(s)
- Jeffrey Victor Leyton
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
40
|
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
|
41
|
Rosenblat TL, McDevitt MR, Mulford DA, Pandit-Taskar N, Divgi CR, Panageas KS, Heaney ML, Chanel S, Morgenstern A, Sgouros G, Larson SM, Scheinberg DA, Jurcic JG. Sequential cytarabine and alpha-particle immunotherapy with bismuth-213-lintuzumab (HuM195) for acute myeloid leukemia. Clin Cancer Res 2010; 16:5303-11. [PMID: 20858843 PMCID: PMC2970691 DOI: 10.1158/1078-0432.ccr-10-0382] [Citation(s) in RCA: 186] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Lintuzumab (HuM195), a humanized anti-CD33 antibody, targets myeloid leukemia cells and has modest single-agent activity against acute myeloid leukemia (AML). To increase the potency of the antibody without the nonspecific cytotoxicity associated with β-emitters, the α-particle-emitting radionuclide bismuth-213 ((213)Bi) was conjugated to lintuzumab. This phase I/II trial was conducted to determine the maximum tolerated dose (MTD) and antileukemic effects of (213)Bi-lintuzumab, the first targeted α-emitter, after partially cytoreductive chemotherapy. EXPERIMENTAL DESIGN Thirty-one patients with newly diagnosed (n = 13) or relapsed/refractory (n = 18) AML (median age, 67 years; range, 37-80) were treated with cytarabine (200 mg/m(2)/d) for 5 days followed by (213)Bi-lintuzumab (18.5-46.25 MBq/kg). RESULTS The MTD of (213)Bi-lintuzumab was 37 MB/kg; myelosuppression lasting >35 days was dose limiting. Extramedullary toxicities were primarily limited to grade ≤2 events, including infusion-related reactions. Transient grade 3/4 liver function abnormalities were seen in five patients (16%). Treatment-related deaths occurred in 2 of 21 (10%) patients who received the MTD. Significant reductions in marrow blasts were seen at all dose levels. The median response duration was 6 months (range, 2-12). Biodistribution and pharmacokinetic studies suggested that saturation of available CD33 sites by (213)Bi-lintuzumab was achieved after partial cytoreduction with cytarabine. CONCLUSIONS Sequential administration of cytarabine and (213)Bi-lintuzumab is tolerable and can produce remissions in patients with AML.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Alpha Particles/adverse effects
- Alpha Particles/therapeutic use
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antimetabolites, Antineoplastic/administration & dosage
- Antimetabolites, Antineoplastic/adverse effects
- Bismuth/adverse effects
- Bismuth/therapeutic use
- Cytarabine/administration & dosage
- Cytarabine/adverse effects
- Drug Administration Schedule
- Female
- Humans
- Immunoconjugates/adverse effects
- Immunoconjugates/therapeutic use
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/radiotherapy
- Male
- Middle Aged
- Radioimmunotherapy/adverse effects
- Radioimmunotherapy/methods
- Radioisotopes/adverse effects
- Radioisotopes/therapeutic use
- Remission Induction
Collapse
Affiliation(s)
- Todd L Rosenblat
- Department of Medicine and the Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
The Role of Monoclonal Antibodies in the Management of Leukemia. Pharmaceuticals (Basel) 2010. [PMCID: PMC4034091 DOI: 10.3390/ph3103258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This article will review the monoclonal antibodies more commonly used in leukemias. In the last three decades, scientists have made considerable progress understanding the structure and the functions of various surface antigens, such as CD20, CD33. The introduction of rituximab, an anti CD20 monoclonal antibody, had a great impact in the treatment of lymphoproliferative disorders. Gemtuzumab, an anti CD 33 conjugated monoclonal antibody has activity in acute mylegenous leukemia (AML). As this field is undergoing a rapid growth, the years will see an increasing use of monoclonal antibodies in hematological malignancies.
Collapse
|
43
|
Ball ED, Elizabeth Broome H. Monoclonal antibodies in the treatment of hematologic malignancy. Best Pract Res Clin Haematol 2010; 23:403-16. [DOI: 10.1016/j.beha.2010.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
44
|
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]
|
45
|
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
|
46
|
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
|
47
|
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
|
48
|
Radioimmunotherapy with [188Re]-labelled anti-CD66 antibody in the conditioning for allogeneic stem cell transplantation for high-risk acute myeloid leukemia. Int J Hematol 2008; 87:414-421. [DOI: 10.1007/s12185-008-0043-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 11/23/2007] [Accepted: 11/27/2007] [Indexed: 10/22/2022]
|
49
|
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
|
50
|
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]
|