1
|
Yang Y, Vedvyas Y, Alcaina Y, Son JY, Min IM, Jin MM. Low-dose targeted radionuclide therapy synergizes with CAR T cells and enhances tumor response. Front Immunol 2024; 15:1355388. [PMID: 38550578 PMCID: PMC10972862 DOI: 10.3389/fimmu.2024.1355388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/29/2024] [Indexed: 04/02/2024] Open
Abstract
Ionizing radiation has garnered considerable attention as a combination partner for immunotherapy due to its potential immunostimulatory effects. In contrast to the more commonly used external beam radiation, we explored the feasibility of combining chimeric antigen receptor (CAR) T cell therapy with targeted radionuclide therapy (TRT), which is achieved by delivering β-emitting 177Lu-DOTATATE to tumor via tumor-infiltrating CAR T cells that express somatostatin receptor 2 (SSTR2). We hypothesized that the delivery of radiation to tumors could synergize with CAR T therapy, resulting in enhanced antitumor immunity and tumor response. To determine the optimal dosage and timing of 177Lu-DOTATATE treatment, we measured CAR T cell infiltration and expansion in tumors longitudinally through positron emission tomography (PET) using a SSTR2-specific positron-emitting radiotracer,18F-NOTA-Octreotide. In animals receiving CAR T cells and a low-dose (2.5 Gy) of TRT following the administration of 177Lu-DOTATATE, we observed a rapid regression of large subcutaneous tumors, which coincided with a dramatic increase in serum proinflammatory cytokines. Tumor burden was also reduced when a higher radiation dose (6 Gy) was delivered to the tumor. However, this higher dose led to cell death in both the tumor and CAR T cells. Our study suggests that there may exist an optimum range of TRT dosage that can enhance T cell activity and sensitize tumor cells to T cell killing, which may result in more durable tumor control compared to a higher radiation dose.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Radiology, Houston Methodist Research Institute, Houston, TX, United States
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Yogindra Vedvyas
- Department of Radiology, Houston Methodist Research Institute, Houston, TX, United States
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Yago Alcaina
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Ju Y. Son
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Irene M. Min
- Department of Radiology, Houston Methodist Research Institute, Houston, TX, United States
- Department of Surgery, Weill Cornell Medicine, New York, NY, United States
| | - Moonsoo M. Jin
- Department of Radiology, Houston Methodist Research Institute, Houston, TX, United States
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
2
|
Gnesin S, Chouin N, Cherel M, Dunn SM, Schaefer N, Faivre-Chauvet A, Prior JO, Delage JA. From bench to bedside: 64Cu/ 177Lu 1C1m-Fc anti TEM-1: mice-to-human dosimetry extrapolations for future theranostic applications. EJNMMI Res 2023; 13:59. [PMID: 37314509 DOI: 10.1186/s13550-023-01010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023] Open
Abstract
The development of diagnostic and therapeutic radiopharmaceuticals is an hot topic in nuclear medicine. Several radiolabeled antibodies are under development necessitating both biokinetic and dosimetry extrapolations for effective human translation. The validation of different animal-to-human dosimetry extrapolation methods still is an open issue. This study reports the mice-to-human dosimetry extrapolation of 64Cu/177Lu 1C1m-Fc anti-TEM-1 for theranostic application in soft-tissue sarcomas. We adopt four methods; direct mice-to-human extrapolation (M1); dosimetry extrapolation considering a relative mass scaling factor (M2), application of a metabolic scaling factor (M3) and combination of M2 and M3 (M4). Predicted in-human dosimetry for the [64Cu]Cu-1C1m-Fc resulted in an effective dose of 0.05 mSv/MBq. Absorbed dose (AD) extrapolation for the [177Lu]Lu-1C1m-Fc indicated that the AD of 2 Gy and 4 Gy to the red-marrow and total-body can be reached with 5-10 GBq and 25-30 GBq of therapeutic activity administration respectively depending on applied dosimetry method. Dosimetry extrapolation methods provided significantly different absorbed doses in organs. Dosimetry properties for the [64Cu]Cu-1C1m-Fc are suitable for a diagnostic in-human use. The therapeutic application of [177Lu]Lu-1C1m-Fc presents challenges and would benefit from further assessments in animals' models such as dogs before moving into the clinic.
Collapse
Affiliation(s)
- Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Nicolas Chouin
- Inserm, CNRS, University of Angers, Oniris, CRCI2NA, University of Nantes, Nantes, France
| | - Michel Cherel
- CHU Nantes, CNRS, Inserm, CRCINA, University of Nantes, 44000, Nantes, France
| | - Steven Mark Dunn
- LAbCore, Ludwig Institute for Cancer Research, Lausanne University Hospital and University of Lausanne, 1066, Epalinges, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | | | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland.
| | - Judith Anna Delage
- Radiopharmacy Unit, Department of Pharmacy, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| |
Collapse
|
3
|
Radioiodine Therapy in Pediatric Differentiated Thyroid Cancer: Dosimetry, Clinical Care, and Future Challenges. Clin Nucl Med 2023; 48:158-167. [PMID: 36240802 DOI: 10.1097/rlu.0000000000004431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
ABSTRACT Thyroid cancer is very rare in children. 131 I therapy after thyroidectomy is established in pediatric differentiated thyroid cancer (DTC). Pediatric DTC guideline is silent on the optimum amount of 131 I that could be safely and effectively administered to children who are more radiosensitive. Like adult DTC, children are also given 131 I therapy empirically based either on age or body weight. Pediatric DTC guideline recommends that patient-specific dosimetry is important in children. Still, due to the low incidence rate and the practical difficulties of dosimetry, it has neither been established nor adopted in routine practice. This review article aims to discuss current approaches of 131 I therapy in children and young adult patients with DTC and dosimetric data obtained by several investigators. Efforts are required to simplify dosimetric procedures and precise results, especially in determining lesion size. We prefer 3-dimensional dosimetry over planar dosimetry, where lesion size could be measured accurately. 124 I PET/CT-based dosimetry is expected to give accurate dosimetric results. The most challenging aspect is that no randomized controlled trials are available to compare the empiric 131 I therapy results versus dosimetry-based treatment outcomes in children and young adults. Suppose dosimetry-based 131 I therapy could be shown to have better outcomes, namely, successful ablation rate, better disease-free survival, and lesser treatment-emergent adverse events than empirical 131 I treatment. In that case, one can argue in favor of the former. Unfortunately, no convincing study is currently available. Thus, there is a need for a randomized control trial to settle this issue.
Collapse
|
4
|
Farshbafnadi M, Razi S, Rezaei N. Transplantation. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
5
|
Pharmacological protection of the thyroid gland against radiation damage from radioactive iodine labeled compounds in children: a systematic review. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00529-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Abstract
Purpose
There is currently no consensus on which protective strategy is most effective to prevent I-131 uptake in the thyroid during medical interventions in children. We aimed to collect the best available evidence to determine which pharmacological intervention is most effective in protecting the thyroid gland from damage by radioactive iodine (RAI).
Methods
Literature searches were performed using PubMed, Embase, OLDMEDLINE, and the Cochrane Central Register of Controlled Trials. Only original studies were included (1950–2022). Studies comparing pharmacological prevention of the thyroid against RAI uptake or occurrence of hypothyroidism, thyroid nodule or thyroid cancer were included. Included studies were graded according to the Grading of Recommendations Assessment, Development and Evaluation considerations. Pharmacological interventions were compared for effectiveness on reduction of thyroidal intake or relevant clinical thyroidal outcomes.
Results
Forty studies were included. Quality of included studies was low and many different outcome variables were used, making meta-analysis impossible. In 81% of studies, the pharmacological intervention could not prevent RAI uptake or thyroid damage. The administration of potassium iodide (KI) 1 h before exposure to RAI seemed most effective to reduce thyroidal uptake, however, hypothyroidism was reported in up to 64% as well as several cases of thyroid carcinoma. The combination of KI, thyroxine and thiamazole reduced RAI uptake and occurrence of hypothyroidism; yet, after follow-up of 9 years, still 50% of patients developed hypothyroidism. KI with potassium perchlorate showed hypothyroidism to occur in up to 12% of patients after short follow-up time.
Conclusions
The lack of well-designed studies impairs making strong recommendations on the optimal way to prevent thyroid damage when using radioactive coupled ligands for medical interventions. To improve the protection of the thyroid against radiation damage by I-131, well-designed randomized clinical trials with sufficient follow-up time, comparing new protective strategies’ effects on valid and well-defined thyroid outcomes are needed.
Collapse
|
6
|
Saha A, Blazar BR. Antibody based conditioning for allogeneic hematopoietic stem cell transplantation. Front Immunol 2022; 13:1031334. [PMID: 36341432 PMCID: PMC9632731 DOI: 10.3389/fimmu.2022.1031334] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/10/2022] [Indexed: 08/25/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapeutic option for many patients with hematological malignancies and nonmalignant hematopoietic disorders. To achieve stable engraftment of donor hematopoietic stem cells (HSCs), recipient HSC deletion is needed to create space for incoming donor HSCs and donor HSCs must escape immune rejection by the recipient. Conventional allo-HSCT requires high dose of irradiation and/or chemotherapy to produce sufficient host stem cell and immune system ablation to permit donor HSC engraftment. However, these procedures also result in nonspecific tissue injury that can cause short- and long-term adverse effects as well as incite and amplify graft-versus-host-disease (GVHD). The delivery of targeted radiotherapy to hematopoietic tissues with the use of a radioimmunoconjugate (ROIC) as a part of transplant preparative regimen has shown clinical benefits. ROIC clinical data provide evidence for decreased relapse without increased transplant-related mortality by delivering higher targeted radiation to sites of malignancy than when given in a nontargeted fashion. An alternative approach to allo-HSCT has been developed and tested in preclinical mouse models in which nonmyeloablative preconditioning with low dose of the alkylating agent (busulfan) or lower systemic dose of irradiation combined with co-stimulatory pathway blockade (CTLA4-Ig, anti-CD40L monoclonal antibody) and/or immunosuppressive drugs have been used. Under these conditions, mixed chimerism and transplantation tolerance to fully MHC mismatched donor marrow was observed. Recently, several novel proof-of-concept antibody-mediated preconditioning methods have been developed that can selectively target hematopoietic stem and immune cells with minimal overall toxicity. Antibody-drug-conjugate (ADC) combined with reduced intensity conditioning or high dose ADC as single dose monotherapy have shown promise for allo-HSCT in preclinical models. The purpose of the current review is to discuss the literature exploring antibody-based conditioning that includes native antibody, radiolabeled antibody conjugates, and ADC for allo-HSCT.
Collapse
Affiliation(s)
- Asim Saha
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Bruce R. Blazar
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
7
|
Cicone F, Sarnelli A, Guidi C, Belli ML, Ferrari ME, Wahl R, Cremonesi M, Paganelli G. Dosimetric Approaches for Radioimmunotherapy of Non-Hodgkin Lymphoma in Myeloablative Setting. Semin Nucl Med 2022; 52:191-214. [PMID: 34996594 DOI: 10.1053/j.semnuclmed.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Radioimmunotherapy (RIT) is a safe and active treatment available for non-Hodgkin lymphomas (NHLs). In particular, two monoclonal antibodies raised against CD20, that is Zevalin (90Y-ibritumomab-tiuxetan) and Bexxar (131I-tositumomab) received FDA approval for the treatment of relapsing/refractory indolent or transformed NHLs. RIT is likely the most effective and least toxic anticancer agent in NHLs. However, its use in the clinical setting is still debated and, in case of relapse after optimized rituximab-containing regimens, the efficacy of RIT at standard dosage is suboptimal. Thus, clinical trials were based on the hypothesis that the inclusion of RIT in myeloablative conditioning would allow to obtain improved efficacy and toxicity profiles when compared to myeloablative total-body irradiation and/or high-dose chemotherapy regimens. Standard-activity RIT has a safe toxicity profile, and the utility of pretherapeutic dosimetry in this setting can be disputed. In contrast, dose-escalation clinical protocols require the assessment of radiopharmaceutical biodistribution and dosimetry before the therapeutic injection, as dose constrains for critical organs may be exceeded when RIT is administered at high activities. The aim of the present study was to review and discuss the internal dosimetry protocols that were adopted for non-standard RIT administration in the myeloablative setting before hematopoietic stem cell transplantation in patients with NHLs.
Collapse
Affiliation(s)
- Francesco Cicone
- Department of Experimental and Clinical Medicine, and Neuroscience Research Centre, PET/RM Unit, "Magna Graecia" University of Catanzaro, Catanzaro, Italy; Nuclear Medicine Unit, University Hospital "Mater Domini", Catanzaro, Italy
| | - Anna Sarnelli
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Claretta Guidi
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maria Luisa Belli
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | - Richard Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Marta Cremonesi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanni Paganelli
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
8
|
Wahl RL, Sgouros G, Iravani A, Jacene H, Pryma D, Saboury B, Capala J, Graves SA. Normal-Tissue Tolerance to Radiopharmaceutical Therapies, the Knowns and the Unknowns. J Nucl Med 2021; 62:23S-35S. [PMID: 34857619 DOI: 10.2967/jnumed.121.262751] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/15/2021] [Indexed: 12/25/2022] Open
Affiliation(s)
- Richard L Wahl
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| | - George Sgouros
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Amir Iravani
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| | | | - Daniel Pryma
- Penn Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Jacek Capala
- National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
9
|
El Fakih R, Lazarus HM, Muffly L, Altareb M, Aljurf M, Hashmi SK. Historical perspective and a glance into the antibody-based conditioning regimens: A new era in the horizon? Blood Rev 2021; 52:100892. [PMID: 34674852 DOI: 10.1016/j.blre.2021.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/10/2021] [Accepted: 10/11/2021] [Indexed: 11/17/2022]
Abstract
The hematopoietic cell transplantation practice has changed significantly over the years. More than 1500 centers around the globe are offering transplant for different types of diseases. This growth was driven by improving the efficacy and the safety of the procedure and the ability to use alternate donors. These improvements made the procedure feasible in virtually all patients in need for it. With the availability of novel therapies and targeted agents, we may be witnessing a new transplant-era. These agents may help to circumvent some of the remaining limitations of the procedure and open the doors for new indications. Herein, we review historical transplant milestones, the accomplishments that led to the modern transplant practice and we discuss the idea of minimal-intensity conditioning and the possibility to adopt chemotherapy and radiation-free preparative regimens in the near future.
Collapse
Affiliation(s)
- Riad El Fakih
- Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| | - Hillard M Lazarus
- Division of Hematology-Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Lori Muffly
- Stanford University, Blood and Marrow Transplant and Cellular therapy, Stanford, CA, USA
| | - Majed Altareb
- Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mahmoud Aljurf
- Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Shahrukh K Hashmi
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, UAE; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
10
|
Fisher DR. Perspectives on Internal Dosimetry for Optimized Radionuclide Therapy. Cancer Biother Radiopharm 2021; 37:161-163. [PMID: 34569812 DOI: 10.1089/cbr.2021.0318] [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: 11/12/2022] Open
Abstract
Abstract A balanced approach to radiopharmaceutical dosimetry involves personalized dosimetry. Planar quantitative imaging can be practical, reliable, and relatively cost-effective. Therapy dose optimization can be achieved for the individual patient using a straightforward tracer study to determine patient-specific biokinetics at three or more imaging time points for organs that assimilate the radiopharmaceutical. Two-dimensional quantitative imaging may be supported and calibrated using a 3D SPECT/CT measurement for the dose-limiting organ at a single time point. Organ volumes are needed from CT images. Measurements require special attention for consistency in camera-to-patient distancing, region-of-interest delineation, and attenuation correction, and operators need training and experience well beyond the requirements for standard nuclear medicine scintigraphy. As with external beam therapy, reimbursement codes are needed to support treatment-planning costs. Postinfusion tumor dosimetry can be important in overall evaluation of radionuclide therapy effectiveness. Clinicians and pharmaceutical companies should recognize the value of a balanced approach to personalized internal dosimetry for maximizing therapy benefit while minimizing toxicity. Prospective clinical trials should employ quantitative dosimetry with standardized methodologies to deliver predictive paradigms and establish the efficacy of new radioimmunotherapy products.
Collapse
Affiliation(s)
- Darrell R Fisher
- Washington State University and Versant Medical Physics and Radiation Safety, Richland, Washington, USA
| |
Collapse
|
11
|
Zhang Y, Kupferschlaeger J, Lang P, Reischl G, Handgretinger R, la Fougere C, Dittmann H. 131Iodine-GD2-ch14.18 scintigraphy to evaluate option for radioimmunotherapy in patients with advanced tumors. J Nucl Med 2021; 63:205-211. [PMID: 34049985 DOI: 10.2967/jnumed.120.261854] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/21/2021] [Indexed: 11/16/2022] Open
Abstract
The tumor-selective ganglioside antigene GD2 is frequently expressed on neuroblastomas and to a lesser extent also on sarcomas and neuroendocrine tumors. Aim of our study was to evaluate tumor targeting and biodistribution of iodine-131-labeled chimeric GD2-antibody clone 14/18 (131I-GD2-ch14.18) in patients with late-stage disease in order to identify eligibility for radioimmunotherapy. Methods: 20 patients (neuroblastoma n = 9; sarcoma n = 9; pheochromocytoma n = 1, neuroendocrine tumor n = 1) were involved in this study. 21 to 131 MBq (1-2 MBq/kg) of I-131-GD2-ch14.18 (0.5 -1.0 mg) were injected intravenously. Planar scintigraphy was performed within 1 h from injection (d0), on d1, d2, d3, and d6 or d7 to analyse tumor uptake and tracer biodistribution. Serial blood samples were collected in 4 individuals. Irradiation dose to tumor lesions and organs was calculated using Olinda® software. Results: The tumor targeting rate on a per-patient base was 65% (13/20) with 6/9 neuroblastomas showing uptake of I-GD2-ch14.18. Tumor lesions showed maximum uptake at 20-64 h p.i. (effective half-life in tumors 33-192 h). The tumor irradiation dose varied between 0.52 and 30.2 mGy/MBq (median: 9.08, n = 13). Visual analysis showed prominent blood pool activity up to d2/d3 p.i.. No pronounced uptake was observed in the bone marrow compartment or in the kidneys. Bone marrow dose was calculated at 0.07-0.47 mGy/MBq (median: 0.14) while blood dose was 1.1-4.7 mGy/MBq. Two patients (1 neuroblastoma and 1 pheochromocytoma) with particularly high tumor uptake underwent radioimmunotherapy using 2.3 and 2.9 GBq of I-GD2-ch14.18 both achieving stable disease. Overall survival was 17 and 6 months, respectively. Conclusion: I-GD2-ch14.18 is cleared slowly from blood resulting in good tumor to background contrast not until 2 d after application. With acceptable red marrow and organ dose, radioimmunotherapy is an option for patients with high tumor uptake. However, due to the variable GD2-expression, decision should be made depending on pretherapeutic dosimetry.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Germany
| | - Juergen Kupferschlaeger
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Germany
| | - Peter Lang
- Clinic for Paediatric Hematology and Oncology, University Hospital Tuebingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, Germany
| | - Rupert Handgretinger
- Clinic for Paediatric Hematology and Oncology, University Hospital Tuebingen, Germany
| | - Christian la Fougere
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Germany
| | - Helmut Dittmann
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Germany
| |
Collapse
|
12
|
Sabanathan D, Campbell DH, Velonas VM, Wissmueller S, Mazure H, Trifunovic M, Poursoltan P, Ho Shon K, Mackay TR, Lund ME, Lu Y, Roach PJ, Bailey DL, Walsh BJ, Gillatt D, Gurney H. Safety and tolerability of Miltuximab ® - a first in human study in patients with advanced solid cancers. ASIA OCEANIA JOURNAL OF NUCLEAR MEDICINE & BIOLOGY 2021; 9:86-100. [PMID: 34250138 PMCID: PMC8255523 DOI: 10.22038/aojnmb.2021.55600.1386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 01/31/2023]
Abstract
OBJECTIVES Miltuximab® is a chimeric antibody targeting Glypican-1 (GPC-1), a cell surface antigen which is overexpressed in solid cancers. Miltuximab® has shown promising safety and efficacy in radioimmunotherapy models of prostate cancer. This first in human study used Miltuximab® radiolabelled with Gallium-67 ([67Ga]Ga-DOTA-Miltuximab®). The primary study endpoint was to establish safety and tolerability of Miltuximab®. Secondary endpoints were biodistribution, tumour targeting and pharmacokinetic analysis. METHODS Four cohorts of three patients (9 with advanced prostate cancer, 2 with pancreatic and 1 with bladder cancer) were dosed with 1 mg, ~250 MBq of [67Ga]Ga-DOTA-Miltuximab®. Cohort 1 received [67Ga]Ga-DOTA-Miltuximab® alone, while cohorts 2-4 were pre-infused with increasing doses (3.5, 11.5 and 24 mg, respectively) of unlabelled Miltuximab®-DOTA 1 hour prior to [67Ga]Ga-DOTA-Miltuximab®. Safety and tolerability were assessed by clinical and standard laboratory assessments. Patients underwent whole body gamma-camera scans and SPECT/CT scans up to 144 h post-infusion. Total organ radiation exposure was determined by dosimetry of whole-body gamma scans. RESULTS The dosing regimen was well tolerated, with no drug-related adverse events observed. Liver and spleen uptake of [67Ga]Ga-DOTA-Miltuximab® was observed. Liver uptake was reduced by pre-infusion of unlabelled Miltuximab®-DOTA. Dosimetry analysis showed a favorable exposure profile. [67Ga]Ga-DOTA-Miltuximab® targeting to tumour sites was observed in two prostate cancer patients who had failed enzalutamide treatment. Higher doses of unlabelled antibody achieved lower liver uptake and increased antibody serum half life. CONCLUSIONS This study is the first in human for Miltuximab® a first in class antibody targeting GPC-1. The trial met its primary endpoint of safety, demonstrating its potential as a safe and tolerable monoclonal antibody. This safety data, together with targeting to tumour lesions and biodistribution information supports the further clinical development of Miltuximab® as a theranostic agent in a planned Phase I human trial.
Collapse
Affiliation(s)
- Dhanusha Sabanathan
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | | | - Vicki M. Velonas
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | | | - Hubert Mazure
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | | | - Pirooz Poursoltan
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Kevin Ho Shon
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | | | - Maria E. Lund
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | - Yanling Lu
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | | | | | - Bradley J. Walsh
- GlyTherix Ltd, 75 Talavera Road, Macquarie Park, Sydney, Australia
| | - David Gillatt
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Howard Gurney
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
13
|
Bobrowicz M, Kubacz M, Slusarczyk A, Winiarska M. CD37 in B Cell Derived Tumors-More than Just a Docking Point for Monoclonal Antibodies. Int J Mol Sci 2020; 21:ijms21249531. [PMID: 33333768 PMCID: PMC7765243 DOI: 10.3390/ijms21249531] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/20/2022] Open
Abstract
CD37 is a tetraspanin expressed prominently on the surface of B cells. It is an attractive molecular target exploited in the immunotherapy of B cell-derived lymphomas and leukemia. Currently, several monoclonal antibodies targeting CD37 as well as chimeric antigen receptor-based immunotherapies are being developed and investigated in clinical trials. Given the unique role of CD37 in the biology of B cells, it seems that CD37 constitutes more than a docking point for monoclonal antibodies, and targeting this molecule may provide additional benefit to relapsed or refractory patients. In this review, we aimed to provide an extensive overview of the function of CD37 in B cell malignancies, providing a comprehensive view of recent therapeutic advances targeting CD37 and delineating future perspectives.
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antineoplastic Agents, Immunological/therapeutic use
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Humans
- Immunotherapy/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Tetraspanins/immunology
- Tetraspanins/metabolism
Collapse
|
14
|
Kumar P, Damle NA, Agarwala S, Dwivedi SN, Bal C. Individualized dosimetry in children and young adults with differentiated thyroid cancer undergoing iodine-131 therapy. J Pediatr Endocrinol Metab 2020; 33:/j/jpem.ahead-of-print/jpem-2020-0072/jpem-2020-0072.xml. [PMID: 32651987 DOI: 10.1515/jpem-2020-0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/23/2020] [Indexed: 11/15/2022]
Abstract
Objectives The amount of Iodine-131 to treat young patients with differentiated thyroid cancer (DTC) has not been established so far. The purpose of this study was to perform and compare blood dosimetry by "Hanscheid's approach"and lesion dosimetry by "Maxon's approach". Methods Seventy-one DTC patients ≤21 years were given diagnostic activity of 74 MBq 131I followed by whole-body scan (WBS) at 2 h (pre-void), 24 h, 48 h, and ≥72 h. Pre-therapy blood and lesion dosimetry were conducted to determine the absorbed doses to blood and lesions and to predict the therapeutic activity. The administered activities were varied from 1.11-5.55 GBq of 131I depending on disease extent. Post therapy dosimetries were again performed by acquiring WBS data at 24 h, 48 h, and ≥72 h. Results In blood dosimetry, the difference between predicted therapy activity (PTA) and actual therapeutic activity (ATA) was statistically significant in remnant and lung lesions but insignificant in nodal metastases (p=0.287). In lesion dosimetry, the difference between PTA and ATA was statistically significant for lung metastasis patients; however, not significant in remnant (p=0.163) and nodal metastases (p=0.054). The difference between predicted and observed absorbed dose was insignificant in blood dosimetry whereas, significant in lesion dosimetry. Conclusions The PTA based on 0.3 Gy recommendations of Hanscheid et al. may be adequate for patients with remnant or nodal metastases but inadequate for lung metastases. Lesion dosimetry demonstrated that there is scope to decrease the 131I empiric ATA for remnant and nodal metastases; at the same time, there is scope to increase in lung metastasis patients.
Collapse
Affiliation(s)
- Praveen Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | | | - Sandeep Agarwala
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Sada Nand Dwivedi
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
15
|
Chow VA, Rajendran JG, Fisher DR, Appelbaum FR, Cassaday RD, Martin PS, Holmberg LA, Gooley TA, Stevenson PA, Pagel JM, Green DJ, Press OW, Gopal AK. A phase II trial evaluating the efficacy of high-dose Radioiodinated Tositumomab (Anti-CD20) antibody, etoposide and cyclophosphamide followed by autologous transplantation, for high-risk relapsed or refractory non-hodgkin lymphoma. Am J Hematol 2020; 95:775-783. [PMID: 32243637 DOI: 10.1002/ajh.25818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/18/2020] [Accepted: 03/28/2020] [Indexed: 01/23/2023]
Abstract
Radiation is the most effective treatment for localized lymphoma, but treatment of multifocal disease is limited by toxicity. Radioimmunotherapy (RIT) delivers tumoricidal radiation to multifocal sites, further augmenting response by dose-escalation. This phase II trial evaluated high-dose RIT and chemotherapy prior to autologous stem-cell transplant (ASCT) for high-risk, relapsed or refractory (R/R) B-cell non-Hodgkin lymphoma (NHL). The primary endpoint was progression free survival (PFS). Secondary endpoints were overall survival (OS), toxicity, and tolerability. Patients age < 60 years with R/R NHL expressing CD20 were eligible. Mantle cell lymphoma (MCL) patients could proceed to transplant in first remission. Patients received I-131-tositumomab delivered at ≤25Gy to critical normal organs, followed by etoposide, cyclophosphamide and ASCT. A group of 107 patients were treated including aggressive lymphoma (N = 29), indolent lymphoma (N = 45), and MCL (N = 33). After a median follow-up of 10.1 years, the 10-year PFS for the aggressive, indolent, and MCL groups were 62%, 64%, 43% respectively. The 10-year OS for the aggressive, indolent, and MCL groups were 61%, 71%, 48% respectively. Toxicities were similar to standard conditioning regimens and non-relapse mortality at 100 days was 2.8%. Late myeloid malignancies were seen in 6% of patients. High-dose I-131-tositumomab, etoposide and cyclophosphamide followed by ASCT appeared feasible, safe, and effective in treating NHL, with estimated PFS at 10-years of 43%-64%. In light of novel cellular therapies for R/R NHL, high-dose RIT-containing regimens yield comparable efficacy and safety and could be prospectively compared.
Collapse
Affiliation(s)
- Victor A Chow
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | - Frederick R Appelbaum
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ryan D Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Paul S Martin
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Leona A Holmberg
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Theodore A Gooley
- Public Health Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Philip A Stevenson
- Public Health Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - John M Pagel
- Center for Blood Disorders and Stem Cell Transplantation, Swedish Cancer Institute, Seattle, WA, USA
| | - Damian J Green
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Oliver W Press
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ajay K Gopal
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
16
|
Kahle XU, Montes de Jesus FM, Glaudemans AWJM, Lub-de Hooge MN, Jorritsma-Smit A, Plattel WJ, van Meerten T, Diepstra A, van den Berg A, Kwee TC, Noordzij W, de Vries EGE, Nijland M. Molecular imaging in lymphoma beyond 18F-FDG-PET: understanding the biology and its implications for diagnostics and therapy. LANCET HAEMATOLOGY 2020; 7:e479-e489. [PMID: 32470439 DOI: 10.1016/s2352-3026(20)30065-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/31/2020] [Accepted: 02/13/2020] [Indexed: 02/08/2023]
Abstract
Mature lymphoproliferative diseases are a heterogeneous group of neoplasms arising from different stages of B-cell and T-cell development. With improved understanding of the molecular processes in lymphoma and novel treatment options, arises a growing need for the molecular characterisation of tumours. Molecular imaging with single-photon-emission CT and PET using specific radionuclide tracers can provide whole-body information to investigate cancer biology, to evaluate phenotypic heterogeneity, to identify resistance to targeted therapy, and to assess the biodistribution of drugs in patients. In this Review, we evaluate the existing literature on molecular imaging in lymphoma, other than 18F-fluordeoxyglucose molecular imaging. The aim is to examine the contribution of molecular imaging to the understanding of the biology of lymphoma and to discuss potential implications for the diagnostics and therapy of this disease. Finally, we discuss possible applications for molecular imaging of patients with lymphoma in the clinical context.
Collapse
Affiliation(s)
- Xaver U Kahle
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Filipe M Montes de Jesus
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Annelies Jorritsma-Smit
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Wouter J Plattel
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tom van Meerten
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Thomas C Kwee
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Marcel Nijland
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
17
|
Dogra P, Butner JD, Nizzero S, Ruiz Ramírez J, Noureddine A, Peláez MJ, Elganainy D, Yang Z, Le AD, Goel S, Leong HS, Koay EJ, Brinker CJ, Cristini V, Wang Z. Image-guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1628. [PMID: 32314552 PMCID: PMC7507140 DOI: 10.1002/wnan.1628] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/06/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug-loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size-dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non-invasive in vivo imaging modalities. This allows for visualization and quantification of the whole-body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non-invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state-of-the-art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - María J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Applied Physics Graduate Program, Rice University, Houston, Texas, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhen Yang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anh-Dung Le
- Nanoscience and Microsystems Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - Shreya Goel
- Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
18
|
Conlon KC, Sportes C, Brechbiel MW, Fowler DH, Gress R, Miljkovic MD, Chen CC, Whatley MA, Bryant BR, Corcoran EM, Kurdziel KA, Pittaluga S, Paik CH, Lee JH, Fleisher TA, Carrasquillo JA, Waldmann TA. 90Y-Daclizumab (Anti-CD25), High-Dose Carmustine, Etoposide, Cytarabine, and Melphalan Chemotherapy and Autologous Hematopoietic Stem Cell Transplant Yielded Sustained Complete Remissions in 4 Patients with Recurrent Hodgkin's Lymphoma. Cancer Biother Radiopharm 2020; 35:249-261. [PMID: 32275165 DOI: 10.1089/cbr.2019.3298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: Despite advances in therapy of Hodgkin's lymphoma (HL), a proportion of patients will not respond or relapse. The authors had previously identified CD25, IL-2Rα, as a target for systemic radioimmunotherapy of HL since most normal cells do not express CD25, but it is expressed by a minority of Hodgkin/Reed-Sternberg (HRS) cells and most Tregs rosetting around HRS cells. Study Design and Treatment: This was a single institution, nonrandomized, open-label phase I/II trial of radiolabeled 90Y-daclizumab, an anti-CD25 monoclonal antibody, BEAM (carmustine, etoposide, cytarabine, and melphalan) conditioning treatment followed by autologous hematopoietic stem cell transplant (ASCT). Four patients with refractory and relapsed HL were treated in this trial with 3 patients receiving a single dose of 564.6-574.6 MBq 90Y-daclizumab and the fourth patient receiving two doses of 580.9-566.1 MBq 90Y-daclizumab followed by high-dose chemotherapy and ASCT. Results: All 4 evaluable patients treated with 90Y-daclizumab obtained complete responses (CRs) that are ongoing 4.5-7 years following their stem cell transplant. The spectrum and severity of adverse events were mild and more importantly none of the patients, including several with multiple therapies before this treatment, developed the myelodysplastic syndrome. Discussion: Targeting by daclizumab was not directed primarily at tumor cells, but rather the nonmalignant CD25-expressing T cells adjacent to the HRS cells and 90Y-daclizumab provided strong enough β emissions to kill CD25-negative tumor cells at a distance by a crossfire effect. Furthermore, the strong β irradiation killed normal cells in the tumor microenvironment. Conclusions: 90Y-daclizumab (anti-CD25), high-dose BEAM chemotherapy and ASCT was well tolerated and yielded sustained complete remissions in all 4 patients with recurrent HL patients who completed their treatment. Significance: Despite advances, a proportion of patients with HL will not have a CR to their initial treatment, and some with CRs will relapse. They demonstrated that the addition of 90Y-daclizumab into the preconditioning regimen for refractory and relapsed HL patients with high-dose BEAM chemotherapy and ASCT provided sustained CRs in the 4 patients studied. Two of these patients were highly refractory to multiple prior treatments with bulky disease at entry into this study, including 1 patient who never entered a remission and had failed 6 different therapeutic regimens. Despite the small number of patients treated in this study, the sustained clinical benefit in these patients indicates a highly effective treatment. The daclizumab was directed primarily not at HRS cells themselves but toward nonmalignant T cells rosetting around malignant cells. 90Y provided strong β emissions that killed antigen nonexpressing tumor cells at a distance by a crossfire effect. Furthermore, the strong β radiation killed normal cells in the tumor microenvironment that nurtured the malignant cells in the lymphomatous mass. The present study supports expanded analysis of 90Y-daclizumab as part of the regimen of ASCT in patients with refractory and relapsed HL.
Collapse
Affiliation(s)
- Kevin C Conlon
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Claude Sportes
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Martin W Brechbiel
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Ronald Gress
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Milos D Miljkovic
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Clara C Chen
- Nuclear Medicine Department, Radiation and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Millie A Whatley
- Nuclear Medicine Department, Radiation and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Bonita R Bryant
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Erin M Corcoran
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Karen A Kurdziel
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chang H Paik
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jae Ho Lee
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas A Fleisher
- Department of Laboratory Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Jorge A Carrasquillo
- Nuclear Medicine Department, Radiation and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Radioimmunotherapy. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
20
|
Detappe A, Reidy M, Yu Y, Mathieu C, Nguyen HVT, Coroller TP, Lam F, Jarolim P, Harvey P, Protti A, Nguyen QD, Johnson JA, Cremillieux Y, Tillement O, Ghobrial IM, Ghoroghchian PP. Antibody-targeting of ultra-small nanoparticles enhances imaging sensitivity and enables longitudinal tracking of multiple myeloma. NANOSCALE 2019; 11:20485-20496. [PMID: 31650133 DOI: 10.1039/c9nr06512a] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Monitoring malignant progression and disease recurrence post-therapy are central challenges to improving the outcomes of patients with multiple myeloma (MM). Whereas current detection methods that rely upon bone marrow examination allow for precise monitoring of minimal residual disease and can help to elucidate clonal evolution, they do not take into account the spatial heterogeneity of the tumor microenvironment. As such, they are uninformative as to the localization of malignant plasma cells and may lead to false negative results. With respect to the latter challenge, clinically-available imaging agents are neither sufficiently sensitive nor specific enough to detect minute plasma cell populations. Here, we sought to explore methods by which to improve detection of MM cells within their natural bone marrow environment, using whole-animal magnetic resonance imaging to longitudinally monitor early-stage disease as well as to enhance tumor detection after systemic therapy. We conducted a proof-of-concept study to demonstrate that ultra-small (<5 nm) gadolinium-containing nanoparticles bound to full-length antibodies against the B-cell maturation antigen (BCMA) exhibit rapid tumor uptake followed by renal clearance, improving the signal-to-noise ratio for MM detection beyond levels that are currently afforded by other FDA-approved clinical imaging modalities. We anticipate that when combined with bone marrow or blood biopsy, such imaging constructs could help to augment the effective management of patients with MM.
Collapse
Affiliation(s)
- Alexandre Detappe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. and Centre Paul Strauss, 3 rue de la porte de l'hôpital, 67000 Strasbourg, France
| | - Mairead Reidy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Yingjie Yu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | - Clelia Mathieu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Hung V-T Nguyen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Thibaud P Coroller
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Fred Lam
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. and Clinical Scholar Program, Division of Neurosurgery, McMaster University, 237 Barton St East, Hamilton General Hospital, Hamilton ON, L8L 2X2, Canada
| | - Petr Jarolim
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Peter Harvey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Andrea Protti
- Lurie Family Imaging Center, Department of Radiology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Department of Radiology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yannick Cremillieux
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR CNRS 5255, 33076 Bordeaux, France
| | - Olivier Tillement
- Institut Lumière Matière, UMR 5306 Université Lyon1-CNRS, Université de Lyon, 69622 Villeurbanne Cedex, France
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - P Peter Ghoroghchian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. and Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| |
Collapse
|
21
|
Eskian M, Khorasanizadeh M, Zinzani PL, Illidge TM, Rezaei N. Novel Methods to Improve the Efficiency of Radioimmunotherapy for Non-Hodgkin Lymphoma. Int Rev Immunol 2019; 38:79-91. [PMID: 30931651 DOI: 10.1080/08830185.2019.1588266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Radioimmunotherapy (RIT) is a novel strategy for treating non-Hodgkin lymphoma (NHL). Several studies have shown the promising results of using RIT in NHL, which have led to FDA approval for two RIT agents in treating low grade NHL. In spite of these favorable results in low-grade NHL, most of the aggressive or relapsed/refractory NHL subjects experience relapses following RIT. Although more aggressive treatments such as myeloablative doses of RIT followed by stem cell transplantation appear to be able to provide a longer survival for some patients these approaches are associated with significant treatment-related adverse events and challenging to deliver in most centers. Therefore, it seems reasonable to develop treatment approaches that enhance the efficiency of RIT, while reducing its toxicity. In this paper, novel methods that improve the efficiency of RIT and reduce its toxicity through various mechanisms are reviewed. Further clinical development of these methods could expand the NHL patient groups eligible for receiving RIT, and even extend the use of RIT to new indications and disease groups in future.
Collapse
Affiliation(s)
- Mahsa Eskian
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,b Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - MirHojjat Khorasanizadeh
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,b Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - Pier Luigi Zinzani
- c Institute of Hematology "L. e A. Seràgnoli", University of Bologna , Bologna , Italy
| | - Tim M Illidge
- d Manchester Academic Health Sciences Centre, University of Manchester, Christie NHS Foundation Trust , Manchester , UK
| | - Nima Rezaei
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,e Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,f Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| |
Collapse
|
22
|
Targeting the Tetraspanins with Monoclonal Antibodies in Oncology: Focus on Tspan8/Co-029. Cancers (Basel) 2019; 11:cancers11020179. [PMID: 30769765 PMCID: PMC6406856 DOI: 10.3390/cancers11020179] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/12/2022] Open
Abstract
Tetraspanins are exposed at the surface of cellular membranes, which allows for the fixation of cognate antibodies. Developing specific antibodies in conjunction with genetic data would largely contribute to deciphering their biological behavior. In this short review, we summarize the main functions of Tspan8/Co-029 and its role in the biology of tumor cells. Based on data collected from recently reported studies, the possibilities of using antibodies to target Tspan8 in immunotherapy or radioimmunotherapy approaches are also discussed.
Collapse
|
23
|
Li Y, Hamlin DK, Chyan MK, Wong R, Dorman EF, Emery RC, Woodle DR, Manger RL, Nartea M, Kenoyer AL, Orozco JJ, Green DJ, Press OW, Storb R, Sandmaier BM, Wilbur DS. cGMP production of astatine-211-labeled anti-CD45 antibodies for use in allogeneic hematopoietic cell transplantation for treatment of advanced hematopoietic malignancies. PLoS One 2018; 13:e0205135. [PMID: 30335787 PMCID: PMC6193629 DOI: 10.1371/journal.pone.0205135] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/19/2018] [Indexed: 11/28/2022] Open
Abstract
The objective of this study was to translate reaction conditions and quality control methods used for production of an astatine-211(211At)-labeled anti-CD45 monoclonal antibody (MAb) conjugate, 211At-BC8-B10, from the laboratory setting to cGMP production. Five separate materials were produced in the preparation of 211At-BC8-B10: (1) p-isothiocyanato-phenethyl-closo-decaborate(2-) (B10-NCS), (2) anti-CD45 MAb, BC8, (3) BC8-B10 MAb conjugate, (4) [211At]NaAt, and (5) 211At-BC8-B10. The 211At-labeling reagent, B10-NCS, was synthesized as previously reported. BC8 was produced, then conjugated with B10-NCS under cGMP conditions to form BC8-B10. [211At]NaAt was produced by α-irradiation of Bi targets, followed by isolation of the 211At using a “wet chemistry” method. The clinical product, 211At-BC8-B10, was prepared by reacting [211At]NaAt with BC8-B10 in NH4OAc buffer (pH 5.5) for 2 min at room temperature, followed by size-exclusion chromatography purification. Quality control tests conducted on the 211At-BC8-B10 included evaluations for purity and identity, as well as pyrogen and sterility tests. Stability of the 211At-BC8-B10 in 25 mg/mL sodium ascorbate solution was evaluated at 1, 2, 4, 6 and 21 h post isolation. For qualification, three consecutive 211At-BC8-B10 clinical preparations were successfully conducted in the cGMP suite, and an additional cGMP clinical preparation was carried out to validate each step required to deliver 211At-BC8-B10 to a patient. These cGMP preparations provided 0.80–1.28 Gbq (21.5–34.5 mCi) of 211At-BC8-B10 with radiochemical purity of >97%. The preparations were found to be sterile and have a pyrogen level <0.50 EU/mL. Cell binding was retained by the 211At-BC8-B10. 211At-BC8-B10 in ascorbic acid solution demonstrated a radiochemical stability of >95% for up to 21 h at room temperature. The experiments conducted have defined conditions for translation of 211At-BC8-B10 production from the laboratory to cGMP suite. This study has allowed the initiation of a phase I/II clinical trial using 211At-BC8-B10 (NCT03128034).
Collapse
Affiliation(s)
- Yawen Li
- Department of Radiation Oncology, University of Washington, Seattle, Washington, United States of America
| | - Donald K. Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, Washington, United States of America
| | - Ming-Kuan Chyan
- Department of Radiation Oncology, University of Washington, Seattle, Washington, United States of America
| | - Roger Wong
- Department of Radiation Oncology, University of Washington, Seattle, Washington, United States of America
| | - Eric F. Dorman
- Department of Radiation Oncology, University of Washington, Seattle, Washington, United States of America
| | - Robert C. Emery
- Department of Radiation Oncology, University of Washington, Seattle, Washington, United States of America
| | - Douglas R. Woodle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ronald L. Manger
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Margaret Nartea
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Aimee L. Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Johnnie J. Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Damian J. Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Oliver W. Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Brenda M. Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - D. Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
24
|
Stokke C, Blakkisrud J, Løndalen A, Dahle J, Martinsen ACT, Holte H, Kolstad A. Pre-dosing with lilotomab prior to therapy with 177Lu-lilotomab satetraxetan significantly increases the ratio of tumor to red marrow absorbed dose in non-Hodgkin lymphoma patients. Eur J Nucl Med Mol Imaging 2018; 45:1233-1241. [PMID: 29470615 PMCID: PMC5953993 DOI: 10.1007/s00259-018-3964-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE 177Lu-lilotomab satetraxetan is a novel anti-CD37 antibody radionuclide conjugate for the treatment of non-Hodgkin lymphoma (NHL). Four arms with different combinations of pre-dosing and pre-treatment have been investigated in a first-in-human phase 1/2a study for relapsed CD37+ indolent NHL. The aim of this work was to determine the tumor and normal tissue absorbed doses for all four arms, and investigate possible variations in the ratios of tumor to organs-at-risk absorbed doses. METHODS Two of the phase 1 arms included cold lilotomab pre-dosing (arm 1 and 4; 40 mg fixed and 100 mg/m2 BSA dosage, respectively) and two did not (arms 2 and 3). All patients were pre-treated with different regimens of rituximab. The patients received either 10, 15, or 20 MBq 177Lu-lilotomab satetraxetan per kg body weight. Nineteen patients were included for dosimetry, and a total of 47 lesions were included. The absorbed doses were calculated from multiple SPECT/CT-images and normalized by administered activity for each patient. Two-sided Student's t tests were used for all statistical analyses. RESULTS Organs with distinct uptake of 177Lu-lilotomab satetraxetan, in addition to tumors, were red marrow (RM), liver, spleen, and kidneys. The mean RM absorbed doses were 0.94, 1.55, 1.44, and 0.89 mGy/MBq for arms 1-4, respectively. For the patients not pre-dosed with lilotomab (arms 2 and 3 combined) the mean RM absorbed dose was 1.48 mGy/MBq, which was significantly higher than for both arm 1 (p = 0.04) and arm 4 (p = 0.02). Of the other organs, the highest uptake was found in the spleen, and there was a significantly lower spleen absorbed dose for arm-4 patients than for the patient group without lilotomab pre-dosing (1.13 vs. 3.20 mGy/MBq; p < 0.01). Mean tumor absorbed doses were 2.15, 2.31, 1.33, and 2.67 mGy/MBq for arms 1-4, respectively. After averaging the tumor absorbed dose for each patient, the patient mean tumor absorbed dose to RM absorbed dose ratios were obtained, given mean values of 1.07 for the patient group not pre-dosed with lilotomab, of 2.16 for arm 1, and of 4.62 for arm 4. The ratios were significantly higher in both arms 1 and 4 compared to the group without pre-dosing (p = 0.05 and p = 0.02). No statistically significant difference between arms 1 and 4 was found. CONCLUSIONS RM is the primary dose-limiting organ for 177Lu-lilotomab satetraxetan treatment, and pre-dosing with lilotomab has a mitigating effect on RM absorbed dose. Increasing the amount of lilotomab from 40 mg to 100 mg/m2 was found to slightly decrease the RM absorbed dose and increase the ratio of tumor to RM absorbed dose. Still, both pre-dosing amounts resulted in significantly higher tumor to RM absorbed dose ratios. The findings encourage continued use of pre-dosing with lilotomab.
Collapse
Affiliation(s)
- Caroline Stokke
- Department of Diagnostic Physics, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.
- Department of Life Sciences and Health, Oslo Metropolitan University, Oslo, Norway.
| | - Johan Blakkisrud
- Department of Diagnostic Physics, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Ayca Løndalen
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Anne C T Martinsen
- Department of Diagnostic Physics, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- The Department of Physics, University of Oslo, Oslo, Norway
| | - Harald Holte
- Department of Oncology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Arne Kolstad
- Department of Oncology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
25
|
Cahan B, Leong L, Wagman L, Yamauchi D, Shibata S, Wilzcynski S, Williams LE, Yazaki P, Colcher D, Frankel P, Wu A, Raubitschek A, Shively J, Wong JYC. Phase I/II Trial of Anticarcinoembryonic Antigen Radioimmunotherapy, Gemcitabine, and Hepatic Arterial Infusion of Fluorodeoxyuridine Postresection of Liver Metastasis for Colorectal Carcinoma. Cancer Biother Radiopharm 2018; 32:258-265. [PMID: 28910150 DOI: 10.1089/cbr.2017.2223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVES Report the feasibility, toxicities, and long-term results of a Phase I/II trial of 90Y-labeled anticarcinoembryonic antigen (anti-CEA) (cT84.66) radioimmunotherapy (RIT), gemcitabine, and hepatic arterial infusion (HAI) of fluorodeoxyuridine (FUdR) after maximal hepatic resection of metastatic colorectal cancer to the liver. METHODS Patients with metastatic colorectal cancer to the liver postresection or ablation to minimum disease were eligible. Each cohort received HAI of FUdR for 14 days on a dose escalation schedule. The maximum HAI FUdR dose level planned was 0.2 mg/kg/day, which is the standard dose for HAI FUdR alone. On day 9, 90Y-cT84.66 anti-CEA at 16.6 mCi/m2 as an i.v. bolus infusion and on days 9-11 i.v. gemcitabine at 105 mg/m2 were given. Patients could receive up to three cycles every 6 weeks of protocol therapy. Four additional cycles of HAI FUdR were allowed after RIT. RESULTS Sixteen patients were treated on this study. A maximum tolerated dose of 0.20 mg/kg/day of HAI FUdR combined with RIT at 16.6 mCi/m2 and gemcitabine at 105 mg/m2 was achieved with only 1 patient experiencing grade 3 reversible toxicity (mucositis). After surgery, 10 patients had no evidence of visible disease and remained without evidence of disease after completion of protocol therapy. The remaining 6 patients demonstrated radiological visible disease after surgery and after protocol therapy 2 patients had a CR, 1 patient had PR, 2 had stable disease, and 1 had progression. With a median follow-up of 41.8 months (18.7-114.6), median progression free survival was 9.6 months. Two patients demonstrated long-term disease control out to 45+ and 113+ months. CONCLUSION This study demonstrates the safety, feasibility, and potential utility of HAI FUdR, RIT, and systemic gemcitabine. The trimodality approach does not have higher hematologic toxicities than seen in prior RIT-alone studies. Future efforts evaluating RIT in colorectal cancer should integrate RIT with systemic and regional therapies in the minimal tumor burden setting.
Collapse
Affiliation(s)
- Benjamin Cahan
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Lucille Leong
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Lawrence Wagman
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - David Yamauchi
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Stephen Shibata
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Sharon Wilzcynski
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Lawrence E Williams
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Paul Yazaki
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - David Colcher
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Paul Frankel
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Anna Wu
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Andrew Raubitschek
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - John Shively
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Jeffrey Y C Wong
- Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| |
Collapse
|
26
|
Green DJ, O'Steen S, Lin Y, Comstock ML, Kenoyer AL, Hamlin DK, Wilbur DS, Fisher DR, Nartea M, Hylarides MD, Gopal AK, Gooley TA, Orozco JJ, Till BG, Orcutt KD, Wittrup KD, Press OW. CD38-bispecific antibody pretargeted radioimmunotherapy for multiple myeloma and other B-cell malignancies. Blood 2018; 131:611-620. [PMID: 29158362 PMCID: PMC5805491 DOI: 10.1182/blood-2017-09-807610] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
Pretargeted radioimmunotherapy (PRIT) has demonstrated remarkable efficacy targeting tumor antigens, but immunogenicity and endogenous biotin blocking may limit clinical translation. We describe a new PRIT approach for the treatment of multiple myeloma (MM) and other B-cell malignancies, for which we developed an anti-CD38-bispecific fusion protein that eliminates endogenous biotin interference and immunogenic elements. In murine xenograft models of MM and non-Hodgkin lymphoma (NHL), the CD38-bispecific construct demonstrated excellent blood clearance and tumor targeting. Dosimetry calculations showed a tumor-absorbed dose of 43.8 Gy per millicurie injected dose of 90Y, with tumor-to-normal organ dose ratios of 7:1 for liver and 15:1 for lung and kidney. In therapy studies, CD38-bispecific PRIT resulted in 100% complete remissions by day 12 in MM and NHL xenograft models, ultimately curing 80% of mice at optimal doses. In direct comparisons, efficacy of the CD38 bispecific proved equal or superior to streptavidin (SA)-biotin-based CD38-SA PRIT. Each approach cured at least 75% of mice at the highest radiation dose tested (1200 µCi), whereas at 600- and 1000-µCi doses, the bispecific outperformed the SA approach, curing 35% more mice overall (P < .004). The high efficacy of bispecific PRIT, combined with its reduced risk of immunogenicity and endogenous biotin interference, make the CD38 bispecific an attractive candidate for clinical translation. Critically, CD38 PRIT may benefit patients with unresponsive, high-risk disease because refractory disease typically retains radiation sensitivity. We posit that PRIT might not only prolong survival, but possibly cure MM and treatment-refractory NHL patients.
Collapse
Affiliation(s)
- Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Shyril O'Steen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Yukang Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Melissa L Comstock
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Aimee L Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Donald K Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | | | - Margaret Nartea
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Mark D Hylarides
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Theodore A Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Kelly D Orcutt
- Department of Chemical Engineering and
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - K Dane Wittrup
- Department of Chemical Engineering and
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| |
Collapse
|
27
|
Blakkisrud J, Holtedahl JE, Løndalen A, Dahle J, Bach-Gansmo T, Holte H, Nygaard S, Kolstad A, Stokke C. Biodistribution and Dosimetry Results from a Phase 1 Trial of Therapy with the Antibody–Radionuclide Conjugate 177Lu-Lilotomab Satetraxetan. J Nucl Med 2017; 59:704-710. [DOI: 10.2967/jnumed.117.195347] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/01/2017] [Indexed: 12/15/2022] Open
|
28
|
Drug Discovery by Molecular Imaging and Monitoring Therapy Response in Lymphoma. Int J Mol Sci 2017; 18:ijms18081639. [PMID: 28749424 PMCID: PMC5578029 DOI: 10.3390/ijms18081639] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/23/2017] [Accepted: 07/23/2017] [Indexed: 12/12/2022] Open
Abstract
Molecular imaging allows a noninvasive assessment of biochemical and biological processes in living subjects. Treatment strategies for malignant lymphoma depend on histology and tumor stage. For the last two decades, molecular imaging has been the mainstay diagnostic test for the staging of malignant lymphoma and the assessment of response to treatment. This technology enhances our understanding of disease and drug activity during preclinical and clinical drug development. Here, we review molecular imaging applications in drug development, with an emphasis on oncology. Monitoring and assessing the efficacy of anti-cancer therapies in preclinical or clinical models are essential and the multimodal molecular imaging approach may represent a new stage for pharmacologic development in cancer. Monitoring the progress of lymphoma therapy with imaging modalities will help patients. Identifying and addressing key challenges is essential for successful integration of molecular imaging into the drug development process. In this review, we highlight the general usefulness of molecular imaging in drug development and radionuclide-based reporter genes. Further, we discuss the different molecular imaging modalities for lymphoma therapy and their preclinical and clinical applications.
Collapse
|
29
|
Seidi K, Jahanban-Esfahlan R, Zarghami N. Tumor rim cells: From resistance to vascular targeting agents to complete tumor ablation. Tumour Biol 2017; 39:1010428317691001. [DOI: 10.1177/1010428317691001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current vascular targeting strategies pursue two main goals: anti-angiogenesis agents aim to halt sprouting and the formation of new blood vessels, while vascular disrupting agents along with coaguligands seek to compromise blood circulation in the vessels. The ultimate goal of such therapies is to deprive tumor cells out of oxygen and nutrients long enough to succumb cancer cells to death. Most of vascular targeting agents presented promising therapeutic potential, but the final goal which is cure is rarely achieved. Nevertheless, in both preclinical and clinical settings, tumors tend to grow back, featuring a highly invasive, metastatic, and extremely resistant form. This review highlights the critical significance of tumor rim cells as the main factor, determining therapy success with vascular targeting agents. We present an overview of different single and combination treatments with vascular targeting agents that enable efficient targeting of tumor rim cells and long-lasting tumor cure. Understanding the nature of tumor rim cells, how they establish, how they manage to survive of vascular targeting agents, and how they contribute in tumor refractoriness, may open new avenues to the development of beneficial strategies, capable to eliminate residual rim cells, and enable tumor ablation once and forever.
Collapse
Affiliation(s)
- Khaled Seidi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Green DJ, Frayo SL, Lin Y, Hamlin DK, Fisher DR, Frost SHL, Kenoyer AL, Hylarides MD, Gopal AK, Gooley TA, Orozco JJ, Till BG, O'Steen S, Orcutt KD, Wilbur DS, Wittrup KD, Press OW. Comparative Analysis of Bispecific Antibody and Streptavidin-Targeted Radioimmunotherapy for B-cell Cancers. Cancer Res 2016; 76:6669-6679. [PMID: 27590740 DOI: 10.1158/0008-5472.can-16-0571] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 08/10/2016] [Accepted: 08/29/2016] [Indexed: 11/16/2022]
Abstract
Streptavidin (SA)-biotin pretargeted radioimmunotherapy (PRIT) that targets CD20 in non-Hodgkin lymphoma (NHL) exhibits remarkable efficacy in model systems, but SA immunogenicity and interference by endogenous biotin may complicate clinical translation of this approach. In this study, we engineered a bispecific fusion protein (FP) that evades the limitations imposed by this system. Briefly, one arm of the FP was an anti-human CD20 antibody (2H7), with the other arm of the FP an anti-chelated radiometal trap for a radiolabeled ligand (yttrium[Y]-DOTA) captured by a very high-affinity anti-Y-DOTA scFv antibody (C825). Head-to-head biodistribution experiments comparing SA-biotin and bispecific FP (2H7-Fc-C825) PRIT in murine subjects bearing human lymphoma xenografts demonstrated nearly identical tumor targeting by each modality at 24 hours. However, residual radioactivity in the blood and normal organs was consistently higher following administration of 1F5-SA compared with 2H7-Fc-C825. Consequently, tumor-to-normal tissue ratios of distribution were superior for 2H7-Fc-C825 (P < 0.0001). Therapy studies in subjects bearing either Ramos or Granta subcutaneous lymphomas demonstrated that 2H7-Fc-C825 PRIT is highly effective and significantly less myelosuppressive than 1F5-SA (P < 0.0001). All animals receiving optimal doses of 2H7-Fc-C825 followed by 90Y-DOTA were cured by 150 days, whereas the growth of tumors in control animals progressed rapidly with complete morbidity by 25 days. In addition to demonstrating reduced risk of immunogenicity and an absence of endogenous biotin interference, our findings offer a preclinical proof of concept for the preferred use of bispecific PRIT in future clinical trials, due to a slightly superior biodistribution profile, less myelosuppression, and superior efficacy. Cancer Res; 76(22); 6669-79. ©2016 AACR.
Collapse
Affiliation(s)
- Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Department of Medicine, University of Washington, Seattle, Washington
| | - Shani L Frayo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Yukang Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Donald K Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | | | - Sofia H L Frost
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Aimee L Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark D Hylarides
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Theodore A Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Shyril O'Steen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kelly D Orcutt
- Department of Chemical Engineering, Massachusetts Institute of Technology, Boston, Massachusetts
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Boston, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Boston, Massachusetts
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
31
|
|
32
|
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
|
33
|
Ménager J, Gorin JB, Fichou N, Gouard S, Morgenstern A, Bruchertseifer F, Davodeau F, Kraeber-Bodéré F, Chérel M, Gaschet J, Guilloux Y. [Alpha-Radioimmunotherapy: principle and relevance in anti-tumor immunity]. Med Sci (Paris) 2016; 32:362-9. [PMID: 27137693 DOI: 10.1051/medsci/20163204014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alpha-radioimmunotherapy (α-RIT) is a targeted anti-tumor therapy using usually a monoclonal antibody specific for a tumor antigen that is coupled to an α-particle emitter. α-emitters represent an ideal tool to eradicate disseminated tumors or metastases. Recent data demonstrate that ionizing radiation in addition to its direct cytotoxic ability can also induce an efficient anti-tumor immunity. This suggests that biologic effects on irradiated tissues could be used to potentiate immunotherapy efficacy and opens the way for development of new therapies combining α-RIT and different types of immunotherapy.
Collapse
Affiliation(s)
- Jérémie Ménager
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France
| | - Jean-Baptiste Gorin
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France
| | - Nolwenn Fichou
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France
| | - Sébastien Gouard
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France
| | - Alfred Morgenstern
- European Commission, Joint research centre, Institute for transuranium elements, Karlsruhe, Allemagne
| | - Frank Bruchertseifer
- European Commission, Joint research centre, Institute for transuranium elements, Karlsruhe, Allemagne
| | - François Davodeau
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France - Institut de Cancérologie de l'Ouest, Saint-Herblain, France - CHU Nantes, département de médecine nucléaire, Nantes, France
| | - Michel Chérel
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France - Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Joëlle Gaschet
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France
| | - Yannick Guilloux
- Centre de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 Inserm, 8, quai Moncousu, BP 70721, 44007 Nantes Cedex 1, France - 6299 CNRS, Nantes, France - Université de Nantes, Nantes, France
| |
Collapse
|
34
|
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
|
35
|
Burtner CR, Chandrasekaran D, Santos EB, Beard BC, Adair JE, Hamlin DK, Wilbur DS, Sandmaier BM, Kiem HP. (211)Astatine-Conjugated Monoclonal CD45 Antibody-Based Nonmyeloablative Conditioning for Stem Cell Gene Therapy. Hum Gene Ther 2016; 26:399-406. [PMID: 25919226 DOI: 10.1089/hum.2015.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Most hematopoietic stem cell gene therapy studies require host conditioning to allow for efficient engraftment of gene-modified cells. Conditioning regimens with lower treatment-related toxicities are especially relevant for the treatment of nonmalignant blood disorders, such as hemoglobinopathies and immunodeficiencies, and for patients who are otherwise ineligible for conventional high-dose conditioning. Radioimmunotherapy, which employs an α- or a β-emitting radionuclide conjugated to a targeting antibody, is effective for delivering cytotoxic doses of radiation to a cell type of interest while minimizing off-target toxicity. Here, we demonstrate the feasibility of using a nonmyeloablative dose of a monoclonal anti-CD45 antibody conjugated to the α-emitter Astatine-211 ((211)At) to promote engraftment of an autologous gene-modified stem cell graft in the canine model. The doses used provided myelosuppression with rapid autologous recovery and minimal off-target toxicity. Engraftment levels were low in all dogs and reflected the low numbers of gene-modified cells infused. Our data suggest that a cell dose exceeding 1×10(6) cells/kg be used with nonmyeloablative doses of (211)At-anti-CD45 monoclonal antibodies for sustained engraftment in the dog model.
Collapse
Affiliation(s)
- Christopher R Burtner
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109
| | - Devikha Chandrasekaran
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109
| | - Erlinda B Santos
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109
| | - Brian C Beard
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109.,2 Division of Oncology, Department of Medicine, University of Washington , Seattle, WA 98195
| | - Jennifer E Adair
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109.,2 Division of Oncology, Department of Medicine, University of Washington , Seattle, WA 98195
| | - Donald K Hamlin
- 3 Radiation Oncology Department, University of Washington , Seattle, WA 98195
| | - D Scott Wilbur
- 3 Radiation Oncology Department, University of Washington , Seattle, WA 98195
| | - Brenda M Sandmaier
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109.,2 Division of Oncology, Department of Medicine, University of Washington , Seattle, WA 98195
| | - Hans-Peter Kiem
- 1 Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA 98109.,2 Division of Oncology, Department of Medicine, University of Washington , Seattle, WA 98195
| |
Collapse
|
36
|
Reagan PM, Friedberg JW. Advancing radioimmunotherapy and its future role in non-Hodgkin lymphoma. Future Oncol 2016; 11:1543-53. [PMID: 25963431 DOI: 10.2217/fon.15.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Radioimmunotherapy is an effective treatment modality with an acceptable toxicity profile in both indolent B-cell non-Hodgkin lymphoma and histologic transformation. Its ease of administration from a patient's perspective sets it apart from chemoimmunotherapy regimens. It has demonstrated efficacy in a range of different treatment scenarios. Despite its promise as a treatment modality, radioimmunotherapy has been seldom used, and one of the previously available agents is now off the market. Radioimmunotherapy has shown impressive activity in both the relapsed and upfront settings in follicular lymphoma, histologic transformation, as consolidation after chemotherapy, and in conjunction with high-dose chemotherapy and autologous stem cell support. Future efforts should focus on its optimal employment in the upfront setting for follicular lymphoma as well as further investigation of the promising activity in histologic transformation.
Collapse
Affiliation(s)
- Patrick M Reagan
- Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Avenue, Box 704, Rochester, NY 14642, USA
| | | |
Collapse
|
37
|
Cassaday RD, Stevenson PA, Gooley TA, Chauncey TR, Pagel JM, Rajendran J, Till BG, Philip M, Orozco JJ, Bensinger WI, Holmberg LA, Shustov AR, Green DJ, Smith SD, Libby EN, Maloney DG, Press OW, Gopal AK. High-dose CD20-targeted radioimmunotherapy-based autologous transplantation improves outcomes for persistent mantle cell lymphoma. Br J Haematol 2015; 171:788-97. [PMID: 26455717 DOI: 10.1111/bjh.13773] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/23/2015] [Indexed: 11/28/2022]
Abstract
Autologous stem cell transplant (ASCT) can improve outcomes for mantle cell lymphoma (MCL) patients, yet relapses are frequent. We hypothesized that high-dose anti-CD20 radioimmunotherapy (RIT)-based conditioning could improve results in this setting. We thus assessed 162 consecutive patients with MCL at our centre undergoing ASCT following high-dose RIT-based (n = 61) or standard (n = 101) conditioning. RIT patients were less likely to be in first remission (48% vs. 72%; P = 0·002), be in complete remission (CR) (26% vs. 61%; P < 0·001) and have chemosensitive disease (84% vs. 96%; P = 0·006). RIT-based conditioning was associated with a reduced risk of treatment failure [hazard ratio (HR) 0·40; P = 0·001] and mortality (HR 0·49; P = 0·01) after adjusting for these imbalances. This difference increased as disease status worsened (from CR to partial remission to stable/progressive disease), with respective HRs of 1·14, 0·53 and 0·04 for mortality, and 0·66, 0·36 and 0·14 for treatment failure. RIT-based conditioning appears to improve outcome following ASCT for MCL patients unable to achieve CR after controlling for imbalances in important risk factors. These data support the further study of RIT and radiation-based strategies in a risk-adapted approach to ASCT for persistent MCL.
Collapse
Affiliation(s)
- Ryan D Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Hematology, Department of Medicine, University of Washington, Seattle, USA
| | - Philip A Stevenson
- Clinical Statistics Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Theodore A Gooley
- Clinical Statistics Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Thomas R Chauncey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Veterans Affairs Puget Sound Health Care System, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - John M Pagel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Joseph Rajendran
- Division of Nuclear Medicine, Department of Radiology, University of Washington, Seattle, USA
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Mary Philip
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Hematology, Department of Medicine, University of Washington, Seattle, USA
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Hematology, Department of Medicine, University of Washington, Seattle, USA
| | - William I Bensinger
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Leona A Holmberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Andrei R Shustov
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Hematology, Department of Medicine, University of Washington, Seattle, USA
| | - Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Stephen D Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Edward N Libby
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - David G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| |
Collapse
|
38
|
90Y-daclizumab, an anti-CD25 monoclonal antibody, provided responses in 50% of patients with relapsed Hodgkin's lymphoma. Proc Natl Acad Sci U S A 2015; 112:13045-50. [PMID: 26438866 DOI: 10.1073/pnas.1516107112] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Despite significant advances in the treatment of Hodgkin's lymphoma (HL), a significant proportion of patients will not respond or will subsequently relapse. We identified CD25, the IL-2 receptor alpha subunit, as a favorable target for systemic radioimmunotherapy of HL. The scientific basis for the clinical trial was that, although most normal cells with exception of Treg cells do not express CD25, it is expressed by a minority of Reed-Sternberg cells and by most polyclonal T cells rosetting around Reed-Sternberg cells. Forty-six patients with refractory and relapsed HL were evaluated with up to seven i.v. infusions of the radiolabeled anti-CD25 antibody (90)Y-daclizumab. (90)Y provides strong β emissions that kill tumor cells at a distance by a crossfire effect. In 46 evaluable HL patients treated with (90)Y-daclizumab there were 14 complete responses and nine partial responses; 14 patients had stable disease, and nine progressed. Responses were observed both in patients whose Reed-Sternberg cells expressed CD25 and in those whose neoplastic cells were CD25(-) provided that associated rosetting T cells expressed CD25. As assessed using phosphorylated H2AX (γ-H2AX) as a bioindicator of the effects of radiation exposure, predominantly nonmalignant cells in the tumor microenvironment manifested DNA damage, as reflected by increased expression of γ-H2AX. Toxicities were transient bone-marrow suppression and myelodysplastic syndrome in six patients who had not been evaluated with bone-marrow karyotype analyses before therapy. In conclusion, repeated (90)Y-daclizumab infusions directed predominantly toward nonmalignant T cells rosetting around Reed-Sternberg cells provided meaningful therapy for select HL patients.
Collapse
|
39
|
Wahl RL, Horner TJ, Lin TS, Kaminski MS. Observational Retrospective Study of Altered Biodistribution of Tositumomab and 131I-Tositumomab. J Nucl Med 2015; 56:1800-3. [PMID: 26338897 DOI: 10.2967/jnumed.115.156190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 08/17/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The tositumomab/(131)I-tositumomab radioimmunotherapy regimen is administered as a dosimetric dose followed by a therapeutic dose. The biodistribution of the dosimetric dose is assessed by quantitative calculations of whole-body residence time (TBRT) and visual examination of whole-body γ-camera images, to determine the administered radioactivity dose and whether a therapeutic dose can be administered. We investigated whether altered biodistribution of (131)I-tositumomab could be identified using quantitative TBRT. METHODS BioClinica, Inc., provided γ-camera images to an independent reviewer to assess altered (131)I-tositumomab biodistribution in patients reported to a registry. RESULTS Of 2,649 therapeutic doses, 5 (0.2%) were cancelled because of altered biodistribution as determined by γ-camera images and TBRT. Of these, 3 γ-camera images were assessed by the independent reviewer; one showed altered biodistribution (0.04%) and was in agreement with the TBRT on-site calculation. CONCLUSION TBRT alone should be used to determine altered biodistribution and hence whether to administer the therapeutic dose.
Collapse
Affiliation(s)
- Richard L Wahl
- Department of Medical Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Thierry J Horner
- Oncology Research and Development, GlaxoSmithKline, Collegeville, Pennsylvania; and
| | - Thomas S Lin
- Oncology Research and Development, GlaxoSmithKline, Collegeville, Pennsylvania; and
| | - Mark S Kaminski
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
40
|
Repetto-Llamazares AHV, Larsen RH, Patzke S, Fleten KG, Didierlaurent D, Pichard A, Pouget JP, Dahle J. Targeted Cancer Therapy with a Novel Anti-CD37 Beta-Particle Emitting Radioimmunoconjugate for Treatment of Non-Hodgkin Lymphoma. PLoS One 2015; 10:e0128816. [PMID: 26066655 PMCID: PMC4466226 DOI: 10.1371/journal.pone.0128816] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/30/2015] [Indexed: 12/31/2022] Open
Abstract
177Lu-DOTA-HH1 (177Lu-HH1) is a novel anti-CD37 radioimmunoconjugate developed to treat non-Hodgkin lymphoma. Mice with subcutaneous Ramos xenografts were treated with different activities of 177Lu-HH1, 177Lu-DOTA-rituximab (177Lu-rituximab) and non-specific 177Lu-DOTA-IgG1 (177Lu-IgG1) and therapeutic effect and toxicity of the treatment were monitored. Significant tumor growth delay and increased survival of mice were observed in mice treated with 530 MBq/kg 177Lu-HH1 as compared with mice treated with similar activities of 177Lu-rituximab or non-specific 177Lu-IgG1, 0.9% NaCl or unlabeled HH1. All mice injected with 530 MBq/kg of 177Lu-HH1 tolerated the treatment well. In contrast, 6 out of 10 mice treated with 530 MBq/kg 177Lu-rituximab experienced severe radiation toxicity. The retention of 177Lu-rituximab in organs of the mononuclear phagocyte system was longer than for 177Lu-HH1, which explains the higher toxicity observed in mice treated with 177Lu-rituximab. In vitro internalization studies showed that 177Lu-HH1 internalizes faster and to a higher extent than 177Lu-rituximab which might be the reason for the better therapeutic effect of 177Lu-HH1.
Collapse
MESH Headings
- Animals
- Antibodies/chemistry
- Antibodies/immunology
- Antigen-Antibody Reactions
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Beta Particles
- Cell Line, Tumor
- Disease Models, Animal
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/therapeutic use
- Iodine Radioisotopes/chemistry
- Lutetium/chemistry
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/mortality
- Lymphoma, Non-Hodgkin/pathology
- Mice
- Mice, Nude
- Radioisotopes
- Radiopharmaceuticals/chemistry
- Radiopharmaceuticals/pharmacokinetics
- Radiopharmaceuticals/therapeutic use
- Rituximab/chemistry
- Rituximab/immunology
- Tetraspanins/chemistry
- Tetraspanins/immunology
- Tetraspanins/metabolism
- Tissue Distribution
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Ada H. V. Repetto-Llamazares
- Nordic Nanovector ASA, Kjelsåsveien 168, 0884, Oslo, Norway
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway
- * E-mail:
| | | | - Sebastian Patzke
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Karianne G. Fleten
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - David Didierlaurent
- UMR 1037 INSERM/UPS, Centre de Recherche en Cancérologie de Toulouse, Toulouse, F-31062, France
| | - Alexandre Pichard
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherche Médicale, U896, Université Montpellier, Montpellier, France
| | - Jean Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherche Médicale, U896, Université Montpellier, Montpellier, France
| | - Jostein Dahle
- Nordic Nanovector ASA, Kjelsåsveien 168, 0884, Oslo, Norway
| |
Collapse
|
41
|
Abstract
The eradication of cancer remains a vexing problem despite recent advances in our understanding of the molecular basis of neoplasia. One therapeutic approach that has demonstrated potential involves the selective targeting of radionuclides to cancer-associated cell surface antigens using monoclonal antibodies. Such radioimmunotherapy (RIT) permits the delivery of a high dose of therapeutic radiation to cancer cells, while minimizing the exposure of normal cells. Although this approach has been investigated for several decades, the cumulative advances in cancer biology, antibody engineering and radiochemistry in the past decade have markedly enhanced the ability of RIT to produce durable remissions of multiple cancer types.
Collapse
Affiliation(s)
- Steven M Larson
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Jorge A Carrasquillo
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Nai-Kong V Cheung
- 1] Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA. [2]
| | - Oliver W Press
- 1] Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, P.O. BOX 19024, Seattle, Washington 98109, USA. [2]
| |
Collapse
|
42
|
Frost SHL, Frayo SL, Miller BW, Orozco JJ, Booth GC, Hylarides MD, Lin Y, Green DJ, Gopal AK, Pagel JM, Bäck TA, Fisher DR, Press OW. Comparative efficacy of 177Lu and 90Y for anti-CD20 pretargeted radioimmunotherapy in murine lymphoma xenograft models. PLoS One 2015; 10:e0120561. [PMID: 25785845 PMCID: PMC4364776 DOI: 10.1371/journal.pone.0120561] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/05/2015] [Indexed: 11/19/2022] Open
Abstract
PURPOSE Pretargeted radioimmunotherapy (PRIT) is a multi-step method of selectively delivering high doses of radiotherapy to tumor cells while minimizing exposure to surrounding tissues. Yttrium-90 (90Y) and lutetium-177 (177Lu) are two of the most promising beta-particle emitting radionuclides used for radioimmunotherapy, which despite having similar chemistries differ distinctly in terms of radiophysical features. These differences may have important consequences for the absorbed dose to tumors and normal organs. Whereas 90Y has been successfully applied in a number of preclinical and clinical radioimmunotherapy settings, there have been few published pretargeting studies with 177Lu. We therefore compared the therapeutic potential of targeting either 90Y or 177Lu to human B-cell lymphoma xenografts in mice. METHODS Parallel experiments evaluating the biodistribution, imaging, dosimetry, therapeutic efficacy, and toxicity were performed in female athymic nude mice bearing either Ramos (Burkitt lymphoma) or Granta (mantle cell lymphoma) xenografts, utilizing an anti-CD20 antibody-streptavidin conjugate (1F5-SA) and an 90Y- or 177Lu-labeled 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-biotin second step reagent. RESULTS The two radionuclides displayed comparable biodistributions in tumors and normal organs; however, the absorbed radiation dose delivered to tumor was more than twice as high for 90Y (1.3 Gy/MBq) as for 177Lu (0.6 Gy/MBq). More importantly, therapy with 90Y-DOTA-biotin was dramatically more effective than with 177Lu-DOTA-biotin, with 100% of Ramos xenograft-bearing mice cured with 37 MBq 90Y, whereas 0% were cured using identical amounts of 177Lu-DOTA-biotin. Similar results were observed in mice bearing Granta xenografts, with 80% of the mice cured with 90Y-PRIT and 0% cured with 177Lu-PRIT. Toxicities were comparable with both isotopes. CONCLUSION 90Y was therapeutically superior to 177Lu for streptavidin-biotin PRIT approaches in these human lymphoma xenograft models.
Collapse
Affiliation(s)
- Sofia H. L. Frost
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- * E-mail:
| | - Shani L. Frayo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Brian W. Miller
- Pacific Northwest National Laboratory, Richland, WA, United States of America
- College of Optical Sciences, The University of Arizona, Tucson, AZ, United States of America
| | - Johnnie J. Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Garrett C. Booth
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Mark D. Hylarides
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Yukang Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Damian J. Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Departments of Medicine and Bioengineering, 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
- Departments of Medicine and Bioengineering, 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
- Departments of Medicine and Bioengineering, University of Washington, Seattle, WA, United States of America
| | - Tom A. Bäck
- Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Oliver W. Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Departments of Medicine and Bioengineering, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
43
|
Berger MD, Branger G, Klaeser B, Taleghani BM, Novak U, Banz Y, Mueller BU, Pabst T. Zevalin and BEAM (Z-BEAM) versus rituximab and BEAM (R-BEAM) conditioning chemotherapy prior to autologous stem cell transplantation in patients with mantle cell lymphoma. Hematol Oncol 2015; 34:133-9. [PMID: 25689832 DOI: 10.1002/hon.2197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/17/2015] [Accepted: 01/21/2015] [Indexed: 11/06/2022]
Abstract
Early relapse is common in patients with mantle cell lymphoma (MCL) highlighting the unmet need for further improvement of therapeutic options for these patients. CD20 inhibition combined with induction chemotherapy as well as consolidation with high-dose chemotherapy (HDCT) is increasingly considered cornerstones within current therapy algorithms of MCL whereas the role of radioimmunotherapy is unclear. This retrospective single center study compared 46 consecutive MCL patients receiving HDCT in first or second remission. Thirty-five patients had rituximab and BEAM (R-BEAM), and 11 patients received ibritumomab tiuxetan (Zevalin®), an Yttrium-90 labeled CD20 targeting antibody, prior to BEAM (Z-BEAM) followed by autologous stem cell transplantation (ASCT). We observed that the 5-year overall survival (OS) in the R-BEAM and Z-BEAM groups was 55% and 71% (p = 0.288), and the 4-year progression free survival (PFS) was 32% and 41%, respectively (p = 0.300). There were no treatment related deaths in both groups, and we observed no differences in toxicities, infection rates or engraftment. Our data suggest that the Z-BEAM conditioning regimen followed by ASCT is well tolerated, but was not associated with significantly improved survival compared to R-BEAM. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Martin D Berger
- Department of Medical Oncology, University Hospital, Berne, Switzerland
| | - Giacomo Branger
- Department of Medical Oncology, University Hospital, Berne, Switzerland
| | - Bernd Klaeser
- Department of Nuclear Medicine, University Hospital, Berne, Switzerland
| | | | - Urban Novak
- Department of Medical Oncology, University Hospital, Berne, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Berne, Berne, Switzerland
| | - Beatrice U Mueller
- Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, University Hospital, Berne, Switzerland
| |
Collapse
|
44
|
Hsieh YT, Chen KC, Cheng CM, Cheng TL, Tao MH, Roffler SR. Impediments to enhancement of CPT-11 anticancer activity by E. coli directed beta-glucuronidase therapy. PLoS One 2015; 10:e0118028. [PMID: 25688562 PMCID: PMC4331512 DOI: 10.1371/journal.pone.0118028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/05/2015] [Indexed: 12/17/2022] Open
Abstract
CPT-11 is a camptothecin analog used for the clinical treatment of colorectal adenocarcinoma. CPT-11 is converted into the therapeutic anti-cancer agent SN-38 by liver enzymes and can be further metabolized to a non-toxic glucuronide SN-38G, resulting in low SN-38 but high SN-38G concentrations in the circulation. We previously demonstrated that adenoviral expression of membrane-anchored beta-glucuronidase could promote conversion of SN-38G to SN-38 in tumors and increase the anticancer activity of CPT-11. Here, we identified impediments to effective tumor therapy with E. coli that were engineered to constitutively express highly active E. coli beta-glucuronidase intracellularly to enhance the anticancer activity of CPT-11. The engineered bacteria, E. coli (lux/βG), could hydrolyze SN-38G to SN-38, increased the sensitivity of cultured tumor cells to SN-38G by about 100 fold and selectively accumulated in tumors. However, E. coli (lux/βG) did not more effectively increase CPT-11 anticancer activity in human tumor xenografts as compared to non-engineered E. coli. SN-38G conversion to SN-38 by E. coli (lux/βG) appeared to be limited by slow uptake into bacteria as well as by segregation of E. coli in necrotic regions of tumors that may be relatively inaccessible to systemically-administered drug molecules. Studies using a fluorescent glucuronide probe showed that significantly greater glucuronide hydrolysis could be achieved in mice pretreated with E. coli (lux/βG) by direct intratumoral injection of the glucuronide probe or by intratumoral lysis of bacteria to release intracellular beta-glucuronidase. Our study suggests that the distribution of beta-glucuronidase, and possibly other therapeutic proteins, in the tumor microenvironment might be an important barrier for effective bacterial-based tumor therapy. Expression of secreted therapeutic proteins or induction of therapeutic protein release from bacteria might therefore be a promising strategy to enhance anti-tumor activity.
Collapse
Affiliation(s)
- Yuan-Ting Hsieh
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Kai-Chuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chiu-Min Cheng
- Department of Aquaculture, National Kaohsiung Marine University, Kaohsiung, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mi-Hua Tao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R. Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
45
|
Astatine-211 conjugated to an anti-CD20 monoclonal antibody eradicates disseminated B-cell lymphoma in a mouse model. Blood 2015; 125:2111-9. [PMID: 25628467 DOI: 10.1182/blood-2014-11-612770] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
α-Emitting radionuclides deposit a large amount of energy within a few cell diameters and may be particularly effective for radioimmunotherapy targeting minimal residual disease (MRD). To evaluate this hypothesis, (211)At-labeled 1F5 monoclonal antibody (mAb) (anti-CD20) was studied in both bulky lymphoma tumor xenograft and MRD animal models. Superior treatment responses to (211)At-labeled 1F5 mAb were evident in the MRD setting. Lymphoma xenograft tumor-bearing animals treated with doses of up to 48 µCi of (211)At-labeled anti-CD20 mAb ([(211)At]1F5-B10) experienced modest responses (0% cures but two- to threefold prolongation of survival compared with negative controls). In contrast, 70% of animals in the MRD lymphoma model demonstrated complete eradication of disease when treated with (211)At-B10-1F5 at a radiation dose that was less than one-third (15 µCi) of the highest dose given to xenograft animals. Tumor progression among untreated control animals in both models was uniformly lethal. After 130 days, no significant renal or hepatic toxicity was observed in the cured animals receiving 15 µCi of [(211)At]1F5-B10. These findings suggest that α-emitters are highly efficacious in MRD settings, where isolated cells and small tumor clusters prevail.
Collapse
|
46
|
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
|
47
|
Abstract
Radiation therapy and immunotherapy are both well-established treatments for malignant disease. Radiotherapy has long been utilized for purposes of providing local tumor control, and the recent success with novel immunomodulatory agents has brought immunotherapy into the forefront of clinical practice for the treatment of many tumor types. Although radiotherapy has traditionally been thought to mediate tumor regression through direct cytotoxic effects, it is now known that radiation also alters the local tumor microenvironment with effects on both the local and systemic anti-tumor immune response. There is growing evidence that the rational integration of the immunomodulatory effects of radiotherapy with the expanding armamentarium of clinically approved immunotherapeutics can yield potent anti-tumor responses exceeding the benefit of either therapy alone. Here we summarize current approaches to the combination of immunotherapy with radiation therapy.
Collapse
Affiliation(s)
- Susan M Hiniker
- Department of Radiation Oncology, Stanford University, Stanford, CA.
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University, Stanford, CA
| |
Collapse
|
48
|
Witzig TE, Tomblyn MB, Misleh JG, Kio EA, Sharkey RM, Wegener WA, Goldenberg DM. Anti-CD22 90Y-epratuzumab tetraxetan combined with anti-CD20 veltuzumab: a phase I study in patients with relapsed/refractory, aggressive non-Hodgkin lymphoma. Haematologica 2014; 99:1738-45. [PMID: 25150258 DOI: 10.3324/haematol.2014.112110] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
A lingering criticism of radioimmunotherapy in non-Hodgkin lymphoma is the use of cold anti-CD20 antibody along with the radiolabeled anti-CD20 antibody. We instead combined radioimmunotherapy with immunotherapy targeting different B-cell antigens. We evaluated the anti-CD22 (90)Y-epratuzumab tetraxetan with the anti-CD20 veltuzumab in patients with aggressive lymphoma in whom at least one prior standard treatment had failed, but who had not undergone stem cell transplantation. Eighteen patients (median age 73 years, median of 3 prior treatments) received 200 mg/m(2) veltuzumab once-weekly for 4 weeks, with (90)Y-epratuzumab tetraxetan at planned doses in weeks 3 and 4, and (111)In-epratuzumab tetraxetan in week 2 for imaging and dosimetry. Veltuzumab effectively lowered levels of B cells in the blood prior to the radioimmunotherapy doses. No significant immunogenicity or change in pharmacokinetics of either agent occurred in combination. (111)In imaging showed tumor targeting with acceptable radiation dosimetry to normal organs. For (90)Y-epratuzumab tetraxetan, transient myelosuppression was dose-limiting with 6 mCi/m(2) (222 MBq/m(2)) × 2 being the maximal tolerated dose. Of 17 assessable patients, nine (53%) had objective responses according to the 2007 revised treatment response criteria, including three (18%) complete responses (2 relapsing after 11 and 13 months, 1 continuing to be clinically disease-free at 19 months), and six (35%) partial responses (1 relapsing after 14 months, 5 at 3 - 7 months). Responses occurred in patients with different lymphoma histologies, treated at different (90)Y dose levels, and with a predicted risk of poor outcome, most importantly including five of the six patients treated with the maximal tolerated dose (2 of whom achieved durable complete responses). In conclusion, the combination of (90)Y-epratuzumab tetraxetan and veltuzumab was well-tolerated with encouraging therapeutic activity in this difficult-to-treat population.
Collapse
Affiliation(s)
| | | | | | - Ebenezer A Kio
- Indiana University Health Center for Cancer Care, Goshen, IN
| | | | | | - David M Goldenberg
- Immunomedics, Morris Plains, NJ Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ, USA
| |
Collapse
|
49
|
Busse N, Erwin W, Pan T. Evaluation of a semiautomated lung mass calculation technique for internal dosimetry applications. Med Phys 2014; 40:122503. [PMID: 24320539 DOI: 10.1118/1.4830433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE The authors sought to evaluate a simple, semiautomated lung mass estimation method using computed tomography (CT) scans obtained using a variety of acquisition techniques and reconstruction parameters for mass correction of medical internal radiation dose-based internal radionuclide radiation absorbed dose estimates. METHODS CT scans of 27 patients with lung cancer undergoing stereotactic body radiation therapy treatment planning with PET∕CT were analyzed retrospectively. For each patient, free-breathing (FB) and respiratory-gated 4DCT scans were acquired. The 4DCT scans were sorted into ten respiratory phases, representing one complete respiratory cycle. An average CT reconstruction was derived from the ten-phase reconstructions. Mid expiration breath-hold CT scans were acquired in the same session for many patients. Deep inspiration breath-hold diagnostic CT scans of many of the patients were obtained from different scanning sessions at similar time points to evaluate the effect of contrast administration and maximum inspiration breath-hold. Lung mass estimates were obtained using all CT scan types, and intercomparisons made to assess lung mass variation according to scan type. Lung mass estimates using the FB CT scans from PET∕CT examinations of another group of ten male and ten female patients who were 21-30 years old and did not have lung disease were calculated and compared with reference lung mass values. To evaluate the effect of varying CT acquisition and reconstruction parameters on lung mass estimation, an anthropomorphic chest phantom was scanned and reconstructed with different CT parameters. CT images of the lungs were segmented using the OsiriX MD software program with a seed point of about -850 HU and an interval of 1000. Lung volume, and mean lung, tissue, and air HUs were recorded for each scan. Lung mass was calculated by assuming each voxel was a linear combination of only air and tissue. The specific gravity of lung volume was calculated using the formula (lung HU - air HU)∕(tissue HU - air HU), and mass = specific gravity × total volume × 1.04 g∕cm(3). RESULTS The range of calculated lung masses was 0.51-1.29 kg. The average male and female lung masses during FB CT were 0.80 and 0.71 kg, respectively. The calculated lung mass varied across the respiratory cycle but changed to a lesser degree than did lung volume measurements (7.3% versus 15.4%). Lung masses calculated using deep inspiration breath-hold and average CT were significantly larger (p < 0.05) than were some masses calculated using respiratory-phase and FB CT. Increased voxel size and smooth reconstruction kernels led to high lung mass estimates owing to partial volume effects. CONCLUSIONS Organ mass correction is an important component of patient-specific internal radionuclide dosimetry. Lung mass calculation necessitates scan-based density correction to account for volume changes owing to respiration. The range of lung masses in the authors' patient population represents lung doses for the same absorbed energy differing from 25% below to 64% above the dose found using reference phantom organ masses. With proper management of acquisition parameters and selection of FB or midexpiration breath hold scans, lung mass estimates with about 10% population precision may be achieved.
Collapse
Affiliation(s)
- Nathan Busse
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | | | | |
Collapse
|
50
|
Repetto-Llamazares AHV, Larsen RH, Giusti AM, Riccardi E, Bruland ØS, Selbo PK, Dahle J. 177Lu-DOTA-HH1, a novel anti-CD37 radio-immunoconjugate: a study of toxicity in nude mice. PLoS One 2014; 9:e103070. [PMID: 25068508 PMCID: PMC4113375 DOI: 10.1371/journal.pone.0103070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/27/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND CD37 is an internalizing B-cell antigen expressed on Non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia cells (CLL). The anti-CD37 monoclonal antibody HH1 was conjugated to the bifunctional chelator p-SCN-Bn-DOTA and labelled with the beta-particle emitting radionuclide 177Lu creating the radio-immunoconjugate (RIC) 177Lu-DOTA-HH1 (177Lu-HH1, trade name Betalutin). The present toxicity study was performed prior to initiation of clinical studies with 177Lu-HH1. METHODOLOGY/PRINCIPAL FINDINGS Nude mice with or without tumor xenografts were treated with 50 to 1000 MBq/kg 177Lu- HH1 and followed for clinical signs of toxicity up to ten months. Acute, life threatening bone marrow toxicity was observed in animals receiving 800 and 1000 MBq/kg 177Lu-HH1. Significant changes in serum concentrations of liver enzymes were evident for treatment with 1000 MBq/kg 177Lu-HH1. Lymphoid depletion, liver necrosis and atrophy, and interstitial cell hyperplasia of the ovaries were also observed for mice in this dose group. CONCLUSIONS/SIGNIFICANCE 177Lu-DOTA-HH1 was well tolerated at dosages about 10 times above those considered relevant for radioimmunotherapy in patients with B-cell derived malignancies.The toxicity profile was as expected for RICs. Our experimental results have paved the way for clinical evaluation of 177Lu-HH1 in NHL patients.
Collapse
Affiliation(s)
- Ada H. V. Repetto-Llamazares
- Nordic Nanovector AS, Oslo, Norway
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | | | | | | | - Øyvind S. Bruland
- Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pål Kristian Selbo
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | | |
Collapse
|