1
|
Minnar CM, Lui G, Gulley JL, Schlom J, Gameiro SR. Preclinical and clinical studies of a tumor targeting IL-12 immunocytokine. Front Oncol 2024; 13:1321318. [PMID: 38260854 PMCID: PMC10802843 DOI: 10.3389/fonc.2023.1321318] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
The clinical success of immune checkpoint inhibitors (ICIs) has demonstrated the promise and challenges of cancer immunotherapy. There is an unmet need to develop novel cancer therapies that can provide clinical benefit for most patients with solid malignancies, which harbor innate or acquired resistance to ICIs. Interleukin-12 (IL-12) is a promising cytokine for cancer therapy given its direct stimulatory effects on innate and adaptive immunity. However, unfavorable pharmacokinetics and a narrow therapeutic index render recombinant IL-12 (rIL-12) less attractive as a cancer therapy. NHS-IL12 is a fusion protein of IL-12 and NHS76 (human IgG1) antibody engineered to target single and double stranded DNA present in necrotic areas solid tumors. In preclinical tumor models, NHS-IL12 elicited significant Th1 immune activation and tumor suppressive effects, primarily mediated by NK and CD8+ T lymphocytes, with engagement of myeloid immunity. NHS-IL12 is currently being evaluated clinically in combination with various therapeutic modalities, including chemotherapy, radiation therapy, immune checkpoint inhibition, vaccines, and epigenetic modulation. Here we review the preclinical and clinical studies involving NHS-IL12 for the treatment of solid malignancies.
Collapse
|
2
|
Zhu T, Hsu JC, Guo J, Chen W, Cai W, Wang K. Radionuclide-based theranostics - a promising strategy for lung cancer. Eur J Nucl Med Mol Imaging 2023; 50:2353-2374. [PMID: 36929181 PMCID: PMC10272099 DOI: 10.1007/s00259-023-06174-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/25/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE This review aims to provide a comprehensive overview of the latest literature on personalized lung cancer management using different ligands and radionuclide-based tumor-targeting agents. BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide. Due to the heterogeneity of lung cancer, advances in precision medicine may enhance the disease management landscape. More recently, theranostics using the same molecule labeled with two different radionuclides for imaging and treatment has emerged as a promising strategy for systemic cancer management. In radionuclide-based theranostics, the target, ligand, and radionuclide should all be carefully considered to achieve an accurate diagnosis and optimal therapeutic effects for lung cancer. METHODS We summarize the latest radiotracers and radioligand therapeutic agents used in diagnosing and treating lung cancer. In addition, we discuss the potential clinical applications and limitations associated with target-dependent radiotracers as well as therapeutic radionuclides. Finally, we provide our views on the perspectives for future development in this field. CONCLUSIONS Radionuclide-based theranostics show great potential in tailored medical care. We expect that this review can provide an understanding of the latest advances in radionuclide therapy for lung cancer and promote the application of radioligand theranostics in personalized medicine.
Collapse
Affiliation(s)
- Tianxing Zhu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
- Lingang Laboratory, Shanghai, 200031, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jingpei Guo
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Weiyu Chen
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Kai Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
3
|
Parakh S, Lee ST, Gan HK, Scott AM. Radiolabeled Antibodies for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:1454. [PMID: 35326605 PMCID: PMC8946248 DOI: 10.3390/cancers14061454] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/14/2022] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
Radioimmunoconjugates consist of a monoclonal antibody (mAb) linked to a radionuclide. Radioimmunoconjugates as theranostics tools have been in development with success, particularly in hematological malignancies, leading to approval by the US Food and Drug Administration (FDA) for the treatment of non-Hodgkin's lymphoma. Radioimmunotherapy (RIT) allows for reduced toxicity compared to conventional radiation therapy and enhances the efficacy of mAbs. In addition, using radiolabeled mAbs with imaging methods provides critical information on the pharmacokinetics and pharmacodynamics of therapeutic agents with direct relevance to the optimization of the dose and dosing schedule, real-time antigen quantitation, antigen heterogeneity, and dynamic antigen changes. All of these parameters are critical in predicting treatment responses and identifying patients who are most likely to benefit from treatment. Historically, RITs have been less effective in solid tumors; however, several strategies are being investigated to improve their therapeutic index, including targeting patients with minimal disease burden; using pre-targeting strategies, newer radionuclides, and improved labeling techniques; and using combined modalities and locoregional application. This review provides an overview of the radiolabeled intact antibodies currently in clinical use and those in development.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
| | - Sze Ting Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Hui K. Gan
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| | - Andrew M. Scott
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| |
Collapse
|
4
|
Li X, Yin M, Xie P, Liu Y, Li X, Qi Y, Ma Y, Li C, Wu G. Self-Expandable Metallic Stent Implantation Combined With Bronchial Artery Infusion Chemoembolization in the Treatment of Lung Cancer With Complete Atelectasis. Front Oncol 2022; 11:733510. [PMID: 35096562 PMCID: PMC8790529 DOI: 10.3389/fonc.2021.733510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background Atelectasis is a common complication of lung cancer, and there are few reports about the treatment methods. This study retrospectively analyzed the safety and effectiveness of endotracheal metal stent implantation combined with arterial infusion chemoembolization in the treatment of non-small cell lung cancer with complete atelectasis. Methods The clinical data of patients with non-small cell lung cancer and complete atelectasis treated by self-expandable metallic stent implantation combined with arterial infusion chemotherapy were retrospectively analyzed. The clinical efficacy was evaluated and postoperative adverse reactions were observed. Progression-free survival and overall survival were analyzed by Kaplan-Meier method. Results In all, 42 endotracheal metallic stents were implanted in 42 patients under fluoroscopy. 5–7 days after stent implantation, CT showed that 24 patients (57.1%) had complete lung recruitment, and that 13 (31.0%) had partial lung recruitment. The technical success rate was 100%, and the clinical success rate was 88.1% (37/42). 5–7 days after stent implantation, bronchial artery infusion chemoembolization was performed in all patients. The median progression-free survival and overall survival were 6 months (95% CI: 2.04-9.66) and 10 months (95% CI: 7.22-12.79), respectively. Conclusion Self-expandable metallic stent implantation combined with arterial infusion chemoembolization may be an effective and safe strategy in the treatment of lung cancer with atelectasis clinically.
Collapse
Affiliation(s)
- Xiaobing Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meipan Yin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengfei Xie
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Liu
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangnan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaozhen Ma
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunxia Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Oliveira MC, Correia JDG. Clinical application of radioiodinated antibodies: where are we? Clin Transl Imaging 2022. [DOI: 10.1007/s40336-021-00477-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
6
|
Suman SK, Subramanian S, Mukherjee A. Combination radionuclide therapy: A new paradigm. Nucl Med Biol 2021; 98-99:40-58. [PMID: 34029984 DOI: 10.1016/j.nucmedbio.2021.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022]
Abstract
Targeted molecular radionuclide therapy (MRT) has shown its potential for the treatment of cancers of multiple origins. A combination therapy strategy employing two or more distinct therapeutic approaches in cancer management is aimed at circumventing tumor resistance by simultaneously targeting compensatory signaling pathways or bypassing survival selection mutations acquired in response to individual monotherapies. Combination radionuclide therapy (CRT) is a newer application of the concept, utilizing a combination of radiolabeled molecular targeting agents with chemotherapy and beam radiation therapy for enhanced therapeutic index. Encouraging results are reported with chemotherapeutic agents in combination with radiolabeled targeting molecules for cancer therapy. With increasing awareness of the various survival and stress response pathways activated after radionuclide therapy, different holistic combinations of MRT agents with radiosensitizers targeting such pathways have also been explored. MRT has also been studied in combination with beam radiotherapy modalities such as external beam radiation therapy and carbon ion radiation therapy to enhance the anti-tumor response. Nanotechnology aids in CRT by bringing together multiple monotherapies on a single nanostructure platform for treating cancers in a more precise or personalized way. CRT will be a key player in managing cancers if correctly tailored to the individual patient profile. The success of CRT lies in an in-depth understanding of the radiobiological principles and pathways activated in response.
Collapse
Affiliation(s)
- Shishu Kant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Suresh Subramanian
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India.
| |
Collapse
|
7
|
Zhao M, Fu X, Zhang Z, Li A, Wang X, Li X. Intracranial 131I-chTNT Brachytherapy in Patients with Deep-Seated Glioma: A Single-center Experience with 10-Year Follow-up from China. Nuklearmedizin 2021; 60:283-288. [PMID: 33836553 DOI: 10.1055/a-1429-1967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The intracranial brachytherapy has been applied for decades, however, no results with long-term follow-up have been reported. This study investigated the long-term efficiency of intra-tumoral injection of 131I-chTNT in patients with deep-seated glioma. METHOD Thirty-five patients undergoing 131I-chTNT brachytherapy between December 2004 and May 2009 were enrolled. 131I-chTNT was injected at a dose of 1.5 mCi/cm3 at an interval of 1 month for consecutive 3 times. Serial ECT scan and MRI were performed during follow-up. Progression-free survival (PFS) and overall survival (OS) were analyzed. Adverse reactions were graded with WHO Toxicity Grading Scale for determining the severity of adverse events. RESULTS ECT scan showed that enhanced accumulation of radioactive agents in the tumor lasted for more than 30 days. Three months after final injection, tumor complete remission (CR) was observed in 4 patients (11.4 %), partial remission (PR) in 11 cases (31.4 %), stable disease (SD) in 10 cases (28.6 %) and progressive disease (PD) in 10 cases (28.6 %). At 6-month, CR, PR, SD and PD were 2, 6, 12 and 15 respectively. After 10 years of follow-up, median progression-free survival (PFS) and overall survival (OS) were 5.4 and 11.4 months. One-year survival was 45.7 %, two and five-year survival was 8.6 %, ten-year survival was 5.7 %. Multivariate analysis showed that pathological grade and tumor diameter were independent prognostic factors for PFS and OS. Grade I-II adverse events occurred after drug injection, including nausea, fever, headache, hairloss and fatigue. CONCLUSION 131I-chTNT intracranial brachytherapy is efficient and safe for patients with deep-seated glioma. It is a reliable option for inoperable glioma patients.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Neurosurgery, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiangping Fu
- Department of Neurosurgery, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhiwen Zhang
- Department of Neurosurgery, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Anmin Li
- Department of Neurosurgery, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaopeng Wang
- Department of Neurosurgery, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xuexiu Li
- Department of Neurosurgery, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Liu W, Tang Y, Ma H, Li F, Hu Y, Yang Y, Yang J, Liao J, Liu N. Astatine-211 labelled a small molecule peptide: specific cell killing in vitro and targeted therapy in a nude-mouse model. RADIOCHIM ACTA 2020. [DOI: 10.1515/ract-2020-0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
Extensive interest in the development of α-emitting radionuclides astatine-211 (211At) stems from the potential superiority for the treatment of smaller tumors, disseminated disease, and metastatic disease. VP2, a small molecule fusion peptide, can specifically bind to the VPAC1 receptor which is over-expressed in malignant epithelial tumors. In our recent study, we performed the preparation of 211At labelled VP2 through a one-step method. In this work, we explored the targeted radionuclide therapy with [211At]At-SPC-VP2 in vitro and in vivo. The cytotoxicity and specific cell killing of [211At]At-SPC-VP2 were evaluated using the CCK-8 assay. Compared with the [211At]NaAt, the VPAC1-targeted radionuclide compound [211At]At-SPC-VP2 showed more effective cytotoxicity in vitro. Targeted radioactive therapy trial was carried out in non-small-cell lung cancer (NSCLC) xenograft mice. For the therapy experiment, 4 groups of mice were injected via the tail vein with 370 kBq, 550 kBq, 740 kBq, 3 × ∼246 kBq of [211At]At-SPC-VP2, of which the second and third injections were given 4 and 8 days after the first injection, respectively. As controls, animals were treated with saline or 550 kBq [211At]NaAt. The body weight and tumor size of mice were monitored before the administration and every 2 days thereafter. Cytotoxic radiation of partial tissue samples such as kidneys, liver and stomach of mice were assessed by immunohistochemical examination. The tumor growth was inhibited and significantly improved survival was achieved in mice treated with [211At]At-SPC-VP2, two-fold prolongation of survival compared with the control group, which received normal saline or 550 kBq [211At]NaAt. No renal or hepatic toxicity was observed in the mice receiving [211At]At-SPC-VP2, but gastric pathological sections showed 211At uptake in stomach resulting in later toxicity, highlighting the importance of further enhancing the stability of labelled compounds.
Collapse
Affiliation(s)
- Weihao Liu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Yu Tang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Huan Ma
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Yingjiang Hu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Yuanyou Yang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Jijun Yang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Jiali Liao
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| |
Collapse
|
9
|
Zeng Y, Yin M, Zhao Y, Liu Y, Li X, Qi Y, Ma Y, Li Z, Li C, Wu G. Combination of Bronchial Arterial Infusion Chemotherapy plus Drug-Eluting Embolic Transarterial Chemoembolization for Treatment of Advanced Lung Cancer-A Retrospective Analysis of 23 Patients. J Vasc Interv Radiol 2020; 31:1645-1653. [PMID: 32951974 DOI: 10.1016/j.jvir.2020.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 05/13/2020] [Accepted: 06/15/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE To determine the efficacy and safety of the combination of bronchial arterial infusion (BAI) chemotherapy and transarterial chemoembolization with the use of drug-eluting embolic (DEE) particles in the treatment of unresectable advanced lung cancer. MATERIALS AND METHODS A retrospective review was performed of 23 patients with unresectable lung cancer (stage III/IV) who received BAI chemotherapy and DEE chemoembolization. Treatment response was assessed by enhanced CT and evaluated on the basis of Response Evaluation Criteria In Solid Tumors at 30 d after the last combination treatment. Patients were followed up until death or March 15, 2020, whichever was first. Overall survival (OS) was estimated by Kaplan-Meier analysis, and factors associated with OS were evaluated by Cox proportional-hazards test. RESULTS Complete response, partial response, stable disease, and progressive disease were seen in 2, 16, 5, and 0 patients at 30 d after the last combination treatment, respectively; therefore, the overall response rate was 78.3% and the disease control rate was 100%. Preprocedure symptoms (hemoptysis in 7 patients and dyspnea in 10) resolved in all cases after combination therapy. Nineteen patients died during follow-up, and 4 survived. Median OS was 15.6 mo (95% confidence interval, 10.1-21.1 mo). On univariate analysis and multivariate analysis, tumor/node/metastasis staging was an independent risk factor for prognosis. There were no serious adverse events during the procedures. CONCLUSIONS The combination of BAI chemotherapy plus DEE chemoembolization appears to be a promising method for treatment of advanced lung cancer.
Collapse
Affiliation(s)
- Yanwei Zeng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Meipan Yin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Yue Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Ying Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Xiangnan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Yu Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Yaozhen Ma
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Chunxia Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China
| | - Gang Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Rd., Zhengzhou 450052, China.
| |
Collapse
|
10
|
Zhang D, Gao M, Jin Q, Ni Y, Zhang J. Updated developments on molecular imaging and therapeutic strategies directed against necrosis. Acta Pharm Sin B 2019; 9:455-468. [PMID: 31193829 PMCID: PMC6543088 DOI: 10.1016/j.apsb.2019.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/07/2018] [Accepted: 01/07/2019] [Indexed: 12/15/2022] Open
Abstract
Cell death plays important roles in living organisms and is a hallmark of numerous disorders such as cardiovascular diseases, sepsis and acute pancreatitis. Moreover, cell death also plays a pivotal role in the treatment of certain diseases, for example, cancer. Noninvasive visualization of cell death contributes to gained insight into diseases, development of individualized treatment plans, evaluation of treatment responses, and prediction of patient prognosis. On the other hand, cell death can also be targeted for the treatment of diseases. Although there are many ways for a cell to die, only apoptosis and necrosis have been extensively studied in terms of cell death related theranostics. This review mainly focuses on molecular imaging and therapeutic strategies directed against necrosis. Necrosis shares common morphological characteristics including the rupture of cell membrane integrity and release of cellular contents, which provide potential biomarkers for visualization of necrosis and necrosis targeted therapy. In the present review, we summarize the updated joint efforts to develop molecular imaging probes and therapeutic strategies targeting the biomarkers exposed by necrotic cells. Moreover, we also discuss the challenges in developing necrosis imaging probes and propose several biomarkers of necrosis that deserve to be explored in future imaging and therapy research.
Collapse
Affiliation(s)
- Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yicheng Ni
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
- Theragnostic Laboratory, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
11
|
Staudacher AH, Li Y, Liapis V, Hou JJC, Chin D, Dolezal O, Adams TE, van Berkel PH, Brown MP. APOMAB Antibody–Drug Conjugates Targeting Dead Tumor Cells are Effective In Vivo. Mol Cancer Ther 2018; 18:335-345. [DOI: 10.1158/1535-7163.mct-18-0842] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/05/2018] [Accepted: 11/05/2018] [Indexed: 11/16/2022]
|
12
|
Lopez T, Ramirez A, Benitez C, Mustafa Z, Pham H, Sanchez R, Ge X. Selectivity Conversion of Protease Inhibitory Antibodies. Antib Ther 2018. [PMID: 30406213 PMCID: PMC7990135 DOI: 10.1093/abt/tby010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Solid tumors are inherently difficult to treat because of large regions of hypoxia and are often chemotherapy- or radiotherapy-resistant. It seems that cancer stem cells reside in hypoxic and adjacent necrotic tumor areas. Therefore, new treatments that are highly selective for tumors and can eradicate cells in both hypoxic and necrotic tumor regions are desirable. Antibody α-radioconjugates couple an α-emitting radionuclide with the specificity of a tumor-targeting monoclonal antibody. The large mass and energy of α-particles result in radiation dose delivery within a smaller area independent of oxygen concentration, thus matching key criteria for killing hypoxic tumor cells. With advances in radionuclide production and chelation chemistry, α-radioconjugate therapy is regaining interest as a cancer therapy. Here, we will review current literature examining radioconjugate therapy specifically targeting necrotic and hypoxic tumor cells and outline how α-radioconjugate therapy could be used to treat tumor regions harboring more resistant cancer cell types. Statement of Significance Tumor-targeting antibodies are excellent vehicles for the delivery of toxic payloads directly to the tumor site. Tumor hypoxia and necrosis promote treatment recurrence, resistance, and metastasis. Targeting these areas with antibody α-radioconjugates would aid in overcoming treatment resistance.
Collapse
Affiliation(s)
- Tyler Lopez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Aaron Ramirez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Chris Benitez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Zahid Mustafa
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Henry Pham
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Ramon Sanchez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Xin Ge
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
13
|
Staudacher AH, Liapis V, Brown MP. Selectivity Conversion of Protease Inhibitory Antibodies. Antib Ther 2018; 1:55-63. [PMID: 30406213 PMCID: PMC7990135 DOI: 10.1093/abt/tby008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 11/14/2022] Open
Abstract
Background: Proteases are one of the largest pharmaceutical targets for drug developments. Their dysregulations result in a wide variety of diseases. Because proteolytic networks usually consist of protease family members that share high structural and catalytic homology, distinguishing them using small molecule inhibitors is often challenging. To achieve specific inhibition, this study described a novel approach for the generation of protease inhibitory antibodies. As a proof of concept, we aimed to convert a matrix metalloproteinase (MMP)-14 specific inhibitor to MMP-9 specific inhibitory antibodies with high selectivity. Methods: An error-prone single-chain Fv (scFv) library of an MMP-14 inhibitor 3A2 was generated for yeast surface display. A dual-color competitive FACS was developed for selection on MMP-9 catalytic domain (cdMMP-9) and counter-selection on cdMMP-14 simultaneously, which were fused/conjugated with different fluorophores. Isolated MMP-9 inhibitory scFvs were biochemically characterized by inhibition assays on MMP-2/-9/-12/-14, proteolytic stability tests, inhibition mode determination, competitive ELISA with TIMP-2 (a native inhibitor of MMPs), and paratope mutagenesis assays. Results: We converted an MMP-14 specific inhibitor 3A2 into a panel of MMP-9 specific inhibitory antibodies with dramatic selectivity shifts of 690-4,500 folds. Isolated scFvs inhibited cdMMP-9 at nM potency with high selectivity over MMP-2/-12/-14 and exhibited decent proteolytic stability. Biochemical characterizations revealed that these scFvs were competitive inhibitors binding to cdMMP-9 near its reaction cleft via their CDR-H3s. Conclusions: This study developed a novel approach able to convert the selectivity of inhibitory antibodies among closely related protease family members. This methodology can be directly applied for mAbs inhibiting many proteases of biomedical importance.
Collapse
Affiliation(s)
- Alexander H Staudacher
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
- School of Medicine, University of Adelaide, Adelaide, Australia
| | - Vasilios Liapis
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
- School of Medicine, University of Adelaide, Adelaide, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, Australia
| |
Collapse
|
14
|
Boros E, Holland JP. Chemical aspects of metal ion chelation in the synthesis and application antibody-based radiotracers. J Labelled Comp Radiopharm 2018; 61:652-671. [PMID: 29230857 PMCID: PMC5997514 DOI: 10.1002/jlcr.3590] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022]
Abstract
Radiometals are becoming increasingly accessible and are utilized frequently in the design of radiotracers for imaging and therapy. Nuclear properties ranging from the emission of γ-rays and β+ -particles (imaging) to Auger electron and β- and α-particles (therapy) in combination with long half-lives are ideally matched with the relatively long biological half-life of monoclonal antibodies in vivo. Radiometal labeling of antibodies requires the incorporation of a metal chelate onto the monoclonal antibody. This chelate must coordinate the metal under mild conditions required for the handling of antibodies, as well as provide high kinetic, thermodynamic, and metabolic stability once the metal ion is coordinated to prevent release of the radionuclide before the target site is reached in vivo. Herein, we review the role of different radiometals that have found applications of the design of radiolabeled antibodies for imaging and radioimmunotherapy. Each radionuclide is described regarding its nuclear synthesis, coordinative preference, and radiolabeling properties with commonly used and novel chelates, as well as examples of their preclinical and clinical applications. An overview of recent trends in antibody-based radiopharmaceuticals is provided to spur continued development of the chemistry and application of radiometals for imaging and therapy.
Collapse
Affiliation(s)
- Eszter Boros
- Stony Brook University, Department of Chemistry, 100 Nicolls road, 11790 Stony Brook, NY, United States
| | - Jason P. Holland
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| |
Collapse
|
15
|
Yang N, Yao S, Liu D. Tumor necrosis factor-related apoptosis-inducing ligand additive with Iodine-131 of inhibits non-small cell lung cancer cells through promoting apoptosis. Oncol Lett 2018; 16:276-284. [PMID: 29928412 PMCID: PMC6006446 DOI: 10.3892/ol.2018.8635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 01/12/2018] [Indexed: 11/26/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) accounts for ~80% of human lung cancer cases and is the most common cause of cancer-associated mortality worldwide. Reports have indicated that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and Iodine-131 (I-131) can induce tumor cell apoptosis. The purpose of the present study was to investigate the additive efficacy of TRAIL and I-131 on NSCLC cells. The present study demonstrated that additive treatment of TRAIL and I-131 (TRAIL-I-131) significantly inhibited the growth and aggressiveness of NSCLC cells compared with single TRAIL or I-131 treatment. Results demonstrated that TRAIL-I-131 treatment induced apoptosis of NSCLC cells, with western blot analysis confirming that TRAIL-I-131 treatment increased proapoptotic Bad and Bax expression levels, while antiapoptotic Bcl-2 and Bcl-w protein levels were decreased in NSCLC cells. The present study demonstrated that TRAIL-I-131 treatment inhibited vascular endothelial growth factor (VEGF) and activator protein-1 (AP-1) in NSCLC cells. Potential mechanism analyses identified that TRAIL-I-131 treatment induced apoptosis of NSCLC cells through caspase-9 activation. In vivo assays revealed that TRAIL-I-131 treatment significantly inhibited NSCLC tumor growth and increased apoptotic bodies in tumor tissues. Immunohistology demonstrated that caspase-9 was upregulated and VEGF was downregulated in tumor tissues in TRAIL-I-131-treated tumors. In conclusion, these results indicate that TRAIL combined with I-131 promoted apoptosis of NSCLC through caspase-9 activation, which may be a promising anticancer therapeutic schedule for the treatment of NSCLC.
Collapse
Affiliation(s)
- Ning Yang
- Department of Nuclear Medicine, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| | - Shuzhan Yao
- Positron Emission Tomography/Computed Tomography Center, Shandong Provincial Hospital, Jinan, Shandong 250012, P.R. China
| | - Dong Liu
- Department of Nuclear Medicine, Central Hospital of Zibo, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
16
|
Therapeutic Antibodies in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 917:95-120. [PMID: 27236554 DOI: 10.1007/978-3-319-32805-8_6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The therapeutic arsenal in solid tumors comprises different anticancer strategies with diverse chemotherapeutic agents and a growing number of biological substances. Large clinical study-based chemotherapeutic protocols combined with biologicals have become an important component in (neo-) adjuvant therapy alongside surgery in solid cancers as well as radiation therapy in some instances. In recent years, monoclonal antibodies have entered the mainstream of cancer therapy. Their first use was as antagonists of oncogenic receptor tyrosine kinases, but today monoclonal antibodies have emerged as long-sought vehicles for the targeted delivery of potent chemotherapeutic agents and as powerful tools to manipulate anticancer immune responses. There is a growing number of FDA approved monoclonal antibodies and small molecules targeting specific types of cancer suggestive of the clinical relevance of this approach.Targeted cancer therapies , also referred to as personalized medicine, are being studied for use alone, in combination with other targeted therapies, and in combination with chemotherapy. The use of monoclonal antibodies in colorectal and gastric cancer for example have shown best outcome when combined with chemotherapy, even though single agent anti-EGFR antibodies seem to be active in particular setting of metastatic colorectal cancer patients. However, it is not well defined whether the addition of anti-VEGF - and anti-EGFR strategies to chemotherapy could improve outcome in those patients susceptible to colorectal cancer-related metastases resection. Among the most promising approaches to activating therapeutic antitumor immunity is the blockade of immune checkpoints, exemplified by the recently FDA-approved agent, Ipilimumab, an antibody that blocks the coinhibitory receptor CTLA-4. Capitalizing on the success of Ipilimumab, agents that target a second coinhibitory receptor, PD-1, or its ligand, PD-L1, are in clinical development. This section attempts to discuss recent progress of targeted agents and in tackling a more general target applicable to gastrointestinal cancer .
Collapse
|
17
|
An atlas of bloodstream-accessible bone marrow proteins for site-directed therapy of acute myeloid leukemia. Leukemia 2017; 32:510-519. [DOI: 10.1038/leu.2017.208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/15/2022]
|
18
|
Targeted radionuclide therapy for lung cancer with iodine-131-labeled peptide in a nude-mouse model. Anticancer Drugs 2017; 28:480-488. [DOI: 10.1097/cad.0000000000000481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
19
|
He M, Li S, Chen Y, Ouyang M, Chen P, Zhang J. 131I-chTNT injection to relieve tracheal obstruction in advanced NSCLC patient. Technol Health Care 2017; 24 Suppl 2:S513-9. [PMID: 27163312 DOI: 10.3233/thc-161176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To relieve large airway obstruction in a patient with advanced non-small cell lung cancer (NSCLC) by injecting the mouse-human chimeric monoclonal antibody radiolabeled with iodine 131 chimeric tumor necrotic treatment (131I-chTNT) and to study the irradiation absorption in the tumor and critical organs. METHODS A 50-year-old patient with NSCLC was treated with radioimmunotherapy. His airway was still obstructed in spite of intensive chemotherapy and radiotherapy.131I-chTNT was injected into the tumor at the right bronchus through a fiberscope. A131I scan was performed during treatment, and a computed tomography (CT) scan of the chest and fiberscope were performed pre- and post-treatment.131I-chTNT distribution in tissues was followed for up to 4 weeks using gamma camera imaging. RESULTS The radiation material accumulated notably in the tumor, relieving the patient's symptoms by suppressing the tumor. Recanalization of the airway was achieved so that the patient was able to breathe easily and cough. CONCLUSION As a new type of radioimmunotherapy,131I-chTNT may be helpful in treatment of advanced lung cancer.
Collapse
|
20
|
Bethge WA, Sandmaier BM. Targeted Cancer Therapy Using Radiolabeled Monoclonal Antibodies. Technol Cancer Res Treat 2016; 4:393-405. [PMID: 16029058 DOI: 10.1177/153303460500400407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Radioimmunotherapy (RIT) combines the advantages of targeted radiation therapy and specific immunotherapy using monoclonal antibodies. RIT can be used either to target tumor cells or to specifically suppress immunocompetent host cells in the setting of allogeneic transplantation. The choice of radionuclide used for RIT depends on its distinct radiation characteristics and the type of malignancy or cells targeted. Beta-emitters with their lower energy and longer path length are more suitable to target bulky, solid tumors whereas α-emitters with their high linear energy transfer and short path length are better suited to target hematopoietic cells (normal or malignant). Different approaches of RIT such as the use of stable radioimmunoconjugates or of pretargeting strategies are available. Encouraging results have been obtained with RIT in patients with hematologic malignancies. The results in solid tumors are somewhat less favorable but new strategies for patients with minimal residual disease using adjuvant and locoregional treatment are evolving. This report outlines basic principles of RIT, gives an overview of available radionuclides and radioimmunoconjugates, and discusses clinical results with special emphasis on their use in hematologic malignancies including use in conditioning regimens for bone marrow transplantation.
Collapse
Affiliation(s)
- Wolfgang A Bethge
- Medical Center, University of Tuebingen, Department of Hematology and Oncology, Otfried-Mueller Str. 10, 72076 Tuebingen, Germany
| | | |
Collapse
|
21
|
Shao H, Zhang J, Sun Z, Chen F, Dai X, Li Y, Ni Y, Xu K. Necrosis targeted radiotherapy with iodine-131-labeled hypericin to improve anticancer efficacy of vascular disrupting treatment in rabbit VX2 tumor models. Oncotarget 2016; 6:14247-59. [PMID: 26036625 PMCID: PMC4546464 DOI: 10.18632/oncotarget.3679] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 03/03/2015] [Indexed: 11/25/2022] Open
Abstract
A viable rim of tumor cells surrounding central necrosis always exists and leads to tumor recurrence after vascular disrupting treatment (VDT). A novel necrosis targeted radiotherapy (NTRT) using iodine-131-labeled hypericin (131I-Hyp) was specifically designed to treat viable tumor rim and improve tumor control after VDT in rabbit models of multifocal VX2 tumors. NTRT was administered 24 hours after VDT. Tumor growth was significantly slowed down by NTRT with a smaller tumor volume and a prolonged tumor doubling time (14.4 vs. 5.7 days), as followed by in vivo magnetic resonance imaging over 12 days. The viable tumor rims were well inhibited in NTRT group compared with single VDT control group, as showed on tumor cross sections at day 12 (1 vs. 3.7 in area). High targetability of 131I-Hyp to tumor necrosis was demonstrated by in vivo SPECT as high uptake in tumor regions lasting over 9 days with 4.26 to 98 times higher radioactivity for necrosis versus the viable tumor and other organs by gamma counting, and with ratios of 7.7-11.7 and 10.5-13.7 for necrosis over peri-tumor tissue by autoradiography and fluorescence microscopy, respectively. In conclusion, NTRT improved the anticancer efficacy of VDT in rabbits with VX2 tumors.
Collapse
Affiliation(s)
- Haibo Shao
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Jian Zhang
- Laboratory of Translational Medicine, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Ziping Sun
- Radiation Medical Institute, Shandong Academy of Medical Sciences, Jinan, China
| | - Feng Chen
- Department of Imaging & Pathology, Theragnostic Laboratory, University of Leuven, Leuven, Belgium
| | - Xu Dai
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Yaming Li
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Yicheng Ni
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China.,Laboratory of Translational Medicine, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China.,Radiation Medical Institute, Shandong Academy of Medical Sciences, Jinan, China.,Department of Imaging & Pathology, Theragnostic Laboratory, University of Leuven, Leuven, Belgium
| | - Ke Xu
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Jang JK, Khawli LA, Canter DC, Hu P, Zhu TH, Wu BW, Angell TE, Li Z, Epstein AL. Systemic delivery of chTNT-3/CpG immunoconjugates for immunotherapy in murine solid tumor models. Cancer Immunol Immunother 2016; 65:511-23. [PMID: 26960932 DOI: 10.1007/s00262-016-1813-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/19/2016] [Indexed: 12/25/2022]
Abstract
CpG oligodeoxynucleotides (CpG) potently activate the immune system by mimicking microbial DNA. Conjugation of CpG to chTNT-3, an antibody targeting the necrotic centers of tumors, enabled CpG to accumulate in tumors after systemic delivery, where it can activate the immune system in the presence of tumor antigens. CpG chemically conjugated to chTNT-3 (chTNT-3/CpG) were compared to free CpG in their ability to stimulate the immune system in vitro and reduce tumor burden in vivo. In subcutaneous Colon 26 adenocarcinoma and B16-F10 melanoma models in BALB/c and C57BL/6 mice, respectively, chTNT-3/CpG, free CpG, or several different control constructs were administered systemically. Intraperitoneal injections of chTNT-3/CpG delayed tumor growth and improved survival and were comparable to intratumorally administered CpG. Compared to saline-treated mice, chTNT-3/CpG-treated mice had smaller average tumor volumes by as much as 72% in Colon 26-bearing mice and 79% in B16-bearing mice. Systemically delivered free CpG and CpG conjugated to an isotype control antibody did not reduce tumor burden or improve survival. In this study, chTNT-3/CpG retained immunostimulatory activity of the CpG moiety and enabled delivery to tumors. Because systemically administered CpG rapidly clear the body and do not accumulate into tumors, chTNT-3/CpG provide a solution to the limitations observed in preclinical and clinical trials.
Collapse
Affiliation(s)
- Julie K Jang
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR 205, Los Angeles, CA, 90033, USA
| | - Leslie A Khawli
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR 205, Los Angeles, CA, 90033, USA
| | - David C Canter
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR 205, Los Angeles, CA, 90033, USA
| | - Peisheng Hu
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR 205, Los Angeles, CA, 90033, USA
| | - Tian H Zhu
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR 205, Los Angeles, CA, 90033, USA
| | - Brian W Wu
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR 205, Los Angeles, CA, 90033, USA
| | - Trevor E Angell
- Department of Endocrinology, Metabolism, and Hypertension, Thyroid Section, Brigham and Women's Hospital, Boston, MA, USA
| | - Zhongjun Li
- Department of Blood Transfusion, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Alan L Epstein
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR 205, Los Angeles, CA, 90033, USA.
| |
Collapse
|
23
|
Parakh S, Parslow AC, Gan HK, Scott AM. Antibody-mediated delivery of therapeutics for cancer therapy. Expert Opin Drug Deliv 2015; 13:401-19. [PMID: 26654403 DOI: 10.1517/17425247.2016.1124854] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Antibody-conjugated therapies (ACTs) combine the specificity of monoclonal antibodies to target cancer cells directly with highly potent payloads, often resulting in superior efficacy and/or reduced toxicity. This represents a new approach to the treatment of cancer. There have been highly promising clinical trial results using this approach with improvements in linker and payload technology. The breadth of current trials examining ACTs in haematological malignancies and solid tumours indicate the potential for clinical impact. AREAS COVERED This review will provide an overview of ACTs currently in clinical development as well as the principles of antibody delivery and types of payloads used, including cytotoxic drugs, radiolabelled isotopes, nanoparticle-based siRNA particles and immunotoxins. EXPERT OPINION The focus of much of the clinical activity in ACTs has, understandably, been on their use as a monotherapy or in combination with standard of care drugs. This will continue, as will the search for better targets, linkers and payloads. Increasingly, as these drugs enter routine clinical care, important questions will arise regarding how to optimise ACT treatment approaches, including investigation of resistance mechanisms, biomarker and patient selection strategies, understanding of the unique toxicities of these drugs, and combinatorial approaches with standard therapies as well as emerging therapeutic agents like immunotherapy.
Collapse
Affiliation(s)
- Sagun Parakh
- a Tumour Targeting Laboratory , Olivia Newton-John Cancer Research Institute , Melbourne , Australia.,b Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre , Austin Health , Heidelberg, Melbourne , Australia.,c School of Cancer Medicine , La Trobe University , Melbourne , Australia
| | - Adam C Parslow
- a Tumour Targeting Laboratory , Olivia Newton-John Cancer Research Institute , Melbourne , Australia.,c School of Cancer Medicine , La Trobe University , Melbourne , Australia
| | - Hui K Gan
- a Tumour Targeting Laboratory , Olivia Newton-John Cancer Research Institute , Melbourne , Australia.,b Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre , Austin Health , Heidelberg, Melbourne , Australia.,c School of Cancer Medicine , La Trobe University , Melbourne , Australia
| | - Andrew M Scott
- a Tumour Targeting Laboratory , Olivia Newton-John Cancer Research Institute , Melbourne , Australia.,c School of Cancer Medicine , La Trobe University , Melbourne , Australia.,d Departmentof Molecular Imaging and Therapy , Austin Health , Melbourne , Australia.,e Department of Medicine , University of Melbourne , Melbourne , Australia
| |
Collapse
|
24
|
Jang JK, Chretin J, Bruyette D, Hu P, Epstein AL. Phase 1 Dose-Escalation Study with LEC/chTNT-3 and Toceranib Phosphate (Palladia ®) in Dogs with Spontaneous Malignancies. ACTA ACUST UNITED AC 2015; 7:167-174. [PMID: 26635918 DOI: 10.4172/1948-5956.1000343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES LEC chemokine promotes TH1 responses and recruits immune cells to inflammatory sites. By linking LEC to an antibody targeting tumor necrosis, LEC/chTNT-3 can be used for the immunotherapeutic treatment of tumors. The primary objective of this study was to determine the safety profile of LEC/chTNT-3 and toceranib phosphate (Palladia®) combination therapy in dogs with spontaneous malignancies. Secondary purpose was to determine objective responses to treatment. METHODS Twenty-three dogs with cancer were enrolled, covering nine different malignancies. In this dose escalation study, dogs received LEC/chTNT-3 for five days, and toceranib every 48 hours for the remainder of the study. Dogs received physical exams, chemistry panel, urinalysis, and complete blood counts on days 0, 10, 28 of the study, and every 6-8 weeks thereafter. RESULTS Lethargy was noted in 13% dogs. There were no statistical differences in the prevalence of anorexia, diarrhea, thrombocytopenia, renal toxicity, or hepatic toxicity before or during the study. There were trends in increases in the prevalence of vomiting, lymphopenia, and neutropenia (all grade 2 or lower, p=0.07) over the initial 28 days of the study. By day 28, 10% of dogs had partial responses, 58% had stable disease, and 32% had progressive disease. CONCLUSIONS LEC/chTNT-3 and toceranib were well tolerated. This combination therapy showed some biological activity against a variety of cancers at a low dose and short duration of LEC/chTNT-3 administration.
Collapse
Affiliation(s)
- Julie K Jang
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - John Chretin
- Veterinary Centers of America West Los Angeles Animal Hospital, Los Angeles, CA, USA
| | - David Bruyette
- Veterinary Centers of America West Los Angeles Animal Hospital, Los Angeles, CA, USA
| | - Peisheng Hu
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Alan L Epstein
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
25
|
Yeong CH, Cheng MH, Ng KH. Therapeutic radionuclides in nuclear medicine: current and future prospects. J Zhejiang Univ Sci B 2015; 15:845-63. [PMID: 25294374 DOI: 10.1631/jzus.b1400131] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The potential use of radionuclides in therapy has been recognized for many decades. A number of radionuclides, such as iodine-131 ((131)I), phosphorous-32 ((32)P), strontium-90 ((90)Sr), and yttrium-90 ((90)Y), have been used successfully for the treatment of many benign and malignant disorders. Recently, the rapid growth of this branch of nuclear medicine has been stimulated by the introduction of a number of new radionuclides and radiopharmaceuticals for the treatment of metastatic bone pain and neuroendocrine and other malignant or non-malignant tumours. Today, the field of radionuclide therapy is enjoying an exciting phase and is poised for greater growth and development in the coming years. For example, in Asia, the high prevalence of thyroid and liver diseases has prompted many novel developments and clinical trials using targeted radionuclide therapy. This paper reviews the characteristics and clinical applications of the commonly available therapeutic radionuclides, as well as the problems and issues involved in translating novel radionuclides into clinical therapies.
Collapse
Affiliation(s)
- Chai-Hong Yeong
- Department of Biomedical Imaging & University of Malaya Research Imaging Centre, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; Department of Nuclear Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | | | | |
Collapse
|
26
|
Effectiveness of combined (131)I-chTNT and radiofrequency ablation therapy in treating advanced hepatocellular carcinoma. Cell Biochem Biophys 2015; 71:777-84. [PMID: 25293788 DOI: 10.1007/s12013-014-0262-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
To investigate the effectiveness of monoclonal antibody ((131)I-chTNT) and radiofrequency ablation (RFA) combination therapy in treating middle-advanced stage hepatocellular carcinoma (HCC). Thirty-four patients diagnosed with HCC patients, divided into two groups comprised of 22 and 12 cases were included in this retrospective study. The two groups received RFA with or without ((131)I-chTNT) therapy, respectively. The patients in these groups were followed up for a median of 31 and 35 months, respectively. Patient survival was evaluated using Kaplan-Meier method and safety profiles were determined by analyzing liver, thyroid, and bone marrow toxicities. This retrospective study showed that survival time of the patients who received combination therapy was significantly longer than that of the RFA group (P = 0.052). The median progress-free survival of patients in the two groups was 23 and 7 months, respectively, and the difference was significant (P = 0.04). Tumor recurred in 3.5-8.7 months in four of the combination group patients, among which three had newly developed lesions. The red blood cells and platelets counts were not altered on day 7 and 1 month of the treatment, however, number of white blood cells was significantly increased on day 7 which was reversed back to the normal range in 2 weeks. The ALT and AST were also not significantly altered on day 7 and 1 month of therapy. In middle-advanced stage HCC patients, the combination of (131)I-chTNT and RFA therapy was found to be significantly more effective than the RFA treatment alone as assessed in short-term follow-up. However, the dose we used was insufficient to completely block the local recurrence of the lesions with a diameter of 5 cm or larger.
Collapse
|
27
|
Liu X, Jiang C, Zhang D, Gao M, Peng F, Huang D, Sun Z, Ni Y, Zhang J, Yin Z. Tumor necrosis targeted radiotherapy of non-small cell lung cancer using radioiodinated protohypericin in a mouse model. Oncotarget 2015; 6:26400-10. [PMID: 26305548 PMCID: PMC4694910 DOI: 10.18632/oncotarget.4568] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/10/2015] [Indexed: 12/31/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death. About 80% of lung cancers are non-small cell lung cancers (NSCLC). Radiotherapy is widely used in treatment of NSCLC. However, the outcome of NSCLC remains unsatisfactory. In this study, a vascular disrupting agent (VDA) combretastatin-A4-phosphate (CA4P) was used to provide massive necrosis targets. (131)I labeled necrosis-avid agent protohypericin ((131)I-prohy) was explored for therapy of NSCLC using tumor necrosis targeted radiotherapy (TNTR). Gamma counting, autoradiography, fluorescence microscopy and histopathology were used for biodistribution analysis. Magnetic resonance imaging (MRI) was used to monitor tumor volume, ratios of necrosis and tumor doubling time (DT). The biodistribution data revealed 131I-prohy was delivered efficiently to tumors. Tracer uptake peaked at 24 h in necrotic tumor of (131)I-prohy with and without combined CA4P (3.87 ± 0.38 and 2.96 ± 0.34%ID/g). (131)I-prohy + CA4P enhanced the uptake of (131)I-prohy in necrotic tumor compared to (131)I-prohy alone. The TNTR combined with CA4P prolonged survival of tumor bearing mice relative to vehicle control group, CA4P control group and (131)I-prohy control group with median survival of 35, 20, 22 and 27 days respectively. In conclusion, TNTR appeared to be effective for the treatment of NSCLC.
Collapse
Affiliation(s)
- Xuejiao Liu
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, P.R.China
| | - Cuihua Jiang
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P.R.China
| | - Dongjian Zhang
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P.R.China
| | - Meng Gao
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
| | - Fei Peng
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
| | - Dejian Huang
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
| | - Ziping Sun
- Shandong Academy of Medical Sciences, Jinan 250062, Shandong, P.R.China
| | - Yicheng Ni
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
- Theragnostic Laboratory, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium
| | - Jian Zhang
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China
| | - Zhiqi Yin
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P.R.China
| |
Collapse
|
28
|
Liu D, Auguste DT. Cancer targeted therapeutics: From molecules to drug delivery vehicles. J Control Release 2015; 219:632-643. [PMID: 26342659 DOI: 10.1016/j.jconrel.2015.08.041] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023]
Abstract
The pitfall of all chemotherapeutics lies in drug resistance and the severe side effects experienced by patients. One way to reduce the off-target effects of chemotherapy on healthy tissues is to alter the biodistribution of drug. This can be achieved in two ways: Passive targeting utilizes shape, size, and surface chemistry to increase particle circulation and tumor accumulation. Active targeting employs either chemical moieties (e.g. peptides, sugars, aptamers, antibodies) to selectively bind to cell membranes or responsive elements (e.g. ultrasound, magnetism, light) to deliver its cargo within a local region. This article will focus on the systemic administration of anti-cancer agents and their ability to home to tumors and, if relevant, distant metastatic sites.
Collapse
Affiliation(s)
- Daxing Liu
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, United States
| | - Debra T Auguste
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, United States.
| |
Collapse
|
29
|
Targeted α-therapy using 227Th-APOMAB and cross-fire antitumour effects: preliminary in-vivo evaluation. Nucl Med Commun 2015; 35:1284-90. [PMID: 25192189 DOI: 10.1097/mnm.0000000000000199] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Resistance to conventional cancer treatments is a major problem associated with solid tumours. Tumour hypoxia is associated with a poor prognosis and with poor treatment outcomes; therefore, there is a need for treatments that can kill hypoxic tumour cells. One potential option is targeted α-radioimmunotherapy, as α-particles can directly kill hypoxic tumour cells. The murine monoclonal antibody DAB4 (APOMAB), which binds dead tumour cells after DNA-damaging treatment, was conjugated and radiolabelled with the α-particle-emitting radionuclide thorium-227 (Th). Mice bearing Lewis lung tumours were administered Th-DAB4 alone or after chemotherapy and the tissue biodistribution of the radioimmunoconjugate was examined, as was the effect of these treatments on tumour growth and survival. Th-DAB4 accumulated in the tumour particularly after chemotherapy, whereas the distribution in healthy tissues did not change. Th-DAB4 as a monotherapy increased survival, with more pronounced responses observed when given after chemotherapy. We have shown that targeted α-therapy of necrotic tumour cells with Th-DAB4 had significant and surprising antitumour activity as it would occur only through a cross-fire effect.
Collapse
|
30
|
Abstract
For 20 years, monoclonal antibodies (mAbs) have been a standard component of cancer therapy, but there is still much room for improvement. Efforts continue to build better cancer therapeutics based on mAbs. Anticancer mAbs function through various mechanisms, including directly targeting the malignant cells, modifying the host response, delivering cytotoxic moieties and retargeting cellular immunity towards the malignant cells. Characteristics of mAbs that affect their efficacy include antigen specificity, overall structure, affinity for the target antigen and how a mAb component is incorporated into a construct that can trigger target cell death. This Review discusses the various approaches to using mAb-based therapeutics to treat cancer and the strategies used to take advantage of the unique potential of each approach, and provides examples of current mAb-based treatments.
Collapse
Affiliation(s)
- George J. Weiner
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, 5970Z‐JPP, Iowa City, Iowa 52242, USA.
| |
Collapse
|
31
|
Li Y, Liu X, Zhang D, Lou B, Peng F, Wang X, Shan X, Jiang C, Gao M, Sun Z, Ni Y, Huang D, Zhang J. Evaluation of a metalloporphyrin (THPPMnCl) for necrosis-affinity in rat models of necrosis. J Drug Target 2015; 23:926-35. [PMID: 25950601 DOI: 10.3109/1061186x.2015.1036358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The combination of an (13I)I-labeled necrosis-targeting agent (NTA) with a vascular disrupting agent is a novel and potentially powerful technique for tumor necrosis treatment (TNT). The purpose of this study was to evaluate a NTA candidate, THPPMnCl, using (131)I isotope for tracing its biodistribution and necrosis affinity. (131)I-THPPMnCl was intravenously injected in rat models with liver, muscle, and tumor necrosis and myocardial infarction (MI), followed by investigations with macroscopic autoradiography, triphenyltetrazolium chloride (TTC) histochemical staining, fluorescence microscopy and H&E stained histology for up to 9 days. (131)I-THPPMnCl displayed a long-term affinity for all types of necrosis and accumulation in the mononuclear phagocytic system especially in the liver. Autoradiograms and TTC staining showed a good targetability of (131)I-THPPMnCl for MI. These findings indicate the potential of THPPMnCl for non-invasive imaging assessment of necrosis, such as in MI. However, (13I)I-THPPMnCl is unlikely suitable for TNT due to its long-term retention in normal tissues.
Collapse
Affiliation(s)
- Yue Li
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Xuejiao Liu
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Dongjian Zhang
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Bin Lou
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Fei Peng
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Xiaoning Wang
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Xin Shan
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Cuihua Jiang
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Meng Gao
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Ziping Sun
- b Radiation Medical Institute, Shandong Academy of Medical Sciences , Jinan , Shandong Province , P.R. China , and
| | - Yicheng Ni
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and.,c Department of Radiology , KU Leuven , Leuven , Belgium
| | - Dejian Huang
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| | - Jian Zhang
- a Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu Province , P.R. China and
| |
Collapse
|
32
|
Ji Y, Jiang C, Zhang X, Liu W, Gao M, Li Y, Wang J, Wang Q, Sun Z, Jiang X, Yao N, Wang X, Fang Z, Yin Z, Ni Y, Zhang J. Necrosis targeted combinational theragnostic approach using radioiodinated Sennidin A in rodent tumor models. Oncotarget 2015; 5:2934-46. [PMID: 24931286 PMCID: PMC4102781 DOI: 10.18632/oncotarget.1728] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Residual cancer cells and subsequent tumor relapse is an obstacle for curative cancer treatment. Tumor necrosis therapy (TNT) has recently been developed to cause residual tumor regression or destruction. Here, we exploited the avidity of the sennidin A (SA) tracer and radioiodinated SA (131I-SA) to necrotic tumors in order to further empower TNT. We showed high uptake and prolonged retention of SA in necrotic tumors and a quick clearance in other non-targeted tissues including the liver. On SPECT-CT images, tumor mass appeared persistently as a hotspot. Based on the prominent targetability of 131I-SA to the tumor necrosis, we designed a combinational theragnostic modality. The vascular disrupting agent (VDA) combretastatin A4 phosphate (CA4P) was used to cause massive tumor necrosis, which formed the target of 131I-SA that subsequently killed the residual tumor cells by cross-fire irradiation of beta particles. Consequently, 131I-SA combined with CA4P significantly inhibited tumor growth, extended tumor doubling time and prolonged mean animal survival. In conclusion, 131I-SA in combination with necrosis inducing drugs/therapies may generate synergetic tumoricidal effects on solid malignancies by means of primary debulking and secondary cleansing process.
Collapse
Affiliation(s)
- Yun Ji
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, P.R.China;Department of Natural Medicinal Chemistry and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, P.R.China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Liu X, Jiang C, Li Y, Liu W, Yao N, Gao M, Ji Y, Huang D, Yin Z, Sun Z, Ni Y, Zhang J. Evaluation of hypericin: effect of aggregation on targeting biodistribution. J Pharm Sci 2014; 104:215-22. [PMID: 25395358 DOI: 10.1002/jps.24230] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 09/30/2014] [Accepted: 10/08/2014] [Indexed: 12/20/2022]
Abstract
Hypericin (Hy) has shown great promise as a necrosis-avid agent in cancer imaging and therapy. Given the highly hydrophobic and π-conjugated planarity characteristics, Hy tends to form aggregates. To investigate the effect of aggregation on targeting biodistribution, nonaggregated formulation (Non-Ag), aggregated formulation with overconcentrated Hy in dimethyl sulfoxide (Ag-DMSO) solution, and aggregated formulation in water solution (Ag-water) were selected by fluorescence measurement. They were labeled with ¹³¹I and evaluated for the necrosis affinity in rat model of reperfused hepatic infarction by gamma counting and autoradiography. The radioactivity ratio of necrotic liver/normal liver was 17.1, 7.9, and 6.4 for Non-Ag, Ag-DMSO, and Ag-water, respectively. The accumulation of two aggregated formulations (Ag-DMSO and Ag-water) in organs of mononuclear phagocyte system (MPS) was 2.62 ± 0.22 and 3.96 ± 0.30 %ID/g in the lung, and 1.44 ± 0.29 and 1.51 ± 0.23 %ID/g in the spleen, respectively. The biodistribution detected by autoradiography showed the same trend as by gamma counting. In conclusion, the Non-Ag showed better targeting biodistribution and less accumulation in MPS organs than aggregated formulations of Hy. The two aggregated formulations showed significantly lower and higher accumulation in targeting organ and MPS organs, respectively.
Collapse
Affiliation(s)
- Xuejiao Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210023, People's Republic of China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, 210028, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bryan RA, Jiang Z, Jandl T, Strauss J, Koba W, Onyedika C, Morgenstern A, Bruchertseifer F, Epstein AL, Dadachova E. Treatment of experimental pancreatic cancer with 213-Bismuth-labeled chimeric antibody to single-strand DNA. Expert Rev Anticancer Ther 2014; 14:1243-9. [PMID: 25156106 DOI: 10.1586/14737140.2014.952285] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Novel approaches to treatment of pancreatic cancer (PCa) are urgently needed. A chimeric monoclonal antibody (mAb) chTNT3 binds to single-strand DNA (ssDNA) and RNA released from the non-viable cells in fast growing tumors. Here the authors investigated whether radioimmunotherapy (RIT) using chTNT3 mAb radiolabeled with 213-Bismuth ((213)Bi) could be effective in treatment of experimental PCa. METHODS Two human PCa cell lines, Panc1 and MiaPaCa-2, were used for in vitro experiments. The xenografts in mice were established using MiaPaCa-2 cells. Therapy compared (213)Bi-chTNT3 (700 μCi) to gemcitabine or cisplatin, untreated controls and 'cold' chTNT3. RESULTS RIT abrogated the tumors growth while tumors in control groups grew aggressively. Chemotherapy was less effective than RIT and toxic to mice while RIT did not have any side effects. CONCLUSIONS RIT with (213)Bi-chTNT3 was safe and effective in the treatment of experimental PCa in comparison with chemotherapy. This makes α-RIT targeting ssDNA a promising modality for further development.
Collapse
Affiliation(s)
- Ruth A Bryan
- Department of Radiology, Albert Einstein College of Medicine, 1695A Eastchester Rd. Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zheng SG, Xu HX, Guo LH, Liu LN, Lu F. The safety and treatment response of combination therapy of radioimmunotherapy and radiofrequency ablation for solid tumor: a study in vivo. PLoS One 2014; 9:e96539. [PMID: 24787957 PMCID: PMC4008584 DOI: 10.1371/journal.pone.0096539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 04/08/2014] [Indexed: 12/18/2022] Open
Abstract
OBJECTION To investigate the safety and treatment response of radioimmunotherapy (RIT) in combination with radiofrequency ablation (RFA) for the treatment of VX2 tumor on rabbit. MATERIALS AND METHODS A total of 36 rabbits bearing VX2 tumor on the thigh were randomly assigned into 3 groups (group I: 1-2 cm; group II: 2-3 cm; group III: 3-4 cm) and 4 subgroups (A: as control, just puncture the tumor using the RFA electrode without power output; B: RFA alone; C: 131I-chTNT intratumoral injection alone; D: RFA+131I-chTNT intratumoral injection 3 days later). The variation of blood assay, weight and survival among different groups and subgroups were used to assess the treatment safety. Ultrasound (US) was used to monitor and assess the tumor response after treatment. RESULTS According to the results of the weight and the blood assay among different groups, subgroups, and at two time points (one day before and the 16th day after treatment), no damages to the liver, kidney function and myelosuppression resulting from the treatment were found. No significant differences in survivals among the four subgroups (p = 0.087) were found. In addition, 131I-chTNT did not show significant inhibition effect on VX2 tumor progression according to US measurements. CONCLUSION 131I-chTNT intratumoral injection alone or in combination with RFA is relatively safe for rabbit without significant toxicity and shows no significant effect on the survival. The treatment response is not as satisfactory as anticipated.
Collapse
Affiliation(s)
- Shu-Guang Zheng
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
- Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui-Xiong Xu
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
- Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Le-Hang Guo
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Lin-Na Liu
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Feng Lu
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, China
| |
Collapse
|
36
|
Parseghian MH, Mechetner E, Osidak MS, Domogatskii SP. Application of monoclonal antibodies for the diagnostic and therapeutic targeting of human tumors with a necrotic component. RUSS J GEN CHEM+ 2014. [DOI: 10.1134/s1070363214020364] [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]
|
37
|
An overview of translational (radio)pharmaceutical research related to certain oncological and non-oncological applications. World J Methodol 2013; 3:45-64. [PMID: 25237623 PMCID: PMC4145570 DOI: 10.5662/wjm.v3.i4.45] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/03/2013] [Accepted: 10/18/2013] [Indexed: 02/06/2023] Open
Abstract
Translational medicine pursues the conversion of scientific discovery into human health improvement. It aims to establish strategies for diagnosis and treatment of diseases. Cancer treatment is difficult. Radio-pharmaceutical research has played an important role in multiple disciplines, particularly in translational oncology. Based on the natural phenomenon of necrosis avidity, OncoCiDia has emerged as a novel generic approach for treating solid malignancies. Under this systemic dual targeting strategy, a vascular disrupting agent first selectively causes massive tumor necrosis that is followed by iodine-131 labeled-hypericin (123I-Hyp), a necrosis-avid compound that kills the residual cancer cells by crossfire effect of beta radiation. In this review, by emphasizing the potential clinical applicability of OncoCiDia, we summarize our research activities including optimization of radioiodinated hypericin Hyp preparations and recent studies on the biodistribution, dosimetry, pharmacokinetic and, chemical and radiochemical toxicities of the preparations. Myocardial infarction is a global health problem. Although cardiac scintigraphy using radioactive perfusion tracers is used in the assessment of myocardial viability, searching for diagnostic imaging agents with authentic necrosis avidity is pursued. Therefore, a comparative study on the biological profiles of the necrosis avid 123I-Hyp and the commercially available 99mTc-Sestamibi was conducted and the results are demonstrated. Cholelithiasis or gallstone disease may cause gallbladder inflammation, infection and other severe complications. While studying the mechanisms underlying the necrosis avidity of Hyp and derivatives, their naturally occurring fluorophore property was exploited for targeting cholesterol as a main component of gallstones. The usefulness of Hyp as an optical imaging agent for cholelithiasis was studied and the results are presented. Multiple uses of automatic contrast injectors may reduce costs and save resources. However, cross-contaminations with blood-borne pathogens of infectious diseases may occur. We developed a radioactive method for safety evaluation of a new replaceable patient-delivery system. By mimicking pathogens with a radiotracer, we assessed the feasibility of using the system repeatedly without septic risks. This overview is deemed to be interesting to those involved in the related fields for translational research.
Collapse
|
38
|
Ye L, Fan J, Shi X, Tao Q, Ye D, Xian Z, Zeng X, Li Y, Feng M, Ju D. Tumor necrosis therapy antibody interleukin-2 fusion protein elicits prolonged and targeted antitumor effects in vivo. Appl Microbiol Biotechnol 2013; 98:4053-61. [DOI: 10.1007/s00253-013-5349-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/17/2013] [Accepted: 10/19/2013] [Indexed: 11/24/2022]
|
39
|
Jang JK, Khawli LA, Park R, Wu BW, Li Z, Canter D, Conti PS, Epstein AL. Cytoreductive chemotherapy improves the biodistribution of antibodies directed against tumor necrosis in murine solid tumor models. Mol Cancer Ther 2013; 12:2827-36. [PMID: 24130055 DOI: 10.1158/1535-7163.mct-13-0383] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Current strategies in cancer treatment employ combinations of different treatment modalities, which include chemotherapy, radiotherapy, immunotherapy, and surgery. Consistent with that approach, the present study demonstrates how chemotherapeutic agents can potentiate the delivery of radiolabeled, necrosis-targeting antibodies (chTNT-3, NHS76) to tumor. All chemotherapeutics in this study (5-fluorouracil, etoposide, vinblastine, paclitaxel, and doxorubicin) resulted in statistically significant increases in tumor uptake of radiolabeled antibodies and their F(ab')2 fragments compared to no pretreatment with chemotherapy. Labeled antibodies were administered at various time points following a single dose of chemotherapy in multiple tumor models, and the biodistribution of the antibodies was determined by measuring radioactivity in harvested tissues. MicroPET/CT was also done to demonstrate clinical relevancy of using chemotherapy pretreatment to increase antibody uptake. Results of biodistribution and imaging data reveal specific time frames following chemotherapy when necrosis-targeting antibodies are best delivered, either for imaging or radiotherapy. Thus, the present work offers the prospect of using cytoreductive chemotherapy to increase tumor accumulation of select therapeutic antibodies, especially when combined with other forms of immunotherapy, for the successful treatment of solid tumors.
Collapse
Affiliation(s)
- Julie K Jang
- Corresponding Author: Alan L. Epstein, Department of Pathology, University of Southern California, Keck School of Medicine, 2011 Zonal Avenue, HMR 205, Los Angeles, CA 90033.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Zheng SG, Xu HX, Lu MD, Yue DC, Xie XY, Liu GJ. Radiofrequency ablation before intratumoral injection of (131)I-chTNT improves the tumor-to-normal tissue ratio in solid VX2 tumor. Cancer Biother Radiopharm 2013; 28:725-30. [PMID: 23964639 DOI: 10.1089/cbr.2012.1418] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE This study was aimed to investigate whether the tumor necrosis induced by radiofrequency ablation (RFA) can improve the ratio of tumor-to-normal tissue (T/NT) after intratumoral injection of (131)I-chTNT. MATERIALS AND METHOD Eighteen New Zealand rabbits bearing VX2 tumor on the thigh were randomly divided into two treatment groups (control group: intratumoral injection of (131)I-chTNT alone; RFA group: RFA + intratumoral injection of (131)I-chTNT 3 days after RFA) and each group was further divided into three subgroups I, II, and III (1-2 cm, 2-3 cm, and 3-4 cm in maximum diameter, respectively), by the tumor size. SPECT was performed to evaluate the T/NT on days 1, 8, and 15 after (131)I-chTNT injection. RESULTS After treatment, all rabbits underwent the SPECT whole-body scan and the T/NT was analyzed. The results showed that T/NT in the RFA group (55.45±41.83) was significantly higher compared with the control group (7.23±5.61) (F=18.89, p=0.001). Meanwhile, a linear ascending trend was found for T/NT in the RFA group along with the follow-up time (r=0.47, p=0.01). The tumor size or the dose of (131)I-TNT injection had no significant effect on the variation of T/NT in both groups (p>0.05). CONCLUSION RFA before intratumoral injection of (131)I-chTNT can dramatically improve T/NT, demonstrating the potential application of this combination therapy.
Collapse
Affiliation(s)
- Shu-Guang Zheng
- 1 Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, China
| | | | | | | | | | | |
Collapse
|
41
|
List T, Neri D. Immunocytokines: a review of molecules in clinical development for cancer therapy. Clin Pharmacol 2013; 5:29-45. [PMID: 23990735 PMCID: PMC3753206 DOI: 10.2147/cpaa.s49231] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The concept of therapeutically enhancing the immune system’s responsiveness to tumors is
long standing. Several cytokines have been investigated in clinical trials for their therapeutic
activity in cancer patients. However, substantial side effects and unfavorable pharmacokinetic
properties have been a major drawback hampering the administration of therapeutically relevant
doses. The use of recombinant antibody–cytokine fusion proteins promises to significantly
enhance the therapeutic index of cytokines by targeting them to the site of disease. This review
aims to provide a concise and complete overview of the preclinical data and clinical results
currently available for all immunocytokines having reached clinical development.
Collapse
Affiliation(s)
- Thomas List
- Department of Chemistry and Applied Biosciences, Swiss Federal institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
42
|
Khawli LA, Hu P, Epstein AL. Targeted and Untargeted Fusion Proteins: Current Approaches to Cancer Immunotherapy. FUSION PROTEIN TECHNOLOGIES FOR BIOPHARMACEUTICALS 2013:295-314. [DOI: 10.1002/9781118354599.ch19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
43
|
Tomblyn MB, Katin MJ, Wallner PE. The New Golden Era for Radioimmunotherapy: Not Just for Lymphomas Anymore. Cancer Control 2013; 20:60-71. [DOI: 10.1177/107327481302000109] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Michael B. Tomblyn
- Department of Radiation Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | | | | |
Collapse
|
44
|
Zhan J, Han Q, Wang K. Development of antibody therapeutics for small cell lung cancer. Expert Opin Investig Drugs 2012; 22:235-44. [PMID: 23176362 DOI: 10.1517/13543784.2013.750293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Small cell lung cancer (SCLC) is one of most aggressive cancers and only modest improvements have been achieved in overall survival over last 30 years. In recent years, antibody therapeutics has been actively studied and shown promise in treatment of SCLC. AREAS COVERED A comprehensive literature search through Medline and the registry database of clinical trials (ClinicalTrials.gov) was performed to collect all relevant preclinical and clinical data. The diverse antibody therapeutics which target against different antigens including VEGE-A, CEA, IGF-1R, CD56, EpCAM, CTLA-4, gangliosides GD2 and GD3, Lewis Y and tenascin-C are now under clinical investigation for therapeutic effects in SCLC. EXPERT OPINION During the last few decades, progresses have been made in antibody therapy for SCLC, however great challenges still remain. The major reasons are the complexity of SCLC and a lack of understanding of cancer immunology. The profound studies of signaling pathways involved in carcinogenesis, proliferation, metastasis and apoptosis in SCLC are crucial for the identification of new therapeutic targets and biomarkers. Moreover, a better understanding of the interplay between cancer and the immune system is a new direction for the design of more effective antibody therapeutics.
Collapse
Affiliation(s)
- Jinbiao Zhan
- Zhejiang University School of Medicine, Department of Biochemistry, Laboratory for Gene and Antibody Engineering, Hangzhou 310058, PR China.
| | | | | |
Collapse
|
45
|
Gillessen S, Gnad-Vogt US, Gallerani E, Beck J, Sessa C, Omlin A, Mattiacci MR, Liedert B, Kramer D, Laurent J, Speiser DE, Stupp R. A phase I dose-escalation study of the immunocytokine EMD 521873 (Selectikine) in patients with advanced solid tumours. Eur J Cancer 2012; 49:35-44. [PMID: 22918078 DOI: 10.1016/j.ejca.2012.07.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 06/26/2012] [Accepted: 07/11/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND EMD 521873 (Selectikine), an immunocytokine comprising a DNA-targeting antibody, aimed at tumour necrosis, fused with a genetically modified interleukin-2 (IL-2) moiety, was investigated in this first-in-human phase I study. METHODS Patients had metastatic or locally advanced solid tumours failing previous standard therapy. Selectikine was administered as a 1-hour intravenous infusion on 3 consecutive days, every 3 weeks. A subgroup of patients also received 300 mg/m(2) cyclophosphamide on day 1 of each cycle. Escalating doses of Selectikine were investigated with the primary objective of determining the maximum tolerated dose (MTD). RESULTS Thirty-nine patients were treated with Selectikine alone at dose levels from 0.075 to 0.9 mg/kg, and nine were treated at doses of 0.45 and 0.6 mg/kg in combination with cyclophosphamide. A dose-dependent linear increase of peak serum concentrations and area under curve was found. The dose-limiting toxicity was grade 3 skin rash at the 0.9 mg/kg dose-level; the MTD was 0.6 mg/kg. Rash and flu-like symptoms were the most frequent side-effects. No severe cardiovascular side-effects (hypotension or vascular leak) were observed. At all dose-levels, transient increases in total lymphocyte, eosinophil and monocyte counts were recorded. No objective tumour responses, but long periods of disease stabilisation were observed. Transient and non-neutralising Selectikine antibodies were detected in 69% of patients. CONCLUSIONS The MTD of Selectikine with or without cyclophosphamide administered under this schedule was 0.6 mg/kg. The recommended phase II dose was 0.45-0.6 mg/kg. Selectikine had a favourable safety profile and induced biological effects typical for IL-2.
Collapse
|
46
|
|
47
|
Abstract
The use of monoclonal antibodies (mAbs) for cancer therapy has achieved considerable success in recent years. Antibody-drug conjugates are powerful new treatment options for lymphomas and solid tumours, and immunomodulatory antibodies have also recently achieved remarkable clinical success. The development of therapeutic antibodies requires a deep understanding of cancer serology, protein-engineering techniques, mechanisms of action and resistance, and the interplay between the immune system and cancer cells. This Review outlines the fundamental strategies that are required to develop antibody therapies for cancer patients through iterative approaches to target and antibody selection, extending from preclinical studies to human trials.
Collapse
Affiliation(s)
- Andrew M Scott
- Ludwig Institute for Cancer Research; University of Melbourne; and Centre for PET, Austin Hospital, Melbourne, Victoria 3084, Australia.
| | | | | |
Collapse
|
48
|
Lechner MG, Russell SM, Bass RS, Epstein AL. Chemokines, costimulatory molecules and fusion proteins for the immunotherapy of solid tumors. Immunotherapy 2012; 3:1317-40. [PMID: 22053884 DOI: 10.2217/imt.11.115] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In this article, the role of chemokines and costimulatory molecules in the immunotherapy of experimental murine solid tumors and immunotherapy used in ongoing clinical trials are presented. Chemokine networks regulate physiologic cell migration that may be disrupted to inhibit antitumor immune responses or co-opted to promote tumor growth and metastasis in cancer. Recent studies highlight the potential use of chemokines in cancer immunotherapy to improve innate and adaptive cell interactions and to recruit immune effector cells into the tumor microenvironment. Another critical component of antitumor immune responses is antigen priming and activation of effector cells. Reciprocal expression and binding of costimulatory molecules and their ligands by antigen-presenting cells and naive lymphocytes ensures robust expansion, activity and survival of tumor-specific effector cells in vivo. Immunotherapy approaches using agonist antibodies or fusion proteins of immunomodulatory molecules significantly inhibit tumor growth and boost cell-mediated immunity. To localize immune stimulation to the tumor site, a series of fusion proteins consisting of a tumor-targeting monoclonal antibody directed against tumor necrosis and chemokines or costimulatory molecules were generated and tested in tumor-bearing mice. While several of these reagents were initially shown to have therapeutic value, combination therapies with methods to delete suppressor cells had the greatest effect on tumor growth. In conclusion, a key conclusion that has emerged from these studies is that successful immunotherapy will require both advanced methods of immunostimulation and the removal of immunosuppression in the host.
Collapse
Affiliation(s)
- Melissa G Lechner
- Department of Pathology, USC Keck School of Medicine, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
49
|
Li J, Chen F, Cona MM, Feng Y, Himmelreich U, Oyen R, Verbruggen A, Ni Y. A review on various targeted anticancer therapies. Target Oncol 2012; 7:69-85. [PMID: 22350489 DOI: 10.1007/s11523-012-0212-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 01/30/2012] [Indexed: 12/11/2022]
Abstract
Translational oncology aims to translate laboratory research into new anticancer therapies. Contrary to conventional surgery, chemotherapy, and radiotherapy, targeted anticancer therapy (TAT) refers to systemic administration of drugs with particular mechanisms that specifically act on well-defined targets or biologic pathways that, when activated or inactivated, may cause regression or destruction of the malignant process, meanwhile with minimized adverse effects on healthy tissues. In this article, we intend to first give a brief review on various known TAT approaches that are deemed promising for clinical applications in the current trend of personalized medicine, and then we will introduce our newly developed approach namely small molecular sequential dual targeting theragnostic strategy as a generalized class of TAT for the management of most solid malignancies, which, after optimization, is expected to help improve overall cancer treatability and curability.
Collapse
Affiliation(s)
- Junjie Li
- Section of Radiology, Department of Diagnostic Sciences, Faculty of Medicine, University of Leuven, Herestraat 49, BE-3000, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Van de Putte M, Marysael T, Fonge H, Roskams T, Cona MM, Li J, Bormans G, Verbruggen A, Ni Y, de Witte PAM. Radiolabeled iodohypericin as tumor necrosis avid tracer: diagnostic and therapeutic potential. Int J Cancer 2012; 131:E129-37. [PMID: 22038886 DOI: 10.1002/ijc.26492] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/08/2011] [Accepted: 09/02/2011] [Indexed: 12/22/2022]
Abstract
It is estimated that 30-80% of solid tumor mass represents necrotic tissue that consists out of a significant number of dead and dying cells. The fact that these necrotic zones are restricted to dysplastic and malignant tissue and are rarely present in normal tissue makes necrosis an interesting target both for cancer diagnosis and therapy. In this study, the avidity of hypericin, [(123) I]iodohypericin and [(131) I]iodohypericin to tumor necrosis was explored for both diagnosis and therapy of experimental malignancies. The intratumoral distribution in RIF-1 tumors was investigated by means of fluorescence microscopy (hypericin) and autoradiography ([(123) I]iodohypericin). Results show high uptake of the tracers in necrosis at 24 hr, lasting for up to 72 hr p.i. Ratios of activity of [(123) I]iodohypericin in necrotic tissue over viable tumor reached up to 19.63 ± 4.66, correlating with 9.20% ID/g in necrosis. Nude mice bearing RIF-1 tumors that received three injections of 300 μCi over a 3-week treatment period showed stabilization in tumor growth for 5 days, as measured by caliper and micro-positron emission tomography using [(18) F]fluorodeoxyglucose. Based on these results, we suggest the potentials of radiolabeled hypericin (1) in diagnostic aspects including prognosis or staging assessment of bulky necrotic cancers, monitoring of treatments and therapeutic follow-up; and (2) in cancer treatment based on tumor necrosis. In conclusion, we showed that hypericin radiolabeled with iodine is a necrosis avid tracer that can be used both as a tumor diagnostic and therapeutic.
Collapse
Affiliation(s)
- Marie Van de Putte
- Laboratorium voor Farmaceutische Biologie, Faculteit Farmaceutische Wetenschappen, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|