1
|
Winter RC, Amghar M, Wacker AS, Bakos G, Taş H, Roscher M, Kelly JM, Benešová-Schäfer M. Future Treatment Strategies for Cancer Patients Combining Targeted Alpha Therapy with Pillars of Cancer Treatment: External Beam Radiation Therapy, Checkpoint Inhibition Immunotherapy, Cytostatic Chemotherapy, and Brachytherapy. Pharmaceuticals (Basel) 2024; 17:1031. [PMID: 39204136 PMCID: PMC11359268 DOI: 10.3390/ph17081031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Cancer is one of the most complex and challenging human diseases, with rising incidences and cancer-related deaths despite improved diagnosis and personalized treatment options. Targeted alpha therapy (TαT) offers an exciting strategy emerging for cancer treatment which has proven effective even in patients with advanced metastatic disease that has become resistant to other treatments. Yet, in many cases, more sophisticated strategies are needed to stall disease progression and overcome resistance to TαT. The combination of two or more therapies which have historically been used as stand-alone treatments is an approach that has been pursued in recent years. This review aims to provide an overview on TαT and the four main pillars of therapeutic strategies in cancer management, namely external beam radiation therapy (EBRT), immunotherapy with checkpoint inhibitors (ICI), cytostatic chemotherapy (CCT), and brachytherapy (BT), and to discuss their potential use in combination with TαT. A brief description of each therapy is followed by a review of known biological aspects and state-of-the-art treatment practices. The emphasis, however, is given to the motivation for combination with TαT as well as the pre-clinical and clinical studies conducted to date.
Collapse
Affiliation(s)
- Ruth Christine Winter
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mariam Amghar
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Anja S. Wacker
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Gábor Bakos
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Harun Taş
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mareike Roscher
- Service Unit for Radiopharmaceuticals and Preclinical Studies, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany;
| | - James M. Kelly
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Martina Benešová-Schäfer
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| |
Collapse
|
2
|
Roy A, Gabani P, Davis EJ, Oppelt P, Merfeld E, Keedy VL, Zoberi I, Chrisinger JSA, Michalski JM, Van Tine B, Spraker MB. Concurrent paclitaxel and radiation therapy for the treatment of cutaneous angiosarcoma. Clin Transl Radiat Oncol 2021; 27:114-120. [PMID: 33604458 PMCID: PMC7876514 DOI: 10.1016/j.ctro.2021.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/25/2022] Open
Abstract
Cutaneous angiosarcoma has poor outcomes with no standardized treatment regimen. Paclitaxel-based chemoRT (CRT) was compared to other therapies at two US institutions. Similar oncologic outcomes and improved survival with paclitaxel CRT. Paclitaxel CRT + surgery provided best oncologic outcomes and survival. Paclitaxel CRT + surgery regimen now being studied in a prospective phase II trial.
Introduction We compared clinical outcomes in patients with cutaneous angiosarcoma receiving concurrent paclitaxel-based chemoradiotherapy (CRT) vs. other modalities (Non-CRT). Materials and methods Patients with non-metastatic cutaneous angiosarcoma diagnosed from 1998 to 2018 at two institutions were identified. In the CRT cohort, paclitaxel 80 mg/m2 weekly was given for up to 12 weeks and patients received radiotherapy (RT) during the final 6 weeks of chemotherapy. The RT dose was 50–50.4 Gy delivered in 1.8–2 Gy per fraction with an optional post-operative boost of 10–16 Gy. Kaplan-Meier and log-rank statistics were used to compare the outcomes between the two groups. P < 0.05 was considered statistically significant. Results Fifty-seven patients were included: 22 CRT and 35 Non-CRT. The CRT cohort had more patients > 60 years (100% vs. 60%, p < 0.001) and tumors >5 cm (68.2% vs 54.3%, p = 0.023). The median follow-up was 25.8 (1.5–155.2) months. There was no significant difference in 2-year local control (LC), distant control (DC), or progression-free survival (PFS) between the two groups. The 2-year overall survival (OS) was significantly higher for the CRT cohort (94.1% vs. 71.6%, p = 0.033). Amongst the subset of patients in the CRT cohort who received trimodality therapy, the 2-year LC, DC, PFS, and OS was 68.6%, 100%, 68.6%, and 100%, respectively. Conclusion The use of concurrent paclitaxel CRT demonstrates promising outcomes. Given these results, we are currently evaluating the safety and efficacy of this regimen in prospective, phase 2 trial (NCT 03921008).
Collapse
Affiliation(s)
- Amit Roy
- Department of Radiation Oncology, Washington University School of Medicine, 4921 Parkview Place, St. Louis, MO 63110, United States
| | - Prashant Gabani
- Baylor Scott and White Health Cancer Center, 300 University Blvd., Bldg. A, Round Rock, TX 78665, United States
| | - Elizabeth J Davis
- Department of Hematology and Oncology, Vanderbilt University School of Medicine, 1161 21st Ave S # D3300, Nashville, TN 37232, United States
| | - Peter Oppelt
- Division of Medical Oncology, Washington University School of Medicine, 4921 Parkview Place, St. Louis, MO 63110, United States
| | - Emily Merfeld
- Department of Human Oncology, University of Wisconsin School of Medicine, 600 Highland Ave, Madison, WI 53792, United States
| | - Vicky L Keedy
- Department of Hematology and Oncology, Vanderbilt University School of Medicine, 1161 21st Ave S # D3300, Nashville, TN 37232, United States
| | - Imran Zoberi
- Department of Radiation Oncology, Washington University School of Medicine, 4921 Parkview Place, St. Louis, MO 63110, United States
| | - John S A Chrisinger
- Department of Pathology and Immunology, Washington University School of Medicine, 4921 Parkview Place, St. Louis, MO 63110, United States
| | - Jeff M Michalski
- Department of Radiation Oncology, Washington University School of Medicine, 4921 Parkview Place, St. Louis, MO 63110, United States
| | - Brian Van Tine
- Division of Medical Oncology, Washington University School of Medicine, 4921 Parkview Place, St. Louis, MO 63110, United States
| | - Matthew B Spraker
- Department of Radiation Oncology, Washington University School of Medicine, 4921 Parkview Place, St. Louis, MO 63110, United States
| |
Collapse
|
3
|
Phase I trial of docetaxel plus lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 ( 177Lu-J591) for metastatic castration-resistant prostate cancer. Urol Oncol 2020; 38:848.e9-848.e16. [PMID: 32600929 DOI: 10.1016/j.urolonc.2020.05.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND Docetaxel remains a standard of care for metatsatic castration resistant porstate cancer (mCRPC) and has radiosensitizing properties. The dose limiting toxicity (DLT) of radioimmunotherapy is myelosuppression; dose fractionation of 177Lu-J591 allows similar administered doses with less toxicity. This study (NCT00916123) was designed to determine the safety, DLT, and maximum tolerated dose of fractionated 177Lu-J591 administered concurrently with standard docetaxel. METHODS Men with progressive mCRPC received docetaxel 75 mg/m2 every 3 weeks with escalating 2 fractionated doses of 177Lu-J591 (1.48 GBq/m2 up to max of 2.96 GBq/m2) with cycle 3. Cycle 4 of docetaxel was planned 6 weeks after cycle 3 to allow for recovery from 177Lu-J591-associated hematologic toxicity. DLT was defined as delay in docetaxel >3 weeks, prolonged myelosuppression or need for >2 platelet transfusions, febrile neutropenia, or grade ≥3 nonhematological toxicity following 177Lu-J591. PSA was assessed prior to each cycle and serial computed tomography (CT) and bone scan were performed. RESULTS Fifteen men with progressive mCRPC received dose-escalated targeted radionuclide therapy in 4 cohorts up to the highest planned dose (2.96 GBq/m2). No DLT was seen at any dose level. Grade 4 neutropenia without fever occurred in 8 (53.5%) and thromboytopenia in 2 (13.3%), with 2 receiving prophylactic platelet transfusion. No grade ≥3 nonhematological toxicity was observed. 11 (73.3%) had >50% PSA decline, with 78.6% having favorable circulating tumor cell counts after 177Lu-J591. All patients had targeting of known sites of disease by planar 177Lu-J591 imaging. CONCLUSION The combination of 177Lu-J591 delivered as a single fractionated cycle with docetaxel/prednisone is feasible in patients with mCRPC. Without preselection for prostate-specific membrane antigen, accurate targeting of known sites of disease and a strong preliminary efficacy signal was observed.
Collapse
|
4
|
Xie J, Gong L, Zhu S, Yong Y, Gu Z, Zhao Y. Emerging Strategies of Nanomaterial-Mediated Tumor Radiosensitization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1802244. [PMID: 30156333 DOI: 10.1002/adma.201802244] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/08/2018] [Indexed: 05/23/2023]
Abstract
Nano-radiosensitization has been a hot concept for the past ten years, and the nanomaterial-mediated tumor radiosensitization method is mainly focused on increasing intracellular radiation deposition by high atomic number (high Z) nanomaterials, particularly gold (Au)-mediated radiation enhancement. Recently, various new nanomaterial-mediated radiosensitive approaches have been successively reported, such as catalyzing reactive oxygen species (ROS) generation, consuming intracellular reduced glutathione (GSH), overcoming tumor hypoxia, and various synergistic radiotherapy ways. These strategies may open a new avenue for enhancing the radiotherapeutic effect and avoiding its side effects. Nevertheless, reviews systematically summarizing these newly emerging methods and their radiosensitive mechanisms are still rare. Therefore, the general strategies of nanomaterial-mediated tumor radiosensitization are comprehensively summarized, particularly aiming at introducing the emerging radiosensitive methods. The strategies are divided into three general parts. First, methods on account of the intrinsic radiosensitive properties of nanoradiosensitizers for radiosensitization are highlighted. Then, newly developed synergistic strategies based on multifunctional nanomaterials for enhancing radiotherapy efficacy are emphasized. Third, nanomaterial-mediated radioprotection approaches for increasing the radiotherapeutic ratio are discussed. Importantly, the clinical translation of nanomaterial-mediated tumor radiosensitization is also covered. Finally, further challenges and outlooks in this field are discussed.
Collapse
Affiliation(s)
- Jiani Xie
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Linji Gong
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Zhu
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuan Yong
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhanjun Gu
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuliang Zhao
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
5
|
Arita T, Asai J, Taura M, Wada M, Takenaka H, Miyashita A, Katoh N. Squamous cell carcinoma of neck metastasized to the lung and cervical lymph nodes, successfully treated with systemic docetaxel and local radiation therapy to the neck in a renal transplant patient. J Dermatol 2017; 44:e346-e347. [PMID: 28862322 DOI: 10.1111/1346-8138.14018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Takahiro Arita
- Department of Dermatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Jun Asai
- Department of Dermatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Maiko Taura
- Department of Dermatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Makoto Wada
- Department of Dermatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Hideya Takenaka
- Department of Dermatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Aya Miyashita
- Department of Dermatology, Omihachiman Community Medical Center, Shiga, Japan
| | - Norito Katoh
- Department of Dermatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
6
|
Response Detection of Castrate-Resistant Prostate Cancer to Clinically Utilised and Novel Treatments by Monitoring Phospholipid Metabolism. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4793465. [PMID: 28717648 PMCID: PMC5498927 DOI: 10.1155/2017/4793465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/30/2017] [Indexed: 12/27/2022]
Abstract
Androgen receptor (AR) activation is the primary driving factor in prostate cancer which is initially responsive to castration but then becomes resistant (castration-resistant prostate cancer (CRPC)). CRPC cells still retain the functioning AR which can be targeted by other therapies. A recent promising development is the use of inhibitors (Epi-1) of protein-protein interaction to inhibit AR-activated signalling. Translating novel therapies into the clinic requires sensitive early response indicators. Here potential response markers are explored. Growth inhibition of prostate cancer cells with flutamide, paclitaxel, and Epi-1 was measured using the MTT assay. To simulate choline-PET scans, pulse-chase experiments were carried out with [3H-methyl]choline and proportion of phosphorylated activity was determined after treatment with growth inhibitory concentrations of each drug. Extracts from treated cells were also subject to 31P-NMR spectroscopy. Cells treated with flutamide demonstrated decreased [3H-methyl]choline phosphorylation, whilst the proportion of phosphorylated [3H-methyl]choline that was present in the lipid fraction was increased in Epi-1-treated cells. Phospholipid breakdown products, glycerophosphorylcholine and glycerophosphoethanolamine levels, were shown by 31P-NMR spectroscopy to be decreased to undetectable levels in cells treated with Epi-1. LNCaP cells responding to treatment with novel protein-protein interaction inhibitors suggest that 31P-NMR spectroscopy may be useful in detecting response to this promising therapy.
Collapse
|
7
|
Pandya HJ, Dhingra K, Prabhakar D, Chandrasekar V, Natarajan SK, Vasan AS, Kulkarni A, Shafiee H. A microfluidic platform for drug screening in a 3D cancer microenvironment. Biosens Bioelectron 2017; 94:632-642. [PMID: 28371753 DOI: 10.1016/j.bios.2017.03.054] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/14/2017] [Accepted: 03/24/2017] [Indexed: 11/16/2022]
Abstract
Development of resistance to chemotherapy treatments is a major challenge in the battle against cancer. Although a vast repertoire of chemotherapeutics is currently available for treating cancer, a technique for rapidly identifying the right drug based on the chemo-resistivity of the cancer cells is not available and it currently takes weeks to months to evaluate the response of cancer patients to a drug. A sensitive, low-cost diagnostic assay capable of rapidly evaluating the effect of a series of drugs on cancer cells can significantly change the paradigm in cancer treatment management. Integration of microfluidics and electrical sensing modality in a 3D tumour microenvironment may provide a powerful platform to tackle this issue. Here, we report a 3D microfluidic platform that could be potentially used for a real-time deterministic analysis of the success rate of a chemotherapeutic drug in less than 12h. The platform (66mm×50mm; L×W) is integrated with the microsensors (interdigitated gold electrodes with width and spacing 10µm) that can measure the change in the electrical response of cancer cells seeded in a 3D extra cellular matrix when a chemotherapeutic drug is flown next to the matrix. B16-F10 mouse melanoma, 4T1 mouse breast cancer, and DU 145 human prostate cancer cells were used as clinical models. The change in impedance magnitude on flowing chemotherapeutics drugs measured at 12h for drug-susceptible and drug tolerant breast cancer cells compared to control were 50,552±144 Ω and 28,786±233 Ω, respectively, while that of drug-susceptible melanoma cells were 40,197±222 Ω and 4069±79 Ω, respectively. In case of prostate cancer the impedance change between susceptible and resistant cells were 8971±1515 Ω and 3281±429 Ω, respectively, which demonstrated that the microfluidic platform was capable of delineating drug susceptible cells, drug tolerant, and drug resistant cells in less than 12h.
Collapse
Affiliation(s)
- Hardik J Pandya
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA
| | - Karan Dhingra
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA
| | - Devbalaji Prabhakar
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA
| | - Vineethkrishna Chandrasekar
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA
| | - Siva Kumar Natarajan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA
| | - Anish S Vasan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA
| | - Ashish Kulkarni
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA.
| | - Hadi Shafiee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital - Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| |
Collapse
|
8
|
Fujisawa Y, Yoshino K, Kadono T, Miyagawa T, Nakamura Y, Fujimoto M. Chemoradiotherapy with taxane is superior to conventional surgery and radiotherapy in the management of cutaneous angiosarcoma: a multicentre, retrospective study. Br J Dermatol 2014; 171:1493-500. [DOI: 10.1111/bjd.13110] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Y. Fujisawa
- Department of Dermatology; University of Tsukuba; 1-1-1 Tennodai Tsukuba Ibaraki 305-8575 Japan
| | - K. Yoshino
- Department of Dermatology; Tokyo Metropolitan Cancer and Infectious disease Center Komagome Hospital; Tokyo Japan
| | - T. Kadono
- Department of Dermatology; University of Tokyo; Tokyo Japan
| | - T. Miyagawa
- Department of Dermatology; University of Tokyo; Tokyo Japan
| | - Y. Nakamura
- Department of Dermatology; University of Tsukuba; 1-1-1 Tennodai Tsukuba Ibaraki 305-8575 Japan
| | - M. Fujimoto
- Department of Dermatology; University of Tsukuba; 1-1-1 Tennodai Tsukuba Ibaraki 305-8575 Japan
| |
Collapse
|
9
|
|
10
|
Yu SD, Liu FY, Wang QR. Notch inhibitor: a promising carcinoma radiosensitizer. Asian Pac J Cancer Prev 2013; 13:5345-51. [PMID: 23317182 DOI: 10.7314/apjcp.2012.13.11.5345] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Radiotherapy is an important part of modern cancer management for many malignancies, and enhancing the radiosensitivity of tumor cells is critical for effective cancer therapies. The Notch signaling pathway plays a key role in regulation of numerous fundamental cellular processes. Further, there is accumulating evidence that dysregulated Notch activity is involved in the genesis of many human cancers. As such, Notch inhibitors are attractive therapeutic agents, although as for other anticancer agents, they exhibit significant and potential side effects. Thus, Notch inhibitors may be best used in combination with other agents or therapy. Herein, we describe evidence supporting the use of Notch inhibitors as novel and potent radiosensitizers in cancer therapy.
Collapse
Affiliation(s)
- Shu-Dong Yu
- Department of Otolaryngology, Qianfoshan Hospital Affiliated to Shandong University, Shandong, China.
| | | | | |
Collapse
|
11
|
Shimizu N, Nakajima NI, Tsunematsu T, Ogawa I, Kawai H, Hirayama R, Fujimori A, Yamada A, Okayasu R, Ishimaru N, Takata T, Kudo Y. Selective enhancing effect of early mitotic inhibitor 1 (Emi1) depletion on the sensitivity of doxorubicin or X-ray treatment in human cancer cells. J Biol Chem 2013; 288:17238-52. [PMID: 23645673 DOI: 10.1074/jbc.m112.446351] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Chemotherapy and radiation in addition to surgery has proven useful in a number of different cancer types, but the effectiveness in normal tissue cannot be avoided in these therapies. To improve the effectiveness of these therapies selectively in cancer tissue is important for avoiding side effects. Early mitotic inhibitor 1 (Emi1) is known to have the function to inhibit anaphase-promoting complex/cyclosome ubiquitin ligase complex, which ubiquitylates the cell cycle-related proteins. It recently has been shown that Emi1 knockdown prevents transition from S to G2 phase by down-regulating geminin via anaphase-promoting complex/cyclosome activation. At present, anticancer drugs for targeting DNA synthesis to interfere with rapidly dividing cells commonly are used. As Emi1 depletion interferes with completion of DNA synthesis in cancer cells, we thought that Emi1 knockdown might enhance the sensitivity for anticancer agents. Here, we confirmed that Emi1 siRNA induced polyploidy for preventing transition from S to G2 phase in several cancer cell lines. Then, we treated Emi1 depleted cells with doxorubicin. Interestingly, increased apoptotic cells were observed after doxorubicin treatment in Emi1 siRNA-treated cancer cells. In addition, Emi1 depletion enhanced the sensitivity of x-ray irradiation in cancer cells. Importantly, synergistic effect of Emi1 knockdown in these combination therapies was not observed in normal cells. These results suggest that Emi1 siRNA can be a useful tool for enhancing of sensitivity of cancer cells to anticancer reagents and radiation.
Collapse
Affiliation(s)
- Natsumi Shimizu
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hoffmann C, Bachran C, Stanke J, Elezkurtaj S, Kaufmann AM, Fuchs H, Loddenkemper C, Schneider A, Cichon G. Creation and characterization of a xenograft model for human cervical cancer. Gynecol Oncol 2010; 118:76-80. [DOI: 10.1016/j.ygyno.2010.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 03/26/2010] [Accepted: 03/29/2010] [Indexed: 11/25/2022]
|
13
|
Kelly MP, Lee ST, Lee FT, Smyth FE, Davis ID, Brechbiel MW, Scott AM. Therapeutic efficacy of 177Lu-CHX-A''-DTPA-hu3S193 radioimmunotherapy in prostate cancer is enhanced by EGFR inhibition or docetaxel chemotherapy. Prostate 2009; 69:92-104. [PMID: 18942092 PMCID: PMC2597150 DOI: 10.1002/pros.20856] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND This study investigated the biodistribution and therapeutic efficacy of Lutetium-177 (177Lu) radiolabeled anti-Lewis Y monoclonal antibody hu3S193 radioimmunotherapy (RIT) in mice bearing prostate cancer xenografts. The ability of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor AG1478 and docetaxel chemotherapy to enhance the efficacy of RIT was also assessed in vivo. METHODS The in vitro cytotoxicity of 177Lu labeled hu3S193 on Le(y) positive DU145 prostate cancer cells was assessed using proliferation assays, with induction of apoptosis measured by ELISA. The in vivo biodistribution and tumor localization of 177Lu-hu3S193 was assessed in mice bearing established DU145 tumor xenografts. The efficacy and maximum tolerated dose of 177Lu-hu3S193 RIT in vivo was determined by a dose escalation study. EGFR inhibitor AG1478 or docetaxel chemotherapy was administered at sub-therapeutic doses in conjunction with RIT in vivo. RESULTS 177Lu-hu3S193 mediated significant induction of cytotoxicity and apoptosis in vitro. In vivo analysis of 177Lu-hu3S193 biodistribution demonstrated specific targeting of DU145 prostate cancer xenografts, with maximal tumor uptake of 33.2 +/- 3.9%ID/g observed at 120 hr post-injection. In RIT studies, 177Lu-hu3S193 caused specific and dose-dependent inhibition of prostate cancer tumor growth. A maximum tolerated dose of 350 microCi was determined for 177Lu-hu3S193. Combination of 177Lu-hu3S193 RIT with EGFR inhibitor AG1478 or docetaxel chemotherapy both significantly improved efficacy. CONCLUSIONS 177Lu-hu3S193 RIT is effective as a single agent in the treatment of Le(y) positive prostate cancer models. The enhancement of RIT by AG1478 or docetaxel indicates the promise of combined modality strategies.
Collapse
Affiliation(s)
- Marcus P Kelly
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Melbourne Centre for ClinicalSciences, Austin Hospital, Heidelberg, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|