To see the other types of publications on this topic, follow the link: Cancer – Radioimmunotherapy.

Journal articles on the topic 'Cancer – Radioimmunotherapy'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Cancer – Radioimmunotherapy.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Sharkey, Robert M., and David M. Goldenberg. "Cancer radioimmunotherapy." Immunotherapy 3, no. 3 (March 2011): 349–70. http://dx.doi.org/10.2217/imt.10.114.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Buraggi, G. L., and E. Seregni. "Radioimmunotherapy of cancer." Biomedicine & Pharmacotherapy 47, no. 6-7 (November 1993): 277. http://dx.doi.org/10.1016/0753-3322(93)90207-2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

&NA;. "Radioimmunotherapy in ovarian cancer." Inpharma Weekly &NA;, no. 996 (July 1995): 8. http://dx.doi.org/10.2165/00128413-199509960-00016.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

SYRIGOS, K. N., and A. A. EPENETOS. "Radioimmunotherapy of Ovarian Cancer." Hybridoma 14, no. 2 (April 1995): 121–24. http://dx.doi.org/10.1089/hyb.1995.14.121.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Crippa, F. "Radioimmunotherapy of Ovarian Cancer." International Journal of Biological Markers 8, no. 3 (July 1993): 187–91. http://dx.doi.org/10.1177/172460089300800309.

Full text
Abstract:
Radioimmunotherapy (RIT) is a new therapeutical approach where radiolabeled monoclonal antibodies (MAb) against tumor-associated antigens are administered to treat tumor lesions. Ovarian cancer is one of the most promising fields for RIT. This paper gives an overview of some biodistribution studies in animal models and in patients with radiolabeled anti-ovarian cancer MAbs, and defines the main criteria which should be considered to plan a clinical trial of RIT in ovarian cancer. As regards the clinical results, the published outcome of various trials and the experience of the National Cancer Institute of Milan are summarized. Even if the number of patients involved in these clinical studies of RIT is too small to provide conclusive indications about its role in the management of ovarian cancer, the preliminary results from qualified groups show its potential in this disease despite the current problems that limit clinical application (above all, the instability of the radiolabeled linkage, the immunogenicity of murine antibodies, the poor absolute tumor radiolabel uptake and the bone marrow toxicity).
APA, Harvard, Vancouver, ISO, and other styles
6

Bertagnolli, M. M. "Radioimmunotherapy for Colorectal Cancer." Clinical Cancer Research 11, no. 13 (July 1, 2005): 4637–38. http://dx.doi.org/10.1158/1078-0432.ccr-05-0485.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Koppe, M. J., R. P. Bleichrodt, W. J. G. Oyen, and O. C. Boerman. "Radioimmunotherapy and colorectal cancer." British Journal of Surgery 92, no. 3 (2005): 264–76. http://dx.doi.org/10.1002/bjs.4936.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Hull, Ashleigh, Yanrui Li, Dylan Bartholomeusz, William Hsieh, Barry Allen, and Eva Bezak. "Radioimmunotherapy of Pancreatic Ductal Adenocarcinoma: A Review of the Current Status of Literature." Cancers 12, no. 2 (February 19, 2020): 481. http://dx.doi.org/10.3390/cancers12020481.

Full text
Abstract:
Pancreatic ductal adenocarcinoma (PDAC) has long been associated with low survival rates. A lack of accurate diagnostic tests and limited treatment options contribute to the poor prognosis of PDAC. Radioimmunotherapy using α- or β-emitting radionuclides has been identified as a potential treatment for PDAC. By harnessing the cytotoxicity of α or β particles, radioimmunotherapy may overcome the anatomic and physiological factors which traditionally make PDAC resistant to most conventional treatments. Appropriate selection of target receptors and the development of selective and cytotoxic radioimmunoconjugates are needed to achieve the desired results of radioimmunotherapy. The aim of this review is to examine the growing preclinical and clinical trial evidence regarding the application of α and β radioimmunotherapy for the treatment of PDAC. A systematic search of MEDLINE® and Scopus databases was performed to identify 34 relevant studies conducted on α or β radioimmunotherapy of PDAC. Preclinical results demonstrated α and β radioimmunotherapy provided effective tumour control. Clinical studies were limited to investigating β radioimmunotherapy only. Phase I and II trials observed disease control rates of 11.2%–57.9%, with synergistic effects noted for combination therapies. Further developments and optimisation of treatment regimens are needed to improve the clinical relevance of α and β radioimmunotherapy in PDAC.
APA, Harvard, Vancouver, ISO, and other styles
9

STEWART, J. S. W., V. HIRD, M. SULLIVAN, D. SNOOK, and A. A. EPENETOS. "Intraperitoneal radioimmunotherapy for ovarian cancer." BJOG: An International Journal of Obstetrics and Gynaecology 96, no. 5 (May 1989): 529–36. http://dx.doi.org/10.1111/j.1471-0528.1989.tb03251.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Stewart, JSW, V. Hird, M. Sullivan, D. Snook, and AA Epenetos. "Intraperitoneal radioimmunotherapy for ovarian cancer." International Journal of Gynecology & Obstetrics 31, no. 1 (January 1990): 96. http://dx.doi.org/10.1016/0020-7292(90)90223-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Pecking, A. P., G. Delorme, and J. L. Floiras. "Radioimmunotherapy of serous ovarian cancer." International Journal of Radiation Oncology*Biology*Physics 19 (January 1990): 235. http://dx.doi.org/10.1016/0360-3016(90)90861-d.

Full text
APA, Harvard, Vancouver, ISO, and other styles
12

Zelenetz, Andrew D. "Radioimmunotherapy for lymphoma." Current Opinion in Oncology 11, no. 5 (September 1999): 375. http://dx.doi.org/10.1097/00001622-199909000-00009.

Full text
APA, Harvard, Vancouver, ISO, and other styles
13

Burke, Patricia A., Sally J. DeNardo, Laird A. Miers, David L. Kukis, and Gerald L. DeNardo. "Combined modality radioimmunotherapy." Cancer 94, S4 (February 12, 2002): 1320–31. http://dx.doi.org/10.1002/cncr.10303.

Full text
APA, Harvard, Vancouver, ISO, and other styles
14

Knox, Susan J., and Ruby F. Meredith. "Clinical radioimmunotherapy." Seminars in Radiation Oncology 10, no. 2 (April 2000): 73–93. http://dx.doi.org/10.1016/s1053-4296(00)80045-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
15

Buchsbaum, Donald J. "Experimental radioimmunotherapy." Seminars in Radiation Oncology 10, no. 2 (April 2000): 156–67. http://dx.doi.org/10.1016/s1053-4296(00)80052-1.

Full text
APA, Harvard, Vancouver, ISO, and other styles
16

Goldenberg, David M., Robert M. Sharkey, Giovanni Paganelli, Jacques Barbet, and Jean-François Chatal. "Antibody Pretargeting Advances Cancer Radioimmunodetection and Radioimmunotherapy." Journal of Clinical Oncology 24, no. 5 (February 10, 2006): 823–34. http://dx.doi.org/10.1200/jco.2005.03.8471.

Full text
Abstract:
This article reviews the methods of pretargeting, which involve separating the targeting antibody from the subsequent delivery of an imaging or therapeutic agent that binds to the tumor-localized antibody. This provides enhanced tumor:background ratios and the delivery of a higher therapeutic dose than when antibodies are directly conjugated with radionuclides, as currently practiced in cancer radioimmunotherapy. We describe initial promising clinical results using streptavidin-antibody constructs with biotin-radionuclide conjugates in the treatment of patients with malignant gliomas, and of bispecific antibodies with hapten-radionuclides in the therapy of tumors expressing carcinoembryonic antigen, such as medullary thyroid and small-cell lung cancers.
APA, Harvard, Vancouver, ISO, and other styles
17

Meredith, Ruby F., and Donald J. Buchsbaum. "Pretargeted radioimmunotherapy." International Journal of Radiation Oncology*Biology*Physics 66, no. 2 (October 2006): S57—S59. http://dx.doi.org/10.1016/j.ijrobp.2006.04.058.

Full text
APA, Harvard, Vancouver, ISO, and other styles
18

Choi, Ik Joon. "Radioimmunotherapy in Head and Neck Cancer." Korean Journal of Otorhinolaryngology-Head and Neck Surgery 61, no. 12 (December 21, 2018): 637–43. http://dx.doi.org/10.3342/kjorl-hns.2018.00696.

Full text
APA, Harvard, Vancouver, ISO, and other styles
19

Carlson, Robert H. "Radioimmunotherapy-Chemotherapy Targets Advanced Pancreatic Cancer." Oncology Times 36, no. 12 (June 2014): 54–55. http://dx.doi.org/10.1097/01.cot.0000451738.18539.ef.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Riva, Pietro, Giancarlo Franceschi, Rossella Gentile, Nada Riva, and Michela Casi. "Radioimmunodetection and Radioimmunotherapy of Breast Cancer." Tumori Journal 83, no. 2 (March 1997): 552–57. http://dx.doi.org/10.1177/030089169708300215.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Riva, P., V. Tison, A. Arista, C. Sturiale, G. Franceschi, N. Riva, M. Casi, G. Moscatelli, F. Campori, and A. Spinelli. "Radioimmunotherapy of Gastrointestinal Cancer and Glioblastomas." International Journal of Biological Markers 8, no. 3 (July 1993): 192–97. http://dx.doi.org/10.1177/172460089300800310.

Full text
Abstract:
Two groups of patients with gastro-intestinal (GI) tumours (41) and recurrent glioblastoma (GBM), (17) underwent radioimmunotherapy after the failure of traditional treatments. A number of different MAbs were employed (anti-CEA and anti-Tenascin) which were labelled with I-131. The radiopharmaceuticals were administered by the intraperitoneal and intratumoral routes. As a rule the cycles were repeated to enhance the effectiveness of RIT. No significant early or late adverse effects were recorded. HAMA development was observed in all GI cases but only in a few GBM patients. The cumulative dose delivered to the target tumors was considerable (mean 8,900 cGy) in the GI group, and was much higher in the GBM patients (mean 51,700 cGy) owing to the particular modality of injection. Survival improved in both series of patients. The objective responses to RIT were promising: in the GI group 10 complete remissions (CR) and 6 partial remissions (PR) were observed, while in the GBM group 3 long-lasting CRs and 3 prolonged PRs were documented.
APA, Harvard, Vancouver, ISO, and other styles
22

Couturier, Olivier, Stéphane Supiot, Marie Degraef-Mougin, Alain Faivre-Chauvet, Thomas Carlier, Jean-François Chatal, François Davodeau, and Michel Cherel. "Cancer radioimmunotherapy with alpha-emitting nuclides." European Journal of Nuclear Medicine and Molecular Imaging 32, no. 5 (April 2005): 601–14. http://dx.doi.org/10.1007/s00259-005-1803-2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Lam, L., X. Liu, and Y. Cao. "Pretargeted radioimmunotherapy, a potential cancer treatment." Drugs of the Future 28, no. 2 (2003): 167. http://dx.doi.org/10.1358/dof.2003.028.02.856929.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Larson, Steven M., Jorge A. Carrasquillo, Nai-Kong V. Cheung, and Oliver W. Press. "Radioimmunotherapy of human tumours." Nature Reviews Cancer 15, no. 6 (May 22, 2015): 347–60. http://dx.doi.org/10.1038/nrc3925.

Full text
APA, Harvard, Vancouver, ISO, and other styles
25

Harwood, Steven J., Linda K. Gibbons, Pamela J. Goldner, William B. Webster, and Robert G. Carroll. "Outpatient radioimmunotherapy with Bexxar." Cancer 94, S4 (February 12, 2002): 1358–62. http://dx.doi.org/10.1002/cncr.10306.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Constanzo, Julie, Lorenzo Galluzzi, and Jean-Pierre Pouget. "Immunostimulatory effects of radioimmunotherapy." Journal for ImmunoTherapy of Cancer 10, no. 2 (February 2022): e004403. http://dx.doi.org/10.1136/jitc-2021-004403.

Full text
Abstract:
Radiation therapy (RT) is known for its ability to kill cancer cells in an immunogenic manner. Recent preclinical data demonstrate that targeted alpha-particle therapy shares with RT the capacity to elicit immunostimulatory effects, standing out as a promising strategy to circumvent immune checkpoint inhibitor resistance in immunologically ‘cold’ tumors.
APA, Harvard, Vancouver, ISO, and other styles
27

Chanan-Khan, Asher, and Myron S. Czuczman. "Radioimmunotherapy in non-Hodgkin lymphoma." Current Opinion in Oncology 14, no. 5 (September 2002): 484–89. http://dx.doi.org/10.1097/00001622-200209000-00003.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Friedberg, Jonathan W. "Radioimmunotherapy for Non-Hodgkin’s Lymphoma." Clinical Cancer Research 10, no. 23 (December 1, 2004): 7789–91. http://dx.doi.org/10.1158/1078-0432.ccr-04-1706.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Wahl, Richard L. "Experimental radioimmunotherapy. A brief overview." Cancer 73, S3 (February 1, 1994): 989–92. http://dx.doi.org/10.1002/1097-0142(19940201)73:3+<989::aid-cncr2820731336>3.0.co;2-u.

Full text
APA, Harvard, Vancouver, ISO, and other styles
30

Kasten, Benjamin B., Soldano Ferrone, Kurt R. Zinn, and Donald J. Buchsbaum. "B7-H3-targeted Radioimmunotherapy of Human Cancer." Current Medicinal Chemistry 27, no. 24 (July 7, 2020): 4016–38. http://dx.doi.org/10.2174/0929867326666190228120908.

Full text
Abstract:
Background: Targeted Radioimmunotherapy (RIT) is an attractive approach to selectively localize therapeutic radionuclides to malignant cells within primary and metastatic tumors while sparing normal tissues from the effects of radiation. Many human malignancies express B7-H3 on the tumor cell surface, while expression on the majority of normal tissues is limited, presenting B7-H3 as a candidate target for RIT. This review provides an overview of the general principles of targeted RIT and discusses publications that have used radiolabeled B7-H3-targeted antibodies for RIT of cancer in preclinical or clinical studies. Methods: Databases including PubMed, Scopus, and Google Scholar were searched for publications through June 2018 using a combination of terms including “B7-H3”, “radioimmunotherapy”, “targeted”, “radiotherapy”, and “cancer”. After screening search results for relevancy, ten publications were included for discussion. Results: B7-H3-targeted RIT studies to date range from antibody development and assessment of novel Radioimmunoconjugates (RICs) in animal models of human cancer to phase II/III trials in humans. The majority of clinical studies have used B7-H3-targeted RICs for intra- compartment RIT of central nervous system malignancies. The results of these studies have indicated high tolerability and favorable efficacy outcomes, supporting further assessment of B7-H3-targeted RIT in larger trials. Preclinical B7-H3-targeted RIT studies have also shown encouraging therapeutic outcomes in a variety of solid malignancies. Conclusion: B7-H3-targeted RIT studies over the last 15 years have demonstrated feasibility for clinical development and support future assessment in a broader array of human malignancies. Future directions worthy of exploration include strategies that combine B7-H3- targeted RIT with chemotherapy or immunotherapy.
APA, Harvard, Vancouver, ISO, and other styles
31

Dadachova, Ekaterina. "Translating radioimmunotherapy from cancer to infectious diseases." Nuclear Medicine and Biology 108-109 (May 2022): S64—S65. http://dx.doi.org/10.1016/s0969-8051(22)00162-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Perkins, Alan, Melanie Hopper, Andrea Murray, Malcolm Frier, and Mike Bishop. "Antibody conjugate radioimmunotherapy of superficial bladder cancer." Brazilian Archives of Biology and Technology 45, spe (September 2002): 87–89. http://dx.doi.org/10.1590/s1516-89132002000500012.

Full text
Abstract:
The administration of antibody conjugates for cancer therapy is now proving to be of clinical value. We are currently undertaking a programme of clinical studies using the monoclonal antibody C595 (IgG3) which reacts with the MUC1 glycoprotein antigen that is aberrantly expressed in a high proportion of bladder tumours. Radioimmunoconjugates of the C595 antibody have been produced with high radiolabelling efficiency and immunoreactivity using Tc-99m and In-111 for diagnostic imaging, and disease staging and the cytotoxic radionuclides Cu-67 and Re-188 for therapy of superficial bladder cancer. A Phase I/II therapeutic trail involving the intravesical administration of antibody directly into the bladder has now begun.
APA, Harvard, Vancouver, ISO, and other styles
33

Stein, Rhona, Robert M. Sharkey, and David M. Goldenberg. "Haematological effects of radioimmunotherapy in cancer patients." British Journal of Haematology 80, no. 1 (January 1992): 69–76. http://dx.doi.org/10.1111/j.1365-2141.1992.tb06402.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Grana, C., M. Bartolomei, P. Rocca, L. Bodei, M. Gatti, M. Caracciolo, N. Colombo, and G. Paganelli. "3-Step Radioimmunotherapy in Advanced Ovarian Cancer." Tumori Journal 88, no. 4 (July 2002): S9. http://dx.doi.org/10.1177/030089160208800450.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Zhao, Xiao-Yan, Doug Schneider, Sandra L. Biroc, Renate Parry, Bruno Alicke, Pamela Toy, Jian-Ai Xuan, et al. "Targeting Tomoregulin for Radioimmunotherapy of Prostate Cancer." Cancer Research 65, no. 7 (April 1, 2005): 2846–53. http://dx.doi.org/10.1158/0008-5472.can-04-4019.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Attard, A. R., M. J. Chappell, and A. R. Bradwell. "Model based calculation for effective cancer radioimmunotherapy." British Journal of Radiology 68, no. 810 (June 1995): 636–45. http://dx.doi.org/10.1259/0007-1285-68-810-636.

Full text
APA, Harvard, Vancouver, ISO, and other styles
37

Jurcic, Joseph G., and David A. Scheinberg. "Recent developments in the radioimmunotherapy of cancer." Current Opinion in Immunology 6, no. 5 (January 1994): 715–21. http://dx.doi.org/10.1016/0952-7915(94)90074-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
38

Kramer, Kim, Nai-Kong V. Cheung, John L. Humm, Ester Dantis, Ron Finn, Samuel J. Yeh, Nuno L. Antunes, Ira J. Dunkel, Marc Souwedaine, and Steven M. Larson. "Targeted radioimmunotherapy for leptomeningeal cancer using131I-3F8." Medical and Pediatric Oncology 35, no. 6 (2000): 716–18. http://dx.doi.org/10.1002/1096-911x(20001201)35:6<716::aid-mpo51>3.0.co;2-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
39

Maraveyas, A., and A. A. Epenetos. "An overview of radioimmunotherapy." Cancer Immunology Immunotherapy 34, no. 2 (March 1991): 71–73. http://dx.doi.org/10.1007/bf01741338.

Full text
APA, Harvard, Vancouver, ISO, and other styles
40

Bihl, H., and Marie-Luise Sautter-Bihl. "Radioimmunotherapy with Monoclonal Antibodies." Nuklearmedizin 33, no. 04 (1994): 167–73. http://dx.doi.org/10.1055/s-0038-1629812.

Full text
Abstract:
SummaryRadioimmunotherapy (RIT) with labeled tumor-associated monoclonal antibodies (MAbs) is a promising concept in oncology, which essentially consists of biological targeting of ionising radiation to tumors. Some encouraging clinical results have been achieved with RIT. However, there are severe problems associated with both understanding the mechanisms and predicting the effectiveness of RIT. This paper reviews the results of some major clinical trials, especially in malignant lymphomas and in some solid tumors. Furthermore, problems with RIT are described such as the significance of dose inhomogeneity and dose-rate effects, the appropriate dose calculation method, the toxicity of RIT and the development of HAMAs. It is suggested that newer technologies including chimeric antibodies, multiple-step targeting protocols, bone marrow transplantation, parallel application of external radiation, heat or bioreductive drugs will enable RIT to make an essential contribution to strategies for combating cancer.
APA, Harvard, Vancouver, ISO, and other styles
41

Kinuya, Seigo, Xiao-Feng Li, Kunihiko Yokoyama, Hirofumi Mori, Kazuhiro Shiba, Naoto Watanabe, Noriyuki Shuke, Hisashi Bunko, Takatoshi Michigishi, and Norihisa Tonami. "Intraperitoneal radioimmunotherapy in treating peritoneal carcinomatosis of colon cancer in mice compared with systemic radioimmunotherapy." Cancer Science 94, no. 7 (July 2003): 650–54. http://dx.doi.org/10.1111/j.1349-7006.2003.tb01498.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Sheng, Hailong, Yan Huang, Yazhi Xiao, Zhenru Zhu, Mengying Shen, Peitao Zhou, Zeqin Guo, et al. "ATR inhibitor AZD6738 enhances the antitumor activity of radiotherapy and immune checkpoint inhibitors by potentiating the tumor immune microenvironment in hepatocellular carcinoma." Journal for ImmunoTherapy of Cancer 8, no. 1 (May 2020): e000340. http://dx.doi.org/10.1136/jitc-2019-000340.

Full text
Abstract:
BackgroundRadioimmunotherapy has a promising antitumor effect in hepatocellular carcinoma (HCC), depending on the regulatory effect of radiotherapy on tumor immune microenvironment. Ionizing radiation (IR)-induced DNA damage repair (DDR) pathway activation leads to the inhibition of immune microenvironment, thus impairing the antitumor effect of radioimmunotherapy. However, it is unclear whether inhibition of the DDR pathway can enhance the effect of radioimmunotherapy. In this study, we aim to explore the role of DDR inhibitor AZD6738 on the combination of radiotherapy and immune checkpoint inhibitors (ICIs) in HCC.MethodsC57BL/6 mouse subcutaneous tumor model was used to evaluate the ability of different treatment regimens in tumor growth control and tumor recurrence inhibition. Effects of each treatment regimen on the alterations of immunophenotypes including the quantification, activation, proliferating ability, exhaustion marker expression, and memory status were assessed by flow cytometry.ResultsAZD6738 further increased radiotherapy-stimulated CD8+T cell infiltration and activation and reverted the immunosuppressive effect of radiation on the number of Tregs in mice xenografts. Moreover, compared with radioimmunotherapy (radiotherapy plus anti-PD-L1 (Programmed death ligand 1)), the addition of AZD6738 boosted the infiltration, increased cell proliferation, enhanced interferon (IFN)-γ production ability of TIL (tumor-infiltrating lymphocyte) CD8+T cells, and caused a decreasing trend in the number of TIL Tregs and exhausted T cells in mice xenografts. Thus, the tumor immune microenvironment was significantly improved. Meanwhile, triple therapy (AZD6738 plus radiotherapy plus anti-PD-L1) also induced a better immunophenotype than radioimmunotherapy in mice spleens. As a consequence, triple therapy displayed greater benefit in antitumor efficacy and mice survival than radioimmunotherapy. Mechanism study revealed that the synergistic antitumor effect of AZD6738 with radioimmunotherapy relied on the activation of cyclic GMP–AMP synthase /stimulator of interferon genes (cGAS/STING) signaling pathway. Furthermore, triple therapy led to stronger immunologic memory and lasting antitumor immunity than radioimmunotherapy, thus preventing tumor recurrence in mouse models.ConclusionsOur findings indicate that AZD6738 might be a potential synergistic treatment for radioimmunotherapy to control the proliferation of HCC cells, prolong survival, and prevent tumor recurrence in patients with HCC by improving the immune microenvironment.
APA, Harvard, Vancouver, ISO, and other styles
43

Green, Damian J., John M. Pagel, Anastasia Pantelias, Nathan Hedin, Yukang Lin, D. Scott Wilbur, Ajay Gopal, Donald K. Hamlin, and Oliver W. Press. "Pretargeted Radioimmunotherapy for B-Cell Lymphomas." Clinical Cancer Research 13, no. 18 (September 15, 2007): 5598s—5603s. http://dx.doi.org/10.1158/1078-0432.ccr-07-1223.

Full text
APA, Harvard, Vancouver, ISO, and other styles
44

Riggs, Simon J., Alan J. Green, Richard H. J. Begent, and Kenneth D. Bagshawe. "Quantitation in131 I-radioimmunotherapy using spect." International Journal of Cancer 41, S2 (1988): 95–98. http://dx.doi.org/10.1002/ijc.2910410722.

Full text
APA, Harvard, Vancouver, ISO, and other styles
45

Larson, Steven M., Chaitanya R. Divgi, Andrew Scott, George Sgouros, Martin C. Graham, Lale Kostakoglu, David Scheinberg, Nai-Kong V. Cheung, Jeffrey Schlom, and Ronald D. Finn. "Current status of radioimmunotherapy." Nuclear Medicine and Biology 21, no. 5 (July 1994): 785–92. http://dx.doi.org/10.1016/0969-8051(94)90050-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Witzig, Thomas E., Leo I. Gordon, Fernando Cabanillas, Myron S. Czuczman, Christos Emmanouilides, Robin Joyce, Brad L. Pohlman, et al. "Randomized Controlled Trial of Yttrium-90–Labeled Ibritumomab Tiuxetan Radioimmunotherapy Versus Rituximab Immunotherapy for Patients With Relapsed or Refractory Low-Grade, Follicular, or Transformed B-Cell Non-Hodgkin’s Lymphoma." Journal of Clinical Oncology 20, no. 10 (May 15, 2002): 2453–63. http://dx.doi.org/10.1200/jco.2002.11.076.

Full text
Abstract:
PURPOSE: Radioimmunotherapy combines biologic and radiolytic mechanisms to target and destroy tumor cells, thus offering a needed therapeutic alternative for refractory non-Hodgkin’s lymphoma (NHL) patients. This phase III randomized study compares the novel radioimmunotherapy yttrium-90 (90Y) ibritumomab tiuxetan with a control immunotherapy, rituximab, in 143 patients with relapsed or refractory low-grade, follicular, or transformed CD20+ transformed NHL. PATIENTS AND METHODS: Patients received either a single intravenous (IV) dose of 90Y ibritumomab tiuxetan 0.4 mCi/kg (n = 73) or rituximab 375 mg/m2 IV weekly for four doses (n = 70). The radioimmunotherapy group was pretreated with two rituximab doses (250 mg/m2) to improve biodistribution and one dose of indium-111 ibritumomab tiuxetan for imaging and dosimetry. The primary end point, overall response rate (ORR), was assessed by an independent, blinded, lymphoma expert panel. RESULTS: ORR was 80% for the 90Y ibritumomab tiuxetan group versus 56% for the rituximab group (P = .002). Complete response (CR) rates were 30% and 16% in the 90Y ibritumomab tiuxetan and rituximab groups, respectively (P = .04). An additional 4% achieved an unconfirmed CR in each group. Kaplan-Meier estimated median duration of response was 14.2 months in the 90Y ibritumomab tiuxetan group versus 12.1 months in the control group (P = .6), and time to progression was 11.2 versus 10.1 months (P = .173) in all patients. Durable responses of ≥ 6 months were 64% versus 47% (P = .030). Reversible myelosuppression was the primary toxicity noted with 90Y ibritumomab tiuxetan. CONCLUSION: Radioimmunotherapy with 90Y ibritumomab tiuxetan is well tolerated and produces statistically and clinically significant higher ORR and CR compared with rituximab alone.
APA, Harvard, Vancouver, ISO, and other styles
47

Bouchelouche, Kirsten, Scott T. Tagawa, Stanley J. Goldsmith, Baris Turkbey, Jacek Capala, and Peter Choyke. "PET/CT Imaging and Radioimmunotherapy of Prostate Cancer." Seminars in Nuclear Medicine 41, no. 1 (January 2011): 29–44. http://dx.doi.org/10.1053/j.semnuclmed.2010.08.005.

Full text
APA, Harvard, Vancouver, ISO, and other styles
48

Scott, Andrew M. "Radioimmunotherapy of Prostate Cancer: Does Tumor Size Matter?" Journal of Clinical Oncology 23, no. 21 (July 20, 2005): 4567–69. http://dx.doi.org/10.1200/jco.2005.01.903.

Full text
APA, Harvard, Vancouver, ISO, and other styles
49

Börjesson, Pontus K. E., Ernst J. Postema, Remco de Bree, Jan C. Roos, C. René Leemans, Kalevi J. A. Kairemo, and Guus A. M. S. van Dongen. "Radioimmunodetection and radioimmunotherapy of head and neck cancer." Oral Oncology 40, no. 8 (September 2004): 761–72. http://dx.doi.org/10.1016/j.oraloncology.2003.11.009.

Full text
APA, Harvard, Vancouver, ISO, and other styles
50

Kraeber-Bodéré, Françoise, Pierre-Yves Salaun, Catherine Ansquer, Delphine Drui, Eric Mirallié, Alain Faivre-Chauvet, Jacques Barbet, David M. Goldenberg, and Jean-François Chatal. "Pretargeted radioimmunotherapy (pRAIT) in medullary thyroid cancer (MTC)." Tumor Biology 33, no. 3 (March 7, 2012): 601–6. http://dx.doi.org/10.1007/s13277-012-0359-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography