1
|
Malinzi J, Basita KB, Padidar S, Adeola HA. Prospect for application of mathematical models in combination cancer treatments. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
2
|
Tagawa ST, Vallabhajosula S, Christos PJ, Jhanwar YS, Batra JS, Lam L, Osborne J, Beltran H, Molina AM, Goldsmith SJ, Bander NH, Nanus DM. Phase 1/2 study of fractionated dose lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 ( 177 Lu-J591) for metastatic castration-resistant prostate cancer. Cancer 2019; 125:2561-2569. [PMID: 31012963 DOI: 10.1002/cncr.32072] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Prostate cancer is radiosensitive. Prostate-specific membrane antigen (PSMA) is selectively overexpressed on advanced, castration-resistant tumors. Lutetium-177-labeled anti-PSMA monoclonal antibody J591 (177 Lu-J591) targets prostate cancer with efficacy and dose-response/toxicity data when delivered as a single dose. Dose fractionation may allow higher doses to be administered safely. METHOD Men with metastatic castration-resistant prostate cancer refractory to or refusing standard treatment options with normal neutrophil and platelet counts were enrolled in initial phase 1b dose-escalation cohorts followed by phase 2a cohorts treated at recommended phase 2 doses (RP2Ds) comprising 2 fractionated doses of 177 Lu-J591 2 weeks apart. 177 Lu-J591 imaging was performed after treatment, but no selection for PSMA expression was performed before enrollment. Phase 2 patients had circulating tumor cell (CTC) counts assessed before and after treatment. RESULTS Forty-nine men received fractionated doses of 177 Lu-J591 ranging from 20 to 45 mCi/m2 ×2 two weeks apart. The dose-limiting toxicity in phase 1 was neutropenia. The RP2Ds were 40 mCi/m2 and 45 mCi/m2 ×2. At the highest RP2D (45 mCi/m2 ×2), 35.3% of patients had reversible grade 4 neutropenia, and 58.8% of patients had thrombocytopenia. This dose showed a greater decrease in prostate-specific antigen (PSA) levels and longer survival (87.5% with any PSA decrease, 58.8% with >30% decrease, 29.4% with >50% decrease; median survival, 42.3 months [95% confidence interval, 19.9-64.7]). Fourteen of 17 (82%) patients with detectable CTCs experienced a decrease in CTC count. Overall, 79.6% of patients had positive PSMA imaging; those with less intense PSMA imaging tended to have poorer responses. CONCLUSION Fractionated administration of 177 Lu-J591 allowed higher cumulative radiation dosing. The frequency and depth of PSA decrease, overall survival, and toxicity (dose-limiting myelosuppression) increased with higher doses.
Collapse
Affiliation(s)
- Scott T Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York.,Department of Urology, Weill Cornell Medicine, New York, New York.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Shankar Vallabhajosula
- Division of Nuclear Medicine, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Paul J Christos
- Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, New York.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Yuliya S Jhanwar
- Division of Nuclear Medicine, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Jaspreet S Batra
- Department of Urology, Weill Cornell Medicine, New York, New York
| | - Linda Lam
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Joseph Osborne
- Division of Nuclear Medicine, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Himisha Beltran
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York.,Dana Farber Cancer Institute, Boston, Massachusetts
| | - Ana M Molina
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Stanley J Goldsmith
- Division of Nuclear Medicine, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Neil H Bander
- Department of Urology, Weill Cornell Medicine, New York, New York.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - David M Nanus
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York.,Department of Urology, Weill Cornell Medicine, New York, New York.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| |
Collapse
|
3
|
Evans-Axelsson S, Timmermand OV, Bjartell A, Strand SE, Elgqvist J. Radioimmunotherapy for Prostate Cancer--Current Status and Future Possibilities. Semin Nucl Med 2016; 46:165-79. [PMID: 26897720 DOI: 10.1053/j.semnuclmed.2015.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) is one of the most common cancers in men and is the second leading cause of cancer-related deaths in the USA. In the United States, it is the second most frequently diagnosed cancer after skin cancer, and in Europe it is number one. According to the American Cancer Society, approximately 221,000 men in the United States would be diagnosed with PCa during 2015, and approximately 28,000 would die of the disease. According to the International Agency for Research on Cancer, approximately 345,000 men were diagnosed with PCa in Europe during 2012, and despite more emphasis placed on early detection through routine screening, 72,000 men died of the disease. Hence, the need for improved therapy modalities is of utmost importance. And targeted therapies based on radiolabeled specific antibodies or peptides are a very interesting and promising alternative to increase the therapeutic efficacy and overall chance of survival of these patients. There are currently several preclinical and some clinical studies that have been conducted, or are ongoing, to investigate the therapeutic efficacy and toxicity of radioimmunotherapy (RIT) against PCa. One thing that is lacking in a lot of these published studies is the dosimetry data, which are needed to compare results between the studies and the study locations. Given the complicated tumor microenvironment and overall complexity of RIT to PCa, old and new targets and targeting strategies like combination RIT and pretargeting RIT are being improved and assessed along with various therapeutic radionuclides candidates. Given alone or in combination with other therapies, these new and improved strategies and RIT tools further enhance the clinical response to RIT drugs in PCa, making RIT for PCa an increasingly practical clinical tool.
Collapse
Affiliation(s)
- Susan Evans-Axelsson
- Department of Translational Medicine, Division of Urological Cancers, Skåne University Hospital, Malmö, Lund University, Lund, Sweden
| | | | - Anders Bjartell
- Department of Translational Medicine, Division of Urological Cancers, Skåne University Hospital, Malmö, Lund University, Lund, Sweden; Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Sven-Erik Strand
- Department of Clinical Sciences, Lund, Division of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Jörgen Elgqvist
- Department of Clinical Sciences, Lund, Division of Medical Radiation Physics, Lund University, Lund, Sweden.
| |
Collapse
|
4
|
Sgouros G, Hobbs RF. Patient-Specific Dosimetry, Radiobiology, and the Previously-Treated Patient. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/174_2012_684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
5
|
Nosanchuk JD, Dadachova E. Radioimmunotherapy of fungal diseases: the therapeutic potential of cytocidal radiation delivered by antibody targeting fungal cell surface antigens. Front Microbiol 2012; 2:283. [PMID: 22275913 PMCID: PMC3257868 DOI: 10.3389/fmicb.2011.00283] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 12/28/2011] [Indexed: 01/26/2023] Open
Abstract
Radioimmunotherapy is the targeted delivery of cytocidal radiation to cells via specific antibody. Although mature for the treatment of cancer, RIT of infectious diseases is in pre-clinical development. However, as there is an obvious and urgent need for novel approaches to treat infectious diseases, RIT can provide us with a powerful approach to combat serious diseases, including invasive fungal infections. For example, RIT has proven more effective than standard amphotericin B for the treatment of experimental cryptococcosis. This review will discuss the concepts of RIT, its applications for infectious diseases, and the strides made to date to bring RIT of infectious diseases to fruition. Finally, we will discuss the potential of PAN-FUNGAL RIT, the targeting of conserved fungal cell surface antigens by RIT, as a treatment modality for fungi prior to the formal microbiological identification of the specific pathogen. In sum, RIT provides a mechanism for the targeted killing of drug susceptible or resistant fungi irrespective of the host immune status and may dramatically reduce the length of therapy currently required for many invasive fungal diseases.
Collapse
Affiliation(s)
- Joshua D Nosanchuk
- Department of Medicine, Albert Einstein College of Medicine Bronx, NY, USA
| | | |
Collapse
|
6
|
Prostate-specific membrane antigen-based therapeutics. Adv Urol 2011; 2012:973820. [PMID: 21811498 PMCID: PMC3145341 DOI: 10.1155/2012/973820] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 05/09/2011] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PC) is the most common noncutaneous malignancy affecting men in the US, leading to significant morbidity and mortality. While significant therapeutic advances have been made, available systemic therapeutic options are lacking. Prostate-specific membrane antigen (PSMA) is a highly-restricted prostate cell-surface antigen that may be targeted. While initial anti-PSMA monoclonal antibodies were suboptimal, the development of monoclonal antibodies such as J591 which are highly specific for the external domain of PSMA has allowed targeting of viable, intact prostate cancer cells. Radiolabeled J591 has demonstrated accurate and selective tumor targeting, safety, and efficacy. Ongoing studies using anti-PSMA radioimmunotherapy with 177Lu-J591 seek to improve the therapeutic profile, select optimal candidates with biomarkers, combine with chemotherapy, and prevent or delay the onset of metastatic disease for men with biochemical relapse. Anti-PSMA monoclonal antibody-drug conjugates have also been developed with completed and ongoing early-phase clinical trials. As PSMA is a selective antigen that is highly overexpressed in prostate cancer, anti-PSMA-based immunotherapy has also been studied and utilized in clinical trials.
Collapse
|
7
|
Stabin MG, Sharkey RM, Siegel JA. RADAR Commentary: Evolution and Current Status of Dosimetry in Nuclear Medicine. J Nucl Med 2011; 52:1156-61. [DOI: 10.2967/jnumed.111.088666] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
8
|
Bouchelouche K, Tagawa ST, Goldsmith SJ, Turkbey B, Capala J, Choyke P. PET/CT Imaging and Radioimmunotherapy of Prostate Cancer. Semin Nucl Med 2011; 41:29-44. [PMID: 21111858 PMCID: PMC3392994 DOI: 10.1053/j.semnuclmed.2010.08.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Prostate cancer is a common cancer in men and continues to be a major health problem. Imaging plays an important role in the clinical management of patients with prostate cancer. An important goal for prostate cancer imaging is more accurate disease characterization through the synthesis of anatomic, functional, and molecular imaging information. Positron emission tomography (PET)/computed tomography (CT) in oncology is emerging as an important imaging tool. The most common radiotracer for PET/CT in oncology, (18)F-fluorodeoxyglucose (FDG), is not very useful in the imaging of prostate cancer. However, in recent years other PET tracers have improved the accuracy of PET/CT imaging of prostate cancer. Among these, choline labeled with (18)F or (11)C, (11)C-acetate, and (18)F-fluoride has demonstrated promising results, and other new radiopharmaceuticals are under development and evaluation in preclinical and clinical studies. Large prospective clinical PET/CT trials are needed to establish the role of PET/CT in prostate cancer patients. Because there are only limited available therapeutic options for patients with advanced metastatic prostate cancer, there is an urgent need for the development of more effective treatment modalities that could improve outcome. Prostate cancer represents an attractive target for radioimmunotherapy (RIT) for several reasons, including pattern of metastatic spread (lymph nodes and bone marrow, sites with good access to circulating antibodies) and small volume disease (ideal for antigen access and antibody delivery). Furthermore, prostate cancer is also radiation sensitive. Prostate-specific membrane antigen is expressed by virtually all prostate cancers, and represents an attractive target for RIT. Antiprostate-specific membrane antigen RIT demonstrates antitumor activity and is well tolerated. Clinical trials are underway to further improve upon treatment efficacy and patient selection. This review focuses on the recent advances of clinical PET/CT imaging and RIT of prostate cancer.
Collapse
Affiliation(s)
- Kirsten Bouchelouche
- PET and Cyclotron Unit, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
9
|
Tagawa ST, Beltran H, Vallabhajosula S, Goldsmith SJ, Osborne J, Matulich D, Petrillo K, Parmar S, Nanus DM, Bander NH. Anti-prostate-specific membrane antigen-based radioimmunotherapy for prostate cancer. Cancer 2010; 116:1075-83. [PMID: 20127956 DOI: 10.1002/cncr.24795] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite recent advances, advanced prostate cancer is suboptimally responsive to current chemotherapeutic agents. Radiolabeled monoclonal antibody therapy that targets prostate-specific membrane antigen (PSMA) shows promise and is an area of active investigation. J591 is a deimmunized IgG monoclonal antibody developed to target the extracellular domain of PSMA. Preclinical and early phase clinical studies using radiolabeled J591 have demonstrated efficacy in targeting tumor cells and decreasing levels of prostate-specific antigen. Radiolabeled J591 is well-tolerated, nonimmunogenic, and can be administered in multiple doses. The dose-limiting toxicity is reversible myelosuppression with little nonhematologic toxicity. Future studies will include approaches to optimize patient selection and incorporate novel strategies to improve the success of anti-PSMA radioimmunotherapy.
Collapse
Affiliation(s)
- Scott T Tagawa
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Sgouros G, Frey E, Wahl R, He B, Prideaux A, Hobbs R. Three-dimensional imaging-based radiobiological dosimetry. Semin Nucl Med 2008; 38:321-34. [PMID: 18662554 PMCID: PMC2597292 DOI: 10.1053/j.semnuclmed.2008.05.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Targeted radionuclide therapy holds promise as a new treatment for cancer. Advances in imaging are making it possible for researchers to evaluate the spatial distribution of radioactivity in tumors and normal organs over time. Matched anatomical imaging, such as combined single-photon emission computed tomography/computed tomography and positron emission tomography/computed tomography, has also made it possible to obtain tissue density information in conjunction with the radioactivity distribution. Coupled with sophisticated iterative reconstruction algorithms, these advances have made it possible to perform highly patient-specific dosimetry that also incorporates radiobiological modeling. Such sophisticated dosimetry techniques are still in the research investigation phase. Given the attendant logistical and financial costs, a demonstrated improvement in patient care will be a prerequisite for the adoption of such highly-patient specific internal dosimetry methods.
Collapse
Affiliation(s)
- George Sgouros
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD 21231, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Palm S, Bäck T, Claesson I, Danielsson A, Elgqvist J, Frost S, Hultborn R, Jensen H, Lindegren S, Jacobsson L. Therapeutic efficacy of astatine-211-labeled trastuzumab on radioresistant SKOV-3 tumors in nude mice. Int J Radiat Oncol Biol Phys 2007; 69:572-9. [PMID: 17869670 DOI: 10.1016/j.ijrobp.2007.06.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 06/09/2007] [Accepted: 06/11/2007] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the potential use of astatine-211 (211At)-labeled trastuzumab for the treatment of HER-2-positive, radioresistant ovarian carcinoma. METHODS AND MATERIALS Four-week-old nude mice were inoculated intraperitoneally with 5 . 10(6) SKOV-3 cells in 0.4 mL saline on Day 0. The endpoint was the total tumor weight in each mouse on Day 63. Three experiments were performed in which the response to single-dose and fractionated treatment with unlabeled and 211At-labeled antibody was evaluated. RESULTS Experiment 1 showed, for the same total amount of trastuzumab, a dose-response relationship between 211At activity (0-400 kBq on Day 7) and therapeutic efficacy (p = 0.001). The effect of varying the amount of unlabeled trastuzumab was studied in Experiment 2. All mice, except for the controls, received 400 kBq 211At-trastuzumab, and different groups received 5, 50, or 500 microg trastuzumab on Day 7. The increase from 5 to 50 microg trastuzumab reduced the tumors by 78% in weight. No tumors were present in mice given 500 microg trastuzumab. In Experiment 3, the effect of a fractionated treatment regimen was studied. Mice that received 100 kBq 211At-trastuzumab on Days 7 and 8 had a 42% smaller tumor burden than did controls. Groups of mice injected with 200 + 100 kBq on Days 7 and 21 and mice injected with 100 kBq on Days 7, 8, and 21 both had 24% less tumor weight than the corresponding controls. CONCLUSION The combination of 500 microg trastuzumab and 400 kBq 211At-trastuzumab had the greatest effect, with complete eradication of the tumors in this nude mouse model.
Collapse
Affiliation(s)
- Stig Palm
- Department of Radiation Physics, Sahlgrenska Academy at Göteborg University, Göteborg, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Fractionated radioimmunotherapy of intraperitoneally growing ovarian cancer in nude mice with 211At-MX35 F(ab')2: therapeutic efficacy and myelotoxicity. Nucl Med Biol 2006; 33:1065-72. [PMID: 17127181 DOI: 10.1016/j.nucmedbio.2006.07.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Revised: 07/25/2006] [Accepted: 07/29/2006] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the therapeutic efficacy and myelotoxicity during fractionated radioimmunotherapy of ovarian cancer in mice. The study was performed using the monoclonal antibody MX35 F(ab')(2) labeled with the alpha-particle emitter (211)At. METHODS Animals were intraperitoneally inoculated with approximately 1x10(7) cells of the cell line NIH:OVCAR-3. Four weeks later, the mice were given the first treatment. Six groups of animals were intraperitoneally injected with approximately 800, 3x approximately 267, approximately 400, 3x approximately 133, approximately 50 or 3x approximately 17 kBq (211)At-MX35 F(ab')(2) (n=18 in each group). The second and third injections for Groups 2, 4 and 6 were given 4 and 8 days after the first injection, respectively. As controls, animals were treated with unlabeled MX35 F(ab')(2) (n=12). Eight weeks after the last injection, the animals were sacrificed and the presence of macro- and microscopic tumors and ascites was determined. Blood counts were determined for each mouse in Groups 1 and 2 before the first injection and 3, 7, 11, 15 and 23 days after the first injection. The calculation of the mean absorbed dose to the bone marrow was based on the ratio between the (211)At-activity concentration in bone and blood [i.e., the bone-to-blood ratio (BBLR)] as well as that between the (211)At-activity concentration in bone marrow and blood [i.e., the bone-marrow-to-blood ratio (BMBLR)] and the cumulated activity and absorbed fraction of the alpha-particles emitted by (211)At in the bone marrow. RESULTS The tumor-free fractions of animals were 56% and 41% when treated with approximately 800 kBq and 3x approximately 267 kBq (211)At-MX35 F(ab')(2), respectively; 39% and 28% when treated with approximately 400 kBq and 3x approximately 133 kBq (211)At-MX35 F(ab')(2), respectively; and 17% and 22% when treated with approximately 50 kBq or 3x approximately 17 kBq (211)At-MX35 F(ab')(2), respectively. The nadir of the white blood cell (WBC) counts was decreased (from 46% to 19%, compared with the baseline WBC counts) and delayed (from Day 4 to Day 11 after the first injection) during the fractionated treatment compared with the single-dose treatment. The percentage of injected activity per gram (%IA/g) for blood, bone and bone marrow all peaked 6 h after injection at 13.80+/-1.34%IA/g, 4.00+/-0.69%IA/g and 8.28+/-1.38%IA/g, respectively. The BBLR and BMBLR were 0.20+/-0.04 and 0.58+/-0.01, respectively. The mean absorbed dose to bone marrow was approximately 0.4 Gy after intraperitoneally injecting approximately 800 kBq (211)At-MX35 F(ab')(2). CONCLUSION No advantage was observed in the therapeutic efficacy of using a fractionated regimen compared with a single administration, with the same total amount of administered activity. Alleviation of the myelotoxicity was observed during the fractionated regimen in terms of decreased suppression and delayed nadir of the WBC counts. No thrombocytopenia was observed during either regimen.
Collapse
|
13
|
Vallabhajosula S, Goldsmith SJ, Kostakoglu L, Milowsky MI, Nanus DM, Bander NH. Radioimmunotherapy of prostate cancer using 90Y- and 177Lu-labeled J591 monoclonal antibodies: effect of multiple treatments on myelotoxicity. Clin Cancer Res 2006; 11:7195s-7200s. [PMID: 16203821 DOI: 10.1158/1078-0432.ccr-1004-0023] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Bone marrow is the dose-limiting organ in radioimmunotherapy. Fractionated dose regimens may decrease myelotoxicity and increase greater total administered dose. We have studied the effect of two or three treatments of 177Lu-J591 and 90Y-J591 monoclonal antibodies (mAb) on myelotoxicity. EXPERIMENTAL DESIGN J591 is a deimmunized anti-PSMA mAb. Seven groups of patients with prostate cancer (n = 35) received 10 to 75 mCi/m2 of 177Lu-J591 and five additional groups (n = 28) received 5 to 20 mCi/m2 of 90Y-J591. Fifteen patients received two to three treatments of 177Lu-J591 (30, 45, or 60 mCi/m2) and four patients received two or three doses of 90Y-J591 (17.5 or 20 mCi/m2). Re-treatment consisted of patients receiving the same 177Lu or 90Y dose as their initial cycle. Time between treatments was 2 to 4 months. RESULTS The single dose maximum tolerated dose was 70 mCi/m2 with 177Lu-J591 and 17.5 mCi/m2 with 90Y-J591. With a single dose of 177Lu, no severe toxicity was observed below 60 mCi/m2. With 177Lu, two doses of 45 or 60 mCi/m2, totaling 90 to 120 mCi/m2, proved to be quite toxic. Three doses of 30 mCi/m2 (total 90 mCi/m2), however, were well tolerated. With 90Y, four patients tolerated two to three doses of 17.5 or 20 mCi/m2. Thrombocytopenia increased at higher doses and after repeat treatments. At higher doses, the nadir was lower and the time to reach nadir was longer. Time for recovery of platelets seems related to the total dose. CONCLUSIONS Multiple (two or three) administrations of 177Lu-J591 (30-60 mCi/m2) or 90Y-J591 (17.5 mCi/m2) over a 4- to 6-month period were tolerated by the patients with manageable thrombocytopenia. Although a single large dose may deliver optimal radiation dose to kill a larger fraction of tumor cells, fractionated therapy offers the advantage of lower myelotoxicity and prolonged tumor response. With 177Lu-J591, dose fractionation in combination with taxanes should be considered as an alternative approach to achieve optimal therapeutic efficacy in patients with prostate cancer.
Collapse
Affiliation(s)
- Shankar Vallabhajosula
- Department of Radiology, Division of Nuclear Medicine, New York Presbyterian Hospital, Weill Medical College of Cornell University, New York 10021, USA.
| | | | | | | | | | | |
Collapse
|
14
|
Janssen M, Frielink C, Dijkgraaf I, Oyen W, Edwards DS, Liu S, Rajopadhye M, Massuger L, Corstens F, Boerman O. Improved tumor targeting of radiolabeled RGD peptides using rapid dose fractionation. Cancer Biother Radiopharm 2005; 19:399-404. [PMID: 15453954 DOI: 10.1089/cbr.2004.19.399] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Arginine-glycine-aspartic acid (RGD) peptides preferentially bind to alphavbeta3 integrin, an integrin expressed on newly formed endothelial cells and on various tumor cells. When labeled with beta-emitting radionuclides, these peptides can be used for peptide-receptor radionuclide therapy of malignant tumors. These studies aimed to investigate whether tumor targeting and tumor therapy could be optimized by dose fractionation. The RGD-peptide DOTA-E-[c(RGDfK)]2 was labeled with 111In for biodistribution experiments and with 90Y for therapy experiments. In mice with NIH:OVCAR-3 ovarian carcinoma xenografts, optimal tumor uptake was obtained at peptide doses up to 1.0 microg (4.8 %ID/g). A peptide dose of 5 microg, required to administer the maximum tolerable dose (MTD) 90Y-DOTA-E-[c(RGDfK)]2, was administered as 5 portions of 1.0 microg. Tumor uptake of the fifth portion was significantly higher than that of the single 5.0 microg portion (3.3 %ID/g versus 2.1 %ID/g). The therapeutic efficacy of 37 MBq 90Y-DOTA-E-[c(RGDfK)]2 (1 x 5.0 microg) was compared with that of 37 MBq administered in five equal portions (5 x 1.0 microg). No difference in tumor growth between the fractionated and the nonfractionated therapy was observed. In conclusion, dose fractionation resulted in higher radiation doses. However, therapeutic efficacy of the radiolabeled peptide was not significantly improved by dose fractionation.
Collapse
Affiliation(s)
- Marcel Janssen
- Department of Nuclear Medicine, University Medical Center Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|