1
|
Zaheer J, Shanmugiah J, Kim S, Kim H, Kim JS. Tumor microenvironment modulation by SERPINE1 increases radioimmunotherapy in murine model of gastric cancer. Sci Rep 2025; 15:16449. [PMID: 40355586 PMCID: PMC12069594 DOI: 10.1038/s41598-025-97983-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
An elevated extracellular matrix (ECM) and interstitial fluid pressure (IFP) in gastric cancer limits the targeting of HER2-expressing cells when radioimmunotherapy (RIT) with 64Cu-trastuzumab (64Cu-TRZ) is utilized. Here, we used Losartan (LOS) to downregulate ECM and IFP in gastric cancer mice model. In our study we treated the gastric cancer mice model with a dose of 40 mg/kg of LOS. We found that the LOS treatment increases a twofold higher Alexa-647-TRZ accumulation which significantly enhanced 64Cu-TRZ. We determined that the LOS-treated samples exhibited reduced mRNA and protein expression of SERPINE1, a gene associated with the ECM degradation. Additionally, LOS treatment resulted in the downregulated mRNA expression of the TGF-β1 and COL13A1, the genes involved in ECM deposition and an upregulated RNA expression of MMP2, a gene associated with the ECM degradation. There were no significant changes in metastatic markers of N-Cadherin and E-Cadherin. Moreover, our study demonstrates that silencing SERPINE1 increases the activity of the MMP2 and decreases COL13A1 with no effect on the N-cadherin and E-cadherin were observed. Our novel combinational therapy of using 64Cu-TRZ with LOS is attributed to the downregulation of SERPINE1 targeting ECM and IFP is highly effective for treatment of gastric cancer.
Collapse
Affiliation(s)
- Javeria Zaheer
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea
- Radiological and Medical Sciences, University of Science and Technology (UST), Seoul, 01812, Republic of Korea
| | - Joycie Shanmugiah
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea
- Radiological and Medical Sciences, University of Science and Technology (UST), Seoul, 01812, Republic of Korea
| | - Seungyoun Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea
- Radiological and Medical Sciences, University of Science and Technology (UST), Seoul, 01812, Republic of Korea
| | - Hyeongi Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea
| | - Jin Su Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea.
- Radiological and Medical Sciences, University of Science and Technology (UST), Seoul, 01812, Republic of Korea.
| |
Collapse
|
2
|
Lampropoulos I, Koutsi M, Kavousanakis ME. Modeling of chemo-radiotherapy targeting growing vascular tumors: A continuum-level approach. PLoS One 2025; 20:e0301657. [PMID: 39813216 PMCID: PMC11734981 DOI: 10.1371/journal.pone.0301657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 12/29/2024] [Indexed: 01/18/2025] Open
Abstract
The aim of this study is to demonstrate the enhanced efficiency of combined therapeutic strategies for the treatment of growing tumors, based on computational experiments of a continuous-level modeling framework. In particular, the tumor growth is simulated within a host tissue and treated as a multiphase fluid, with each cellular species considered as a distinct fluid phase. Our model integrates the impact of chemical species on tumor dynamics, and we model -through reaction-diffusion equations- the spatio-temporal evolution of oxygen, vascular endothelial growth factor (VEGF) and chemotherapeutic agents. Simulations of a growing tumor exposed to external radiation showcase the rapid impact of radiotherapy on tumor suppression, however this effect diminishes over time. To enhance the therapeutic efficiency of radiotherapy, we investigate the combination of external radiation with the anti-VEGF drug bevacizumab and the cytotoxic drug docetaxel. Our simulations demonstrate that this synergistic approach integrates the immediate effectiveness of radiation therapy with the enduring tumor-suppressive capabilities of chemotherapy.
Collapse
Affiliation(s)
- Ioannis Lampropoulos
- School of Chemical Engineering, National Technical University of Athens, Zografou, Athens, Greece
| | - Marina Koutsi
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Michail E. Kavousanakis
- School of Chemical Engineering, National Technical University of Athens, Zografou, Athens, Greece
| |
Collapse
|
3
|
Wen X, Zeng X, Cheng X, Zeng X, Liu J, Zhang Y, Li Y, Chen H, Huang J, Guo Z, Chen X, Zhang X. PD-L1-Targeted Radionuclide Therapy Combined with αPD-L1 Antibody Immunotherapy Synergistically Improves the Antitumor Effect. Mol Pharm 2022; 19:3612-3622. [PMID: 35652897 DOI: 10.1021/acs.molpharmaceut.2c00281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune checkpoint blockers (ICBs) targeting programmed death receptor 1 (PD-1) ligand 1 (PD-L1) for immunotherapy have radically reformed oncology. It is of great significance to enhance the response rate of ICB in cancer patients. Here, a radioiodinated anti-PD-L1 antibody (131I-αPD-L1) was developed for PD-L1-targeted single-photon emission computed tomography (SPECT) imaging and αPD-L1 immunotherapy. Flow cytometry and immunofluorescence staining were performed to identify PD-L1 upregulation in a time- and dose-dependent manner after being induced by 131I-αPD-L1. ImmunoSPECT imaging and biodistributions of 131I-αPD-L1 in CT26, MC38, 4T1, and B16F10 tumor models were conducted to visualize the high tumor uptake and low background signal. Compared to monotherapy alone, concurrent administration of αPD-L1 mAb and 131I-αPD-L1 revealed improved tumor control in murine tumor models. The combination of 11.1 MBq of 131I-αPD-L1 and 200 μg of αPD-L1 mAb resulted in significant tumor growth delay and prolonged survival. This radioligand synergized immunotherapy strategy holds great potential for cancer management.
Collapse
Affiliation(s)
- Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xueyuan Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xingxing Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jia Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yiren Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Haojun Chen
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Jinxiong Huang
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology and Surgery, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| |
Collapse
|
4
|
Kumar C, Sharma R, Repaka KM, Pareri AU, Dash A. Camptothecin enhances 131I-rituximab-induced G1-arrest and apoptosis in Burkitt lymphoma cells. J Cancer Res Ther 2021; 17:943-950. [PMID: 34528546 DOI: 10.4103/jcrt.jcrt_1012_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background Rituximab is a chimeric monoclonal antibody against CD20. It is an established immunotherapeutic agent for non-Hodgkin's lymphoma. Even though rituximab has been used in clinics for decades, only 50% of the patients respond to rituximab therapy. To enhance the in vitro effect of rituximab, it was labeled with Iodine-131 (131I) and combined effect of 131I-rituximab and camptothecin (CPT) was studied on a tumor cell line expressing CD20. Objective The aim is to study the magnitude of cell killing and the underlying mechanism responsible for enhancing in vitro therapeutic efficacy. Materials and Methods Rituximab was labeled with 131I by the iodogen method. Raji cells were pretreated with CPT (250 nM) for an hour followed by 131I-rituximab (0.37 and 3.7 MBq) and incubated for 24 h in a humidified atmosphere of CO2 incubator at 37°C. Subsequently, Raji cells were harvested and thoroughly washed to carry out studies of cellular toxicity, apoptosis, cell cycle, and mitogen-activated protein kinase (MAPK) pathways. Results Maximal inhibition of cell proliferation and enhancement of apoptotic cell death was observed in the cells treated with the combination of CPT and 131I-rituximab, compared to controls of CPT-treated and 131I-rituximab-treated cells. Raji cells undergo G1 arrest after 131I-rituximab treatment, which leads to apoptosis and was confirmed by the downregulation of bclxl protein. Expression of p38 was decreased while an increase in phosphorylation of p38 was observed in the combination treatment of CPT and 131I-rituximab. Conclusions It was concluded from the findings that CPT enhanced 131I-rituximab-induced apoptosis, G1 cell cycle arrest and p38 MAPK phosphorylation in Raji cells.
Collapse
Affiliation(s)
- Chandan Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
| | - Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
| | - Krishna Mohan Repaka
- Radiopharmaceutical Quality Control Program, Board of Radiation and Isotope Technology, Navi Mumbai, Maharashtra, India
| | | | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
| |
Collapse
|
5
|
Pareri AU, Koijam AS, Kumar C. Breaking the Silence of Tumor Response: Future Prospects of Targeted Radionuclide Therapy. Anticancer Agents Med Chem 2021; 22:1845-1858. [PMID: 34477531 DOI: 10.2174/1871520621666210903152354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023]
Abstract
Therapy-induced tumor resistance has always been a paramount hurdle in the clinical triumph of cancer therapy. Resistance acquired by tumor through interventions of chemotherapeutic drugs, ionizing radiation, and immunotherapy in the patientsis a severe drawback and major cause of recurrence of tumor and failure of therapeutic responses. To counter acquired resistance in tumor cells, several strategies are practiced such as chemotherapy regimens, immunotherapy, and immunoconjugates, but the outcome is very disappointing for the patients as well as clinicians. Radionuclide therapy using alpha or beta-emitting radionuclide as payload became state-of-the-art for cancer therapy. With the improvement in dosimetric studies, development of high-affinity target molecules, and design of several novel chelating agents which provide thermodynamically stable complexes in vivo, the scope of radionuclide therapy has increased by leaps and bounds. Additionally, radionuclide therapy along with the combination of chemotherapy is gaining importance in pre-clinics, which is quite encouraging. Thus, it opens an avenue for newer cancer therapy modalities where chemotherapy, radiation therapy, and immunotherapy are unable to break the silence of tumor response. This article describes, in brief, the causes of tumor resistance and discusses the potential of radionuclide therapy to enhance tumor response.
Collapse
Affiliation(s)
| | | | - Chandan Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Mumbai-400085, India
| |
Collapse
|
6
|
Ren J, Xu M, Chen J, Ding J, Wang P, Huo L, Li F, Liu Z. PET imaging facilitates antibody screening for synergistic radioimmunotherapy with a 177Lu-labeled αPD-L1 antibody. Theranostics 2021; 11:304-315. [PMID: 33391476 PMCID: PMC7681088 DOI: 10.7150/thno.45540] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: The low response rate of immunotherapy, such as anti-PD-L1/PD-1 and anti-CTLA4, has limited its application to a wider population of cancer patients. One widely accepted view is that inflammation within the tumor microenvironment is low or ineffective for inducing the sufficient infiltration and/or activation of lymphocytes. Here, a highly tumor-selective anti-PD-L1 (αPD-L1) antibody was developed through PET imaging screening, and it was radiolabeled with Lu-177 for PD-L1-targeted radioimmunotherapy (RIT) and radiation-synergized immunotherapy. Methods: A series of αPD-L1 antibodies were radiolabeled with zirconium-89 for PET imaging to screen the most suitable antibodies for RIT. Mice were divided into an immunotherapy group, a RIT group and a radiation-synergized immunotherapy group to evaluate the therapeutic effect. Alterations in the tumor microenvironment after treatment were assessed using flow cytometry and immunofluorescence microscopy. Results: Radiation-synergistic RIT can achieve a significantly better therapeutic effect than immunotherapy or RIT alone. The dosages of the radiopharmaceuticals and αPD-L1 antibodies were reduced, the infiltration of CD4+ and CD8+ T cells in the tumor microenvironment was increased, and no side effects were observed. This radiation-synergistic RIT strategy successfully showed a strong synergistic effect with αPD-L1 checkpoint blockade therapy, at least in the mouse model. Conclusions: PET imaging of 89Zr-labeled antibodies is an effective method for antibody screening. RIT with a 177Lu-labeled αPD-L1 antibody could successfully upregulate antitumor immunity in the tumor microenvironment and turn "cold" tumors "hot" for immunotherapy.
Collapse
|
7
|
Kim EH, Ko HY, Yu AR, Kim H, Zaheer J, Kang HJ, Lim YC, Cho KD, Joo HY, Kang MK, Lee JJ, Lee SS, Kang HJ, Lim SM, Kim JS. Inhibition of HIF-1α by Atorvastatin During 131I-RTX Therapy in Burkitt's Lymphoma Model. Cancers (Basel) 2020; 12:E1203. [PMID: 32403237 PMCID: PMC7281655 DOI: 10.3390/cancers12051203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDS Radioimmunotherapy (RIT) serves as a targeted therapy for non-Hodgkin lymphomas (NHL). Although HIF(Hypoxia-inducible factors)-1α is an important biomarker during radiation therapy, its role in NHL is unclear. Atorvastatin (ATV) is used as a combination drug for chemotherapy. METHODS We investigated whether ATV downregulated tumor radio-resistance and enhanced the anticancer effect of 131I-RTX (rituximab) in Raji xenograft mouse models. First, the increased uptake and enhanced therapeutic effect of 131I-RTX by ATV was confirmed using molecular imaging in Raji xenograft subcutaneous model and orthotropic model with SPECT and IVIS images. Second, we examined the profile of differentially expressed miRNAs using miRNA array. RESULTS We found that miR-346 inhibited HIF-1α/VEGF (Vascular endothelial growth factor) during ATV combination therapy with 131I-RTX. The underlying mechanism of ATV involved induction of anti-angiogenesis and radiosensitivity by downregulating HIF-1α in Raji cells. CONCLUSION Our findings suggested that combination therapy with ATV and 131I-RTX is a promising strategy for enhancing the potency of 131I-RTX therapy in poorly responding patients and those with radio-resistance.
Collapse
Affiliation(s)
- Eun-Ho Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (E.-H.K.); (H.-Y.J.)
- Department of Biochemistry, School of Medicine, Catholic University of Daegu, 33, 17-gil, Duryugongwon-ro, Nam-gu, Daegu 705-718, Korea
| | - Hae Young Ko
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - A Ram Yu
- Laboratory Animal Center, Osong Medical Innovation Foundation, Osong, Chungbuk 28159, Korea; (A.R.Y.); (M.K.K.); (J.J.L.)
| | - Hyeongi Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
| | - Javeria Zaheer
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
- Radiologcial and Medico-Oncological Sciences, University of science and technology (UST), Seoul 01812, Korea
| | - Hyun Ji Kang
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
- Radiologcial and Medico-Oncological Sciences, University of science and technology (UST), Seoul 01812, Korea
| | - Young-Cheol Lim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
| | - Kyung Deuk Cho
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
| | - Hyun-Yoo Joo
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (E.-H.K.); (H.-Y.J.)
| | - Min Kyoung Kang
- Laboratory Animal Center, Osong Medical Innovation Foundation, Osong, Chungbuk 28159, Korea; (A.R.Y.); (M.K.K.); (J.J.L.)
| | - Jae Jun Lee
- Laboratory Animal Center, Osong Medical Innovation Foundation, Osong, Chungbuk 28159, Korea; (A.R.Y.); (M.K.K.); (J.J.L.)
| | - Seung-Sook Lee
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
| | - Hye Jin Kang
- Division of Hematology/Oncology, Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
| | - Sang Moo Lim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Jin Su Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (H.Y.K.); (H.K.); (J.Z.); (H.J.K.); (Y.-C.L.); (K.D.C.); (S.M.L.)
- Radiologcial and Medico-Oncological Sciences, University of science and technology (UST), Seoul 01812, Korea
| |
Collapse
|
8
|
Zaheer J, Kim H, Lee YJ, Kim JS, Lim SM. Combination Radioimmunotherapy Strategies for Solid Tumors. Int J Mol Sci 2019; 20:ijms20225579. [PMID: 31717302 PMCID: PMC6888084 DOI: 10.3390/ijms20225579] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 10/31/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Combination radioimmunotherapy is an emerging approach for the treatment of solid tumors where radio immunotherapy alone has proven to be reasonably ineffective. Radioimmunotherapy (RIT) using monoclonal antibodies (mAbs) labeled with radionuclides is an attractive approach for cancer treatment because tumor-associated mAbs with cytotoxic radionuclides can selectively bind to tumor antigens. However, due to various limitations, mAbs cannot reach solid tumors, consequently reducing RIT efficacy. Combination RIT is a pragmatic approach through which the addition of drugs or other agents not only help mAbs to reach the targeted site but also improves its efficacy. Thus, the combination of drugs or moieties with RIT can be applied to overcome the barriers that RIT faces for solid tumors. This review covers the RIT approach, along with the mechanism of action of mAb used in RIT, limitations of solid tumors, and strategies that can be used in combination RIT to enhance the treatment regimen for solid tumors.
Collapse
Affiliation(s)
- Javeria Zaheer
- Division of RI application, Korea Institute of Radiological and Medical Sciences, (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (J.Z.); (H.K.); (Y.-J.L.); (S.M.L.)
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea
| | - Hyeongi Kim
- Division of RI application, Korea Institute of Radiological and Medical Sciences, (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (J.Z.); (H.K.); (Y.-J.L.); (S.M.L.)
| | - Yong-Jin Lee
- Division of RI application, Korea Institute of Radiological and Medical Sciences, (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (J.Z.); (H.K.); (Y.-J.L.); (S.M.L.)
| | - Jin Su Kim
- Division of RI application, Korea Institute of Radiological and Medical Sciences, (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (J.Z.); (H.K.); (Y.-J.L.); (S.M.L.)
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea
- Correspondence: ; Tel.: +82-2-970-1661
| | - Sang Moo Lim
- Division of RI application, Korea Institute of Radiological and Medical Sciences, (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea; (J.Z.); (H.K.); (Y.-J.L.); (S.M.L.)
| |
Collapse
|
9
|
Zaheer J, Kim H, Lee YJ, Lim SM, Kim JS. Comparison between Fractionated Dose and Single Dose of Cu-64 Trastuzumab Therapy in the NCI-N87 Gastric Cancer Mouse Model. Int J Mol Sci 2019; 20:ijms20194708. [PMID: 31547586 PMCID: PMC6801605 DOI: 10.3390/ijms20194708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/19/2019] [Accepted: 09/22/2019] [Indexed: 11/16/2022] Open
Abstract
For optimum radioimmunotherapy (RIT), deep penetration and uniform distribution into the tumor core is important. The solid tumor microenvironment, consisting of a highly fibrotic or desmoplastic tumor, abnormal tumor vasculature, high fluid pressure, and the absence of fluid lymphatics, limits the distribution of monoclonal antibodies mAbs to the tumor core. To investigate the optimal rationale for therapeutic mAbs administration and the microdistribution of mAbs, single and serial fractional dosage regimens of Cu-64-trastuzumab (TRZ) with paclitaxel were evaluated. Groups of nude mice were inoculated with gastric cancer cell line NCI-N87 tumor cells. When the tumor size reached 200 ± 20 mm3, the mice were divided into two groups for injection of Alexa-647-TRZ. One group (n = 5) was injected with 15 mg/kg in a single dose (SD), and the other group (n = 5) with two doses of 7.5 mg/kg (fractionated dose (FD)). In both cases, the injections were done intravenously in combination with intraperitoneal paclitaxel either as a SD of 70 mg/kg or fractionated into two doses of 40 and 30 mg/kg. Tumors were harvested, flash frozen, and sectioned (8 µm) five days after Alexa-647-TRZ injection. Rhodamine lectin (rhodamine-labeled Ricinus communis agglutinin I, 1 mg in 0.2 mL of phosphate-buffered saline (PBS)) was intravenously injected to delineate the functional vessel for a wait time of 5 min before animal euthanization. Microscopic images were acquired with an IN Cell Analyzer. The amount of TRZ that penetrated the tumor surface and the tumor vessel was calculated by area under the curve (AUC) analysis. For RIT efficacy (n = 21), Cu-64-TRZ was injected following the same dose schedule to observe tumor volume and survival ratio for 30 days. The SD and FD regimens of Alexa-647-TRZ were observed to have no significant difference in penetration of mAbs from the tumor edge and vessel, nor was the total accumulation across the whole tumor tissue significantly different. Additionally, the SD and FD regimens of Cu-64-TRZ were not proven to be significantly efficacious. Our study reveals that SD and FD in a treatment design with Cu-64-TRZ and paclitaxel shows no significant difference in therapeutic efficacy on tumor growth inhibition in vivo in mice bearing human gastric cancer xenografts overexpressing HER2 antigen.
Collapse
Affiliation(s)
- Javeria Zaheer
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Hyeongi Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Yong-Jin Lee
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Sang Moo Lim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Jin Su Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| |
Collapse
|
10
|
Measurement of Tumor Pressure and Strategies of Imaging Tumor Pressure for Radioimmunotherapy. Nucl Med Mol Imaging 2019; 53:235-241. [PMID: 31456855 PMCID: PMC6694369 DOI: 10.1007/s13139-019-00598-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/10/2019] [Accepted: 05/22/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor interstitial pressure is a fundamental feature of cancer biology. Elevation in tumor pressure affects the efficacy of cancer treatment and results in the heterogenous intratumoral distribution of drugs and macromolecules. Monoclonal antibodies (mAb) play a prominent role in cancer therapy and molecular nuclear imaging. Therapy using mAb labeled with radionuclides—also known as radioimmunotherapy (RIT)—is an effective form of cancer treatment. RIT is clinically effective for the treatment of lymphoma and other blood cancers; however, its clinical use for solid tumor was limited because their high interstitial pressure prevents mAb from penetrating into the tumor. This pressure can be decreased using anti-cancer drugs or additional external therapy. In this paper, we reviewed the intratumoral pressure using direct tumor-pressure measurement strategies, such as the wick-in-needle and pressure catheter transducer method, and indirect tumor-pressure measurement strategies via magnetic resonance.
Collapse
|
11
|
Duan C, Li Y, Yang W, Zhang C. Combination of 131I-anti-endoglin monoclonal antibody and 5-fluorouracil may be a promising combined-modality radioimmunotherapy strategy for the treatment of hepatocellular carcinoma. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2018.1475254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
Affiliation(s)
- Chongling Duan
- The Laboratory Center for Basic Medicine and Biomedical Isotope Research Center, School of Basic Medical Science, Shandong University, Jinan, P.R. China
| | - Yanxiang Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Weifei Yang
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, P.R. China
| | - Chao Zhang
- The Laboratory Center for Basic Medicine and Biomedical Isotope Research Center, School of Basic Medical Science, Shandong University, Jinan, P.R. China
| |
Collapse
|
12
|
Bourgeois M, Bailly C, Frindel M, Guerard F, Chérel M, Faivre-Chauvet A, Kraeber-Bodéré F, Bodet-Milin C. Radioimmunoconjugates for treating cancer: recent advances and current opportunities. Expert Opin Biol Ther 2017; 17:813-819. [DOI: 10.1080/14712598.2017.1322577] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Mickaël Bourgeois
- Department of Nuclear Medicine, University Hospital, 44093 Nantes, France
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Nantes, France
- Department of radiopharmacy, ARRONAX Cyclotron, Saint Herblain, France
| | - Clément Bailly
- Department of Nuclear Medicine, University Hospital, 44093 Nantes, France
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Nantes, France
| | - Mathieu Frindel
- Department of Nuclear Medicine, Institut de Cancérologie de l’Ouest (ICO) – Site Gauducheau, Saint Herblain, France
| | - François Guerard
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Nantes, France
| | - Michel Chérel
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Nantes, France
- Department of Nuclear Medicine, Institut de Cancérologie de l’Ouest (ICO) – Site Gauducheau, Saint Herblain, France
| | - Alain Faivre-Chauvet
- Department of Nuclear Medicine, University Hospital, 44093 Nantes, France
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- Department of Nuclear Medicine, University Hospital, 44093 Nantes, France
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Nantes, France
- Department of Nuclear Medicine, Institut de Cancérologie de l’Ouest (ICO) – Site Gauducheau, Saint Herblain, France
| | - Caroline Bodet-Milin
- Department of Nuclear Medicine, University Hospital, 44093 Nantes, France
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Nantes, France
- Department of Nuclear Medicine, Institut de Cancérologie de l’Ouest (ICO) – Site Gauducheau, Saint Herblain, France
| |
Collapse
|
13
|
Kumar C, Shetake N, Desai S, Kumar A, Samuel G, Pandey BN. Relevance of radiobiological concepts in radionuclide therapy of cancer. Int J Radiat Biol 2016; 92:173-86. [PMID: 26917443 DOI: 10.3109/09553002.2016.1144944] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Radionuclide therapy (RNT) is a rapidly growing area of clinical nuclear medicine, wherein radionuclides are employed to deliver cytotoxic dose of radiation to the diseased cells/tissues. During RNT, radionuclides are either directly administered or delivered through biomolecules targeting the diseased site. RNT has been clinically used for diverse range of diseases including cancer, which is the focus of the review. CONCLUSIONS The major emphasis in RNT has so far been given towards developing peptides/antibodies and other molecules to conjugate a variety of therapeutic radioisotopes for improved targeting/delivery of radiation dose to the tumor cells. Despite that, many of the RNT approaches have not achieved their desired therapeutic success probably due to poor knowledge about complex and dynamic (i) fate of radiolabeled molecules; (ii) radiation dose delivered; (iii) cellular heterogeneity in tumor mass; and (iv) cellular radiobiological response. Based on understanding gathered during recent years, it may be stated that besides the absorbed dose, the net radiobiological response of tumor/normal cells also determines the clinical response of radiotherapeutic modalities including RNT. The radiosensitivity of tumor/normal cells is governed by radiobiological phenomenon such as radiation-induced bystander effect, genomic instability, adaptive response and low dose hyper-radiosensitivity. These concepts have been well investigated in the context of external beam radiotherapy, but their clinical implications during RNT have received meagre attention. In this direction, a few studies performed using in vitro and in vivo models envisage the possibilities of exploiting the radiobiological knowledge for improved therapeutic outcome of RNT.
Collapse
Affiliation(s)
- Chandan Kumar
- a Radiopharmaceutical Chemistry Section , Bhabha Atomic Research Centre , Mumbai
| | - Neena Shetake
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai
| | - Sejal Desai
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Amit Kumar
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Grace Samuel
- c Isotope Production and Applications Division , Bhabha Atomic Research Centre , Mumbai
| | - Badri N Pandey
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| |
Collapse
|
14
|
Kim JS. Combination Radioimmunotherapy Approaches and Quantification of Immuno-PET. Nucl Med Mol Imaging 2016; 50:104-11. [PMID: 27275358 PMCID: PMC4870465 DOI: 10.1007/s13139-015-0392-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/18/2015] [Accepted: 12/23/2015] [Indexed: 11/30/2022] Open
Abstract
Monoclonal antibodies (mAbs), which play a prominent role in cancer therapy, can interact with specific antigens on cancer cells, thereby enhancing the patient's immune response via various mechanisms, or mAbs can act against cell growth factors and, thereby, arrest the proliferation of tumor cells. Radionuclide-labeled mAbs, which are used in radioimmunotherapy (RIT), are effective for cancer treatment because tumor associated-mAbs linked to cytotoxic radionuclides can selectively bind to tumor antigens and release targeted cytotoxic radiation. Immunological positron emission tomography (immuno-PET), which is the combination of PET with mAb, is an attractive option for improving tumor detection and mAb quantification. However, RIT remains a challenge because of the limited delivery of mAb into tumors. The transport and uptake of mAb into tumors is slow and heterogeneous. The tumor microenvironment contributed to the limited delivery of the mAb. During the delivery process of mAb to tumor, mechanical drug resistance such as collagen distribution or physiological drug resistance such as high intestinal pressure or absence of lymphatic vessel would be the limited factor of mAb delivery to the tumor at a potentially lethal mAb concentration. When α-emitter-labeled mAbs were used, deeper penetration of α-emitter-labeled mAb inside tumors was more important because of the short range of the α emitter. Therefore, combination therapy strategies aimed at improving mAb tumor penetration and accumulation would be beneficial for maximizing their therapeutic efficacy against solid tumors.
Collapse
Affiliation(s)
- Jin Su Kim
- />Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Gil, Gongneung-Dong, Nowon-Gu, Seoul, 01812 Korea
- />Korea Drug Development Platform using Radio-Isotope(KDePRI), Seoul, Korea
- />Radiologcial and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, Korea
| |
Collapse
|
15
|
Narayanan S, Mony U, Vijaykumar DK, Koyakutty M, Paul-Prasanth B, Menon D. Sequential release of epigallocatechin gallate and paclitaxel from PLGA-casein core/shell nanoparticles sensitizes drug-resistant breast cancer cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1399-406. [DOI: 10.1016/j.nano.2015.03.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/24/2015] [Accepted: 03/30/2015] [Indexed: 12/26/2022]
|
16
|
Lindenblatt D, Fischer E, Cohrs S, Schibli R, Grünberg J. Paclitaxel improved anti-L1CAM lutetium-177 radioimmunotherapy in an ovarian cancer xenograft model. EJNMMI Res 2014; 4:54. [PMID: 26116117 PMCID: PMC4452682 DOI: 10.1186/s13550-014-0054-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/26/2014] [Indexed: 12/18/2022] Open
Abstract
Background Today’s standard treatment of advanced-stage ovarian cancer, including surgery followed by a paclitaxel-platinum-based chemotherapy, is limited in efficacy. Recently, we could show that radioimmunotherapy (RIT) with 177Lu-labelled anti-L1 cell adhesion molecule (L1CAM) monoclonal antibody chCE7 is effective in ovarian cancer therapy. We investigated if the efficacy of anti-L1CAM RIT can be further improved by its combination with paclitaxel (PTX). Methods In vitro cell viability and cell cycle arrest of human ovarian cancer cells were assessed upon different treatment conditions. For therapy studies, nude mice (n = 8) were injected subcutaneously with IGROV1 human ovarian carcinoma cells and received a single dose of 6 MBq 177Lu-DOTA-chCE7 alone or in combination with 600 μg PTX (31.6 mg/kg). Tumour growth delay and survival were determined. To investigate whether PTX can influence the tumour uptake of the radioimmunoconjugates (RICs), a biodistribution study (n = 4) and SPECT/CT images were acquired 120 h post injections of 2 MBq 177Lu-DOTA-chCE7 alone or in combination with 600 μg PTX. Results Lu-DOTA-chCE7 in combination with PTX revealed a significantly decreased cell viability of ovarian carcinoma cells in vitro and was effective in a synergistic manner (combination index < 1). PTX increased the RIT efficacy by arresting cells in the radiosensitive G2/M phase of the cell cycle 24 h post treatment start. In vivo combination therapy including 177Lu-DOTA-chCE7 and PTX resulted in a significantly prolonged overall survival (55 days vs. 18 days/PTX and 29 days/RIT), without weight loss and/or signs of toxicity. Biodistribution studies revealed no significant difference in tumour uptakes of 177Lu-DOTA-chCE7 72 h post injection regardless of an additional PTX administration. Conclusions Combination of anti-L1CAM 177Lu-RIT with PTX is a more effective therapy resulting in a prolonged overall survival of human ovarian carcinoma-bearing nude mice compared with either monotherapy. The combination is promising for future clinical applications. Electronic supplementary material The online version of this article (doi:10.1186/s13550-014-0054-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dennis Lindenblatt
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen PSI, Switzerland,
| | | | | | | | | |
Collapse
|
17
|
Mikhail AS, Eetezadi S, Ekdawi SN, Stewart J, Allen C. Image-based analysis of the size- and time-dependent penetration of polymeric micelles in multicellular tumor spheroids and tumor xenografts. Int J Pharm 2014; 464:168-77. [DOI: 10.1016/j.ijpharm.2014.01.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/25/2013] [Accepted: 01/06/2014] [Indexed: 01/02/2023]
|
18
|
Örbom A, Eriksson SE, Elgström E, Ohlsson T, Nilsson R, Tennvall J, Strand SE. The Intratumoral Distribution of Radiolabeled 177Lu-BR96 Monoclonal Antibodies Changes in Relation to Tumor Histology over Time in a Syngeneic Rat Colon Carcinoma Model. J Nucl Med 2013; 54:1404-10. [DOI: 10.2967/jnumed.112.117028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
19
|
Callahan DJ, Liu W, Li X, Dreher MR, Hassouneh W, Kim M, Marszalek P, Chilkoti A. Triple stimulus-responsive polypeptide nanoparticles that enhance intratumoral spatial distribution. NANO LETTERS 2012; 12:2165-70. [PMID: 22417133 PMCID: PMC3474318 DOI: 10.1021/nl300630c] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
To address the limited tumor penetration of nanoparticle drug delivery vehicles, we report the first pH-responsive polypeptide micelle that dissociates at the low extracellular pH of solid tumors. This histidine-rich elastin-like polypeptide block copolymer self-assembles at 37 °C into spherical micelles that are stabilized by Zn(2+) and are disrupted as the pH drops from 7.4 to 6.4. These pH-sensitive micelles demonstrate better in vivo penetration and distribution in tumors than a pH-insensitive control.
Collapse
Affiliation(s)
- Daniel J. Callahan
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham NC, 27708
- Center for Biologically Inspired Materials and Material Systems, Duke University, Box 90300, Durham, NC, 27708
| | - Wenge Liu
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham NC, 27708
| | - Xinghai Li
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham NC, 27708
| | - Matthew R. Dreher
- Center for Interventional Oncology, Clinical Center, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892
| | - Wafa Hassouneh
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham NC, 27708
- Center for Biologically Inspired Materials and Material Systems, Duke University, Box 90300, Durham, NC, 27708
| | - Minkyu Kim
- Center for Biologically Inspired Materials and Material Systems, Duke University, Box 90300, Durham, NC, 27708
- Department of Mechanical Engineering and Materials Science, Duke University, Box 90300, Durham, NC, 27708
| | - Piotr Marszalek
- Center for Biologically Inspired Materials and Material Systems, Duke University, Box 90300, Durham, NC, 27708
- Department of Mechanical Engineering and Materials Science, Duke University, Box 90300, Durham, NC, 27708
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham NC, 27708
- Center for Biologically Inspired Materials and Material Systems, Duke University, Box 90300, Durham, NC, 27708
| |
Collapse
|