1
|
Goto H, Shiraishi Y, Okada S. Continuing progress in radioimmunotherapy for hematologic malignancies. Blood Rev 2025; 69:101250. [PMID: 39609167 DOI: 10.1016/j.blre.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024]
Abstract
Radioimmunotherapy (RIT) involves combining a cytotoxic radionuclide with an antibody (Ab) targeting a tumor antigen. Compared with conventional therapies, RIT improves the therapeutic efficacy of Ab and ameliorates toxicity. This comprehensive review describes the current advancements and future prospects in RIT for treating hematologic malignancies based on recent investigations. Although β-particle RITs targeting CD20 are effective with low toxicity in patients with relapsed/refractory indolent or transformed non-Hodgkin's lymphoma, these treatments have not gained popularity because of the increasing availability of new therapies. RIT using single-domain antibodies is expected to improve tumor penetrance and reduce radiation exposure to non-target organs. To enhance RIT efficacy, α-particle RIT and pretargeted radioimmunotherapy (PRIT) are currently being developed. Alpha-particle RIT demonstrates substantial antitumor activity and reduced bystander effects due to its high linear energy transfer and short particle range. PRIT may increase the tumor-to-whole body dose ratio.
Collapse
Affiliation(s)
- Hiroki Goto
- Division of Radioisotope and Tumor Pathobiology, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| | - Yoshioki Shiraishi
- Radioisotope Center, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| |
Collapse
|
2
|
Benedetto R, Massicano AVF, Silva JJ, Boas CAWV, Mengatti J, Araújo EBD. Development of radioimmunoconjugate for diagnosis and management of head-and-neck subclinical cancer and colorectal carcinoma. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902017000417039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
3
|
Jang BS, Lee SM, Kim HS, Shin IS, Razjouyan F, Wang S, Yao Z, Pastan I, Dreher MR, Paik CH. Combined-modality radioimmunotherapy: synergistic effect of paclitaxel and additive effect of bevacizumab. Nucl Med Biol 2011; 39:472-83. [PMID: 22172384 DOI: 10.1016/j.nucmedbio.2011.10.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 10/24/2011] [Accepted: 10/28/2011] [Indexed: 11/19/2022]
Abstract
INTRODUCTION This study was undertaken to investigate the effect of paclitaxel and bevacizumab on the therapeutic efficacy of (90)Y-labeled B3 monoclonal antibody, directed against Le(y) antigen, for the treatment of Le(y)-positive A431 tumors implanted subcutaneously in the right hind flank of nude mice. METHODS When the tumor size reached ~200 mm(3), the mice received a single dose of intravenous (iv) (90)Y-labeled B3 (60 μCi/150 μg or 100 μCi/150 μg B3), intraperitoneal paclitaxel (40 mg/kg) or iv bevacizumab (5 mg/kg) for monotherapy. To investigate the effect of combined therapies on survival, the mice were treated with two or three agents in the following combinations: (90)Y-B3 on day 0 and paclitaxel on day 1; bevacizumab on -1 day and (90)Y-B3 on day 0; bevacizumab on -1 day and paclitaxel on day 1; bevacizumab, (90)Y-B3 and paclitaxel each at 1-day intervals. The mice with no treatment were used as a control. The tumor volume at 1000 mm(3) was used as a surrogate end point of survival. RESULTS Compared to control animals, paclitaxel delayed tumor growth with a significantly longer median survival time (P<.001), whereas bevacizumab alone showed a less pronounced effect on a median survival time (P=.18). (90)Y-B3 increased the median survival time in a dose-dependent manner (P<.05). The combined therapy of bevacizumab with paclitaxel produced a trend toward an increase of the median survival time compared to paclitaxel alone (P=.06), whereas bevacizumab combined with (90)Y-B3 showed a statistically insignificant increase in the median survival time compared to (90)Y-B3 alone (P=.25). The tumor sizes of all animals in these groups reached the surrogate end point of survival by day 35. In contrast, the combined therapy involving (90)Y-B3 with paclitaxel showed a striking synergistic effect in shrinking tumors and prolonging the survival time (P<.001); on day 120, three of nine mice (33%) and six of six mice (100%) were alive without tumor when treated with 60 μCi (90)Y-B3 and 100 μCi (90)Y-B3, respectively. The addition of bevacizumab treatment 1 day before the combined therapy of 60 μCi (90)Y-B3 with paclitaxel did not produce a statistically significant increase in survival when compared to the (90)Y-B3 with paclitaxel (P>.10). Fluorescence microscopy analysis indicated that paclitaxel increased, whereas bevacizumab decreased, the accumulation and penetration of Alexa Fluor 647-B3 into tumor microenvironment compared to the control (P<.05). CONCLUSION Our findings on the paclitaxel effect support a hypothesis that the increased tumor accumulation and penetration of (90)Y-B3 as well as the high radiosensitization of tumor cells by paclitaxel may be the major factors responsible for the synergistic effect of the combined therapy involving (90)Y-B3 with paclitaxel.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Bevacizumab
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/radiotherapy
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Proliferation/radiation effects
- Combined Modality Therapy
- Drug Synergism
- Humans
- Mice
- Microscopy, Fluorescence
- Microvessels/drug effects
- Microvessels/metabolism
- Microvessels/radiation effects
- Paclitaxel/pharmacology
- Paclitaxel/therapeutic use
- Radioimmunotherapy/methods
- Xenograft Model Antitumor Assays
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
- Beom-Su Jang
- Radiopharmaceutical Laboratory, Nuclear Medicine, Clinical Center, National Cancer Institute/NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Shin IS, Lee SM, Kim HS, Yao Z, Regino C, Sato N, Cheng KT, Hassan R, Campo MF, Albone EF, Choyke PL, Pastan I, Paik CH. Effect of chelator conjugation level and injection dose on tumor and organ uptake of 111In-labeled MORAb-009, an anti-mesothelin antibody. Nucl Med Biol 2011; 38:1119-27. [PMID: 21741258 DOI: 10.1016/j.nucmedbio.2011.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 04/27/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Radiolabeling of a monoclonal antibody (mAb) with a metallic radionuclide requires the conjugation of a bifunctional chelator to the mAb. The conjugation, however, can alter the physical and immunological properties of the mAb, consequently affecting its tumor-targeting pharmacokinetics. In this study, we investigated the effect of the amount of 2-(p-isothiocyanatobenzyl)-cyclohexyl-diethylenetriamine-pentaacetic acid (CHX-A″) conjugated to MORAb-009, a mAb directed against mesothelin, and the effect of MORAb dose on the biodistribution of (111)In-labeled MORAb-009. METHODS We used nude mice bearing the A431/K5 tumor as a mesothelin-positive tumor model and the A431 tumor as a mesothelin-negative control. To find the optimal level of CHX-A″ conjugation, CHX-A″-MORAb-009 conjugates with 2.4, 3.5 and 5.5 CHX-A″ molecules were investigated. To investigate the effect of injected MORAb-009 dose on neutralizing the shed mesothelin in the circulation, biodistribution studies were performed after the intravenous co-injection of (111)In-labeled MORAb-009 (2.4 CHX-A″/MORAb-009) with three different doses: 0.2, 2 and 30 μg of MORAb-009. RESULTS The tumor uptake in A431/K5 tumor was four times higher than that in A431 tumor, indicating that the tumor uptake in A431/K5 was mesothelin mediated. The conjugate with 5.5 CHX-A″ showed a lower isoelectric point (pI) and lower immunoreactivity (IR) than the 2.4 CHX-A″ conjugate. These differences were reflected in the biodistribution of the (111)In label. The (111)In-labeled MORAb-009 conjugated with 2.4 CHX-A″ produced higher tumor uptake and lower liver and spleen uptakes than the 5.5 CHX-A″ conjugate. The biodistribution studies also revealed that the tumor uptake was significantly affected by the injected MORAb-009 dose and tumor size. The 30-μg dose produced higher tumor uptake than the 0.2- and 2-μg doses, whereas the 30-μg dose produced lower liver and spleen uptakes than the 0.2-μg dose. CONCLUSION This study demonstrates that the number of chelate conjugation and the injected dose are two important parameters to achieve high tumor and low non-target organ uptake of (111)In-labeled MORAb-009. This study also suggests that the injected dose of mAb could be individualized based on the tumor size or the blood level of shed antigen in a patient to achieve the ideal tumor-to-organ radioactivity ratios.
Collapse
Affiliation(s)
- In Soo Shin
- Radiopharmaceutical Laboratory, Division of Nuclear Medicine, Department of Radiology and Imaging Sciences, NIH Clinical Center, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
González GP, García IG, González JG, Sánchez LP, Mirabal MV, Marín CC, Ruiz FLG, Iglesias EG, de Queralta RL, Toirac RR, Ávila MA, Díaz AL, Saura PAL, Gavilondo JV, González JPO. Phase I Clinical Trial of the 131I-Labeled Anticarcinoembryonic Antigen CIGB-M3 Multivalent Antibody Fragment. Cancer Biother Radiopharm 2011; 26:353-63. [DOI: 10.1089/cbr.2010.0899] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Idrian García García
- Clinical Trials Division, Cancer Research Department, Center for Genetic Engineering and Biotechnology (CIGB), La Habana, Cuba
| | | | - Lincidio Pérez Sánchez
- Recombinant Antibody Group Laboratory, Pharmaceuticals Division, Cancer Research Department, Center for Genetic Engineering and Biotechnology (CIGB), La Habana, Cuba
| | | | - Carlos Calderón Marín
- Nuclear Medicine Department, National Institute of Oncology and Radiobiology, La Habana, Cuba
| | - Fausto L. García Ruiz
- Nuclear Medicine Department, National Institute of Oncology and Radiobiology, La Habana, Cuba
| | - Elizeth Garcia Iglesias
- Clinical Trials Division, Cancer Research Department, Center for Genetic Engineering and Biotechnology (CIGB), La Habana, Cuba
| | | | - Ramón Ropero Toirac
- Nuclear Medicine Department, National Institute of Oncology and Radiobiology, La Habana, Cuba
| | - Marta Ayala Ávila
- Recombinant Antibody Group Laboratory, Pharmaceuticals Division, Cancer Research Department, Center for Genetic Engineering and Biotechnology (CIGB), La Habana, Cuba
| | - Adlín López Díaz
- Nuclear Medicine Department, “Hermanos Ameijeiras” Hospital, La Habana, Cuba
| | - Pedro A. López Saura
- Clinical Trials Division, Cancer Research Department, Center for Genetic Engineering and Biotechnology (CIGB), La Habana, Cuba
| | - Jorge V. Gavilondo
- Recombinant Antibody Group Laboratory, Pharmaceuticals Division, Cancer Research Department, Center for Genetic Engineering and Biotechnology (CIGB), La Habana, Cuba
| | - Juan P. Oliva González
- Nuclear Medicine Department, National Institute of Oncology and Radiobiology, La Habana, Cuba
| |
Collapse
|
6
|
Nanomolecular HLA-DR10 antibody mimics: A potent system for molecular targeted therapy and imaging. Cancer Biother Radiopharm 2010; 23:783-96. [PMID: 20443696 DOI: 10.1089/cbr.2008.0589] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To mimic the molecular specificity and cell selectivity of monoclonal antibody (mAb) binding while decreasing size, nanomolecules (selective high-affinity ligands; SHALs), based on in silico modeling, have been created to bind to human leukocyte antigen-DR (HLA-DR10), a signaling receptor protein upregulated on the malignant B-lymphocytes of non-Hodgkin's lymphoma and chronic lymphocytic leukemia. SHALs were synthesized with a biotin or DOTA chelate (1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid), using a solid-phase lysine-polyethyleneglycol backbone to link sets of ligands shown previously to bind to HLA-DR10. Using cell-binding and death assays and confocal microscopy, SHAL uptake, residualization, and cytocidal activity were evaluated in HLA-DR10 expressing and nonexpressing live, human lymphoma cell lines. All of the SHALs tested were selective for, and accumulated in, expressing cells. Reflecting binding to HLA-DR10 inside the cells, SHALs having the Ct ligand (3-(2-([3-chloro-5-trifluoromethyl)-2-pyridinyl]oxy)-anilino)-3-oxopropanionic acid) residualized in expressing cells greater than 179 times more than accountable by cell-surface membrane HLA-DR10. Confocal microscopy confirmed the intracellular residualization of these SHALs. Importantly, SHALs with a Ct ligand had direct cytocidal activity, similar in potency to that of Lym-1 mAb and rituximab, selectively for HLA-DR10 expressing lymphoma cells and xenografts. The results show that SHALs containing the Ct ligand residualize intracellularly and have cytocidal effects mediated by HLA-DR10. These SHALs have extraordinary potential as novel molecules for the selective targeting of lymphoma and leukemia for molecular therapy and imaging. Further, these SHALs can be used to transport and residualize cytotoxic agents near critical sites inside these malignant cells.
Collapse
|
7
|
DeNardo G, DeNardo S. Dose intensified molecular targeted radiotherapy for cancer-lymphoma as a paradigm. Semin Nucl Med 2010; 40:136-44. [PMID: 20113681 DOI: 10.1053/j.semnuclmed.2009.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although most patients with locoregional cancer are cured by surgery, radiotherapy, chemotherapy, and combinations thereof, those with distant metastases are not despite systemic chemotherapy. These patients respond to local radiotherapy but generally need systemic therapy. Non-Hodgkin's lymphoma (NHL) provides a paradigm for the role of molecular targeted radiotherapy (MTRT) because these patients have multifocal disease in most cases. Although patients with NHL achieve remissions after multiple cycles of chemotherapy, less than one half of those with aggressive NHL are cured and almost none of those with low grade NHL. Furthermore, NHL, like other cancers, becomes chemoresistant, yet remains responsive to radiotherapy. MTRT, radiation targeted by molecules, is a good strategy for the treatment of multifocal and radiosensitive cancers. Radioimmunotherapy (RIT) is an MTRT approach using MAbs, or parts thereof, to target the radionuclide that delivers radiation. Two anti-CD20 monoclonal antibodies (MAbs), one labeled with (111)In for imaging or (90)Y for therapy and a second labeled with (131)I for imaging and therapy, have proven effective and safe for MTRT for NHL patients. The importance of the radiation is demonstrated in the data from the randomized pivotal trial of (90)Y-ibritumomab; response rates were distinctly better in the (90)Y-ibritumomab arm than in the rituximab arm. Furthermore, the efficacy of (131)I-tositumomab was greater than that of the same MAb alone in another pivotal trial. Although hematologic toxicity is dose limiting for MTRT, febrile neutropenia is uncommon. MTRT is also not associated with mucositis, hair loss, or persistent nausea or vomiting, unlike chemotherapy. Randomized trials of MTRT in different strategies have not been conducted, but there is evidence of better outcomes, particularly for strategies that provide dose intensification, such as pretargeted MTRT, multiple dosing ("fractionation"), and MTRT with stem cell transplantation (SCT). Pretargeted RIT separates delivery of the targeting molecule from radionuclide delivery, provides dose escalation, and is more effective than direct one-step RIT, although more complicated to implement. Improved drugs and strategies for MTRT have documented potential for better patient outcomes. Smaller radionuclide carriers, such as those used for pretargeted MTRT, should be incorporated into the management of patients with NHL and other cancers soon after the patients have proven incurable. Expected improvements using better drugs, strategies, and combinations with other drugs seem likely to make MTRT integral in the management of many patients with cancer and likely to lead to cures of NHL.
Collapse
Affiliation(s)
- Gerald DeNardo
- Davis Medical Center, University of California, Sacramento, CA, USA.
| | | |
Collapse
|
8
|
Abstract
This article provides an overview of a semiempirical pretargeting model now under development. After a brief review of the pretargeting concept, the strategies available, and the complexities of optimizing the dosage and timing, a semiempirical model is described that is not only capable of optimizing dosage and timing but also capable of predicting the results of pretargeting as a function of most pretargeting variables. The model requires knowledge of the pharmacokinetics of both the pretargeting agent (usually an antibody) and the effector, the accessibility of the pretargeting antibody for the effector, and their quantitative relationships in vivo. Several misconceptions that often surround pretargeting are also clarified.
Collapse
Affiliation(s)
- Guozheng Liu
- Department of Radiology, Division of Nuclear Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | |
Collapse
|
9
|
DeNardo GL, DeNardo SJ, Balhorn R. Systemic radiotherapy can cure lymphoma: a paradigm for other malignancies? Cancer Biother Radiopharm 2008; 23:383-97. [PMID: 18771343 DOI: 10.1089/cbr.2007.0523-u] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The cytocidal potency of a molecule can be augmented by conjugating a radionuclide for molecular targeted radionuclide therapy (MTRT) for cancer. Radioimmunotherapy (RIT) should be incorporated into the management of patients with B-cell non-Hodgkin's lymphoma (NHL) soon after the patients have proven incurable. Better drugs, strategies, and combinations with other drugs seem certain to make RIT integral to the management of patients with NHL and likely to lead to a cure of the currently incurable NHL. These improved drugs, strategies, and combinations thereof also offer opportunities for RIT to become part of the management of solid malignancies, including epithelial cancers. Smaller radionuclide carriers, such as those used for pretargeted strategies, provide dose intensification. The potential of pretargeted RIT to improve patient outcomes is striking.
Collapse
Affiliation(s)
- Gerald L DeNardo
- Department of Internal Medicine, University of California, Davis, Sacramento, CA 95816, USA.
| | | | | |
Collapse
|
10
|
Fidarova EF, El-Emir E, Boxer GM, Qureshi U, Dearling JLJ, Robson MP, Begent RHJ, Trott KR, Pedley RB. Microdistribution of targeted, fluorescently labeled anti-carcinoembryonic antigen antibody in metastatic colorectal cancer: implications for radioimmunotherapy. Clin Cancer Res 2008; 14:2639-46. [PMID: 18451227 DOI: 10.1158/1078-0432.ccr-07-2031] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Most radioimmunotherapy studies on radiolabeled antibody distribution are based on autoradiographic and radioluminographic data, which provide a lack of detailed information due to low resolution. We used fluorescently labeled anti-carcinoembryonic antigen (CEA) antibody (A5B7) to investigate quantitatively the kinetics and microdistribution of antibody in a clinically relevant orthotopic colorectal cancer model (LS174T) using high-resolution digital microscopy. EXPERIMENTAL DESIGN Nude mice bearing LS174T liver orthotopic tumors received a single i.v. injection of fluorescently labeled A5B7 and were sacrificed at 10 minutes, 1 hour, or 24 hours postinjection. Before sacrifice, mice were injected with the perfusion marker Hoechst 33342. An anti-CD31 antibody was used to detect blood vessel distribution. Cryostat sections were processed with immunofluorescence procedures and analyzed with fluorescence microscopy and image analysis techniques. The fluorescence images were related to morphologic images of the same or adjacent tumor sections. RESULTS Fluorescently labeled antibody showed rapid, selective uptake into tumor deposits, with a strong negative correlation with tumor size at 10 minutes and 1 hour (P < or = 0.01). By 24 hours, the correlation was no longer significant. The study showed movement of antibody across the tumor with time and a tendency to localize more uniformly by later time points (24 hours). The rate of antibody motility was similar in small and large tumor metastases, but small deposits showed more rapid antibody localization. Intratumoral vessels were positively related to tumor size (P < or = 0.001). CONCLUSION The obtained data suggest that radioimmunotherapy can be highly efficient in an adjuvant or minimal residual disease setting.
Collapse
Affiliation(s)
- Elena F Fidarova
- Cancer Research UK Targeting and Imaging Group, Department of Oncology, University College London, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
|
12
|
Nemecek ER, Green DJ, Fisher DR, Pagel JM, Lin Y, Gopal AK, Durack LD, Rajendran JG, Wilbur DS, Nilsson R, Sandberg B, Press OW. Extracorporeal Adsorption Therapy: A Method to Improve Targeted Radiation Delivered by Radiometal-Labeled Monoclonal Antibodies. Cancer Biother Radiopharm 2008; 23:181-91. [DOI: 10.1089/cbr.2007.0433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Eneida R. Nemecek
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Damian J. Green
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | - John M. Pagel
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Yukang Lin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| | - Ajay K. Gopal
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | | | - D. Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | | | | | - Oliver W. Press
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
13
|
El Emir E, Qureshi U, Dearling JL, Boxer GM, Clatworthy I, Folarin AA, Robson MP, Nagl S, Konerding MA, Pedley RB. Predicting Response to Radioimmunotherapy from the Tumor Microenvironment of Colorectal Carcinomas. Cancer Res 2007; 67:11896-905. [DOI: 10.1158/0008-5472.can-07-2967] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
DeNardo GL. Editorial:The Conundrum of Personalized Cancer Management, Drug Development, and Economics. Cancer Biother Radiopharm 2007; 22:719-21. [DOI: 10.1089/cbr.2007.500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Gerald L. DeNardo
- Division of Hematology and Oncology, Department of Internal Medicine, University of California–Davis, Davis, CA
| |
Collapse
|
15
|
Schillaci O, DeNardo GL, DeNardo SJ, Goldstein DS, Kroger LA, O'Donnell RT, Lamborn KR. Effect of Antilymphoma Antibody,131I-Lym-1, on Peripheral Blood Lymphocytes in Patients with Non-Hodgkin's Lymphoma. Cancer Biother Radiopharm 2007; 22:521-30. [PMID: 17803447 DOI: 10.1089/cbr.2007.374a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Anti-CD20 monoclonal antibodies (mAbs), unlabeled rituximab (Rituxan, Biogen Idec Inc., Cambridge, MA; and Genentech Inc., South San Francisco, CA) or radiolabeled 90Y-ibritumomab (Zevalin, Biogen Idec Inc., Cambridge, MA) and 131I-tositumomab (Bexxar; Glaxo Smith Kline, Research Triangle Park, NC), have proven to be effective therapy for non-Hodgkin's lymphoma (NHL), but also induce immediate and persistent decreases in normal peripheral blood lymphocytes (PBLs). Lym-1, a mAb that selectively targets malignant lymphocytes, also has induced therapeutic responses and prolonged survival in patients with NHL when labeled with iodine-131 (131I). We have retrospectively examined its effect on PBLs in 41 NHL patients that had received 131I-Lym-1 therapy. Absolute lymphocyte counts (ALCs) were evaluated before and after the first and last 131I-Lym-1 infusion. Modest decreases in PBLs were observed in most of the patients. Using strict criteria to define recovery, time to recovery was determined for 19 patients, with the remainder censored because of insufficient follow-up (median follow up for censored patients: 22 days). Using Kaplan-Meier estimates, it would be predicted that 31% of patients would recover by 28 days and that median time to recovery would be 44 days after the last 131I-Lym-1 infusion. No predictors were found for time to recovery, considering such factors as the administered Lym-1 or 131I dose, spleen volume, or radiation doses to the body, marrow, or spleen. The data suggest that the effect of 131I-Lym-1 on ALC is the result of a nonspecific radiation effect, rather than a specific Lym-1 mAb effect. The shorter time required for ALC recovery after 131I-Lym-1 when compared to that reported for anti-CD20 mAbs, whether radiolabeled or otherwise, is probably related to differing mechanisms for lymphocytotoxicity and lesser Lym-1 antigenic density on normal B-lymphocytes.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/adverse effects
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/therapeutic use
- Cell Survival/drug effects
- Female
- Humans
- Kaplan-Meier Estimate
- Lymphocytes/immunology
- Lymphocytes/pathology
- Lymphoma, Non-Hodgkin/immunology
- Lymphoma, Non-Hodgkin/pathology
- Lymphoma, Non-Hodgkin/radiotherapy
- Male
- Middle Aged
- Radioimmunotherapy
Collapse
Affiliation(s)
- Orazio Schillaci
- Department of Biopathology and Diagnostic Imaging, University Tor Vergata, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
West J, Perkins J, Hok S, Balhorn R, Lightstone FC, Cosman M, DeNardo SJ, DeNardo GL. Direct antilymphoma activity of novel, first-generation "antibody mimics" that bind HLA-DR10-positive non-Hodgkin's lymphoma cells. Cancer Biother Radiopharm 2007; 21:645-54. [PMID: 17257080 DOI: 10.1089/cbr.2006.21.645] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A first-generation series of novel small molecules, collectively known as selective high-affinity ligands (SHALs), were designed and synthesized to mimic the binding of Lym-1, a monoclonal antibody (mAb) shown to be an effective cytotoxic and radionuclide carrier molecule for targeting non-Hodgkin's lymphoma (NHL). Created as radionuclide targeting molecules, these SHALs were intended to have the human leukocyte antigen-DR (HLA-DR) selectivity of Lym-1 mAb and the pharmacokinetics of a small molecule. Because of the remarkable bioactivity of Lym-1 in vitro, the direct antilymphoma activity of three of these SHALs was tested. Two of these SHALs were bidentate and consisted of two ligands connected to the carboxyl and amino groups of lysine and polyethylene glycol (PEG); the third SHAL was a dimeric version of one of the former two SHALs linked with PEG. The three SHALs tested were: LeLPLDB, that contained one deoxycholate and one 5-leu-enkephalin as ligands; (LeacPLD)2LPB, a bis version of LeLPLDB intended to improve "functional affinity"; and ItPLDB, that contained the ligands, deoxycholate and triiodothyronine. Micromolar concentrations of all three SHALs showed binding to Raji, an HLA-DR10-positive human malignant B-cell line but no binding to CEM or Jurkat's, HLA-DR10-negative malignant T-cell lines. Additionally, the Raji cell membrane distributions of all three SHALs and of Lym-1 were remarkably similar. Unlike Lym-1, which causes substantial growth inhibition and cell death in NHL cell lines, these SHALs had no direct antilymphoma activity. In summary, three first-generation SHALs lacked direct antilymphoma activity, although they had selective NHL B-cell binding like Lym-1 mAb. Because of their small size, these SHALs have potential as radionuclide carrier substitutes for Lym-1 mAb to target the HLA-DR10 NHL-related cell-surface protein.
Collapse
Affiliation(s)
- Jeremy West
- Department of Internal Medicine and Radiology, University of California, Davis, Sacramento, CA 95816, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Jain M, Venkatraman G, Batra SK. Optimization of radioimmunotherapy of solid tumors: biological impediments and their modulation. Clin Cancer Res 2007; 13:1374-82. [PMID: 17309914 DOI: 10.1158/1078-0432.ccr-06-2436] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In contrast to the overwhelming success of radiolabeled antibodies in treating hematologic malignancies, only modest success has been achieved in the radioimmunotherapy of solid tumors. One of the major limitations in successful application of radioimmunotherapy is the large molecular size of the intact immunoglobulin that results in prolonged serum half-life and poor tumor penetration and uptake. With the advent of antibody engineering, small molecular weight antibody fragments exhibiting improved pharmacokinetics and tumor penetration have been generated. However, their clinical application has been limited by suboptimal tumor uptake and short tumor residence time. There is a greater realization that optimization of the molecular size of the antibodies alone is not sufficient for clinical success of radioimmunotherapy. In addition to their size, radiolabeled antibodies encounter other impediments before reaching their target antigens expressed on the cell surface of solid tumors. Some of the barriers include poor blood flow in large tumors, permeability of vascular endothelium, elevated interstitial fluid pressure of tumor stroma, and heterogeneous antigen expression. Recent research has considerably improved our understanding and appreciation of these forces, and the new wave of optimization strategies involves the use of biological modifiers to modulate the impediments posed by solid tumors. In combination with radiolabeled antibodies, various agents are being used to improve the tumor blood flow, enhance vascular permeability, lower tumor interstitial fluid pressure by modulating stromal cells and extracellular matrix components, up-regulate the expression of target antigens, and improve the penetration and retention of the radiopharmaceuticals. This review outlines ongoing research efforts involving biological modifiers to optimize the uptake and efficacy of radiolabeled antibodies for the treatment of solid tumors.
Collapse
Affiliation(s)
- Maneesh Jain
- Department of Biochemistry and Molecular Biology, Department of Pathology and Microbiology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| | | | | |
Collapse
|
18
|
Pitini V, Baldari S, Altavilla G, Arrigo C, Naro C, Perniciaro F. Salvage therapy for primary central nervous system lymphoma with 90Y-Ibritumomab and Temozolomide. J Neurooncol 2007; 83:291-3. [PMID: 17245621 DOI: 10.1007/s11060-006-9324-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Accepted: 12/26/2006] [Indexed: 11/25/2022]
Abstract
Aggressive initial treatment of Primary Central Nervous System Lymphoma (PCNSL) has achieved prolonged survival and occasional cures. However, some patients do not respond to initial therapy and others relapse after an initial remission. The optimal salvage regimen is not known and many different strategies have been proposed. In this report we describe the efficacy of a combination of (90)Y-Ibritumomab Tiuxetan (Zevalin) and Temozolamide as a maintenance therapy for recurrent PCNS Lymphoma in two patients that are both alive and in complete remission after 9 and 10 months respectively. This combination merits further study and provides a reasonable therapeutic alternative for older patients with progressive PCNSL.
Collapse
Affiliation(s)
- Vincenzo Pitini
- Medical Oncology (Pad. H 50 piano), University of Messina, Via Consolare Valeria, 98125, Messina, Italy.
| | | | | | | | | | | |
Collapse
|