51
|
Niu YQ, Liu JH, Aymonier C, Fermani S, Kralj D, Falini G, Zhou CH. Calcium carbonate: controlled synthesis, surface functionalization, and nanostructured materials. Chem Soc Rev 2022; 51:7883-7943. [PMID: 35993776 DOI: 10.1039/d1cs00519g] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Calcium carbonate (CaCO3) is an important inorganic mineral in biological and geological systems. Traditionally, it is widely used in plastics, papermaking, ink, building materials, textiles, cosmetics, and food. Over the last decade, there has been rapid development in the controlled synthesis and surface modification of CaCO3, the stabilization of amorphous CaCO3 (ACC), and CaCO3-based nanostructured materials. In this review, the controlled synthesis of CaCO3 is first examined, including Ca2+-CO32- systems, solid-liquid-gas carbonation, water-in-oil reverse emulsions, and biomineralization. Advancing insights into the nucleation and crystallization of CaCO3 have led to the development of efficient routes towards the controlled synthesis of CaCO3 with specific sizes, morphologies, and polymorphs. Recently-developed surface modification methods of CaCO3 include organic and inorganic modifications, as well as intensified surface reactions. The resultant CaCO3 can then be further engineered via template-induced biomineralization and layer-by-layer assembly into porous, hollow, or core-shell organic-inorganic nanocomposites. The introduction of CaCO3 into nanostructured materials has led to a significant improvement in the mechanical, optical, magnetic, and catalytic properties of such materials, with the resultant CaCO3-based nanostructured materials showing great potential for use in biomaterials and biomedicine, environmental remediation, and energy production and storage. The influences that the preparation conditions and additives have on ACC preparation and stabilization are also discussed. Studies indicate that ACC can be used to construct environmentally-friendly hybrid films, supramolecular hydrogels, and drug vehicles. Finally, the existing challenges and future directions of the controlled synthesis and functionalization of CaCO3 and its expanding applications are highlighted.
Collapse
Affiliation(s)
- Yu-Qin Niu
- Research Group for Advanced Materials & Sustainable Catalysis (AMSC), State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310032, China. .,Qing Yang Institute for Industrial Minerals, You Hua, Qing Yang, Chi Zhou 242804, China
| | - Jia-Hui Liu
- Research Group for Advanced Materials & Sustainable Catalysis (AMSC), State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310032, China. .,Qing Yang Institute for Industrial Minerals, You Hua, Qing Yang, Chi Zhou 242804, China
| | - Cyril Aymonier
- Univ Bordeaux, ICMCB, Bordeaux INP, UMR 5026, CNRS, F-33600 Pessac, France
| | - Simona Fermani
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, I-40126 Bologna, Italy. .,Interdepartmental Centre for Industrial Research Health Sciences & Technologies, University of Bologna, 40064 Bologna, Italy
| | - Damir Kralj
- Laboratory for Precipitation Processes, Ruđer Bošković Institute, P. O. Box 1016, HR-10001 Zagreb, Croatia
| | - Giuseppe Falini
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, I-40126 Bologna, Italy.
| | - Chun-Hui Zhou
- Research Group for Advanced Materials & Sustainable Catalysis (AMSC), State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310032, China. .,Qing Yang Institute for Industrial Minerals, You Hua, Qing Yang, Chi Zhou 242804, China
| |
Collapse
|
52
|
Gu J, Zhao G, Yu J, Xu P, Yan J, Jin Z, Chen S, Wang Y, Zhang LW, Wang Y. Injectable pH-responsive hydrogel for combinatorial chemoimmunotherapy tailored to the tumor microenvironment. J Nanobiotechnology 2022; 20:372. [PMID: 35953828 PMCID: PMC9367026 DOI: 10.1186/s12951-022-01561-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/17/2022] [Indexed: 11/10/2022] Open
Abstract
Although combination chemoimmunotherapy shows promising clinical results for cancer treatment, this approach is largely restricted by variable objective response rate and severe systemic adverse effects of immunotherapeutic antibody and chemotherapeutic drugs. Therefore, an in situ-formed therapeutic silk-chitosan composite scaffold is fabricated in this study to allow local release of the chemotherapeutic drug doxorubicin (DOX) and JQ1 (small molecular inhibitor used for the extraterminal protein BRD4 and bromodomain) with control release kinetics. DOX-JQ1@Gel contains a pH-degradable group that releases therapeutics in a weak acidic tumor microenvironment. The released DOX could directly kill tumor cells or lead to immunogenic cell death, thereby triggering the response of antitumor immunity. Meanwhile, chemotherapy-triggered antigen release and JQ1-mediated PD-L1 checkpoint blockade cumulatively contribute to trigger the response of antitumor immunity. Finally, the DOX-JQ1@Gel is locally injected to evaluate its synergistic cancer therapeutic effect, which is expected to improve objective response rate of immunotherapy and minimize systemic side effects.
Collapse
Affiliation(s)
- Jun Gu
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, 215228, China
| | - Gang Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Jiangkun Yu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Pei Xu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Jiabin Yan
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, 215228, China
| | - Zhengshuai Jin
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, 215228, China
| | - Sheng Chen
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, 215228, China.
| | - Yong Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Leshuai W Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yangyun Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
53
|
Hu T, Gong H, Xu J, Huang Y, Wu F, He Z. Nanomedicines for Overcoming Cancer Drug Resistance. Pharmaceutics 2022; 14:pharmaceutics14081606. [PMID: 36015232 PMCID: PMC9412887 DOI: 10.3390/pharmaceutics14081606] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Clinically, cancer drug resistance to chemotherapy, targeted therapy or immunotherapy remains the main impediment towards curative cancer therapy, which leads directly to treatment failure along with extended hospital stays, increased medical costs and high mortality. Therefore, increasing attention has been paid to nanotechnology-based delivery systems for overcoming drug resistance in cancer. In this respect, novel tumor-targeting nanomedicines offer fairly effective therapeutic strategies for surmounting the various limitations of chemotherapy, targeted therapy and immunotherapy, enabling more precise cancer treatment, more convenient monitoring of treatment agents, as well as surmounting cancer drug resistance, including multidrug resistance (MDR). Nanotechnology-based delivery systems, including liposomes, polymer micelles, nanoparticles (NPs), and DNA nanostructures, enable a large number of properly designed therapeutic nanomedicines. In this paper, we review the different mechanisms of cancer drug resistance to chemotherapy, targeted therapy and immunotherapy, and discuss the latest developments in nanomedicines for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Hanlin Gong
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Jiayue Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Yuan Huang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Fengbo Wu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Correspondence: (F.W.); or (Z.H.); Tel.: +86-28-85422965 (Z.H.); Fax: +86-28-85422664 (Z.H.)
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Correspondence: (F.W.); or (Z.H.); Tel.: +86-28-85422965 (Z.H.); Fax: +86-28-85422664 (Z.H.)
| |
Collapse
|
54
|
Li X, Zhang Y, Wu X, Chen J, Yang M, Ma F, Shi L. In Situ Antigen-Capturing Nanochaperone Toward Personalized Nanovaccine for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203100. [PMID: 35843873 DOI: 10.1002/smll.202203100] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Personalized cancer vaccination using nanomaterials holds great potential for cancer immunotherapy. Here, a nanochaperone (PBA-nChap) is tailored for in situ capture of tumor-associated antigens (TAAs) to improve cancer immunotherapy. The PBA-nChap is capable of i) efficiently capturing TAAs in situ; ii) protecting TAAs from degradation; iii) transporting TAAs to antigen-presenting cells and promoting cross-presentation. Intratumor injection of PBA-nChap in combination with pretreatment with photodynamic therapy (PDT) significantly enhances immune response and exhibits excellent antitumor efficacy. Moreover, nanovaccine prepared by simply co-culturing PBA-nChap with tumor cell fragments from surgery resected primary tumor in vitro synergized with immune checkpoint blockade (ICB) therapy can effectively inhibit tumor recurrence and metastasis after an operation. This work provides a promising platform for personalized cancer vaccination.
Collapse
Affiliation(s)
- Xue Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Yongxin Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Xiaohui Wu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Jiajing Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Menglin Yang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Feihe Ma
- State Key Laboratory of Separation Membranes and Membrane Processes and School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| |
Collapse
|
55
|
Zhang S, Lin A, Tao Z, Fu Y, Xiao L, Ruan G, Li Y. Microsphere‐containing hydrogel scaffolds for tissue engineering. Chem Asian J 2022; 17:e202200630. [PMID: 35909078 DOI: 10.1002/asia.202200630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/25/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Shihao Zhang
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Anqi Lin
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Ziwei Tao
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Yingying Fu
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Lan Xiao
- Queensland University of Technology Centre for Biomedical Technologies AUSTRALIA
| | | | - Yulin Li
- East China University of Science and Technology Meilong Road 130 Shanghai CHINA
| |
Collapse
|
56
|
Liu Q, Ruan H, Sheng Z, Sun X, Li S, Cui W, Li C. Nanoantidote for repression of acidosis pH promoting COVID-19 infection. VIEW 2022; 3:20220004. [PMID: 35937939 PMCID: PMC9347551 DOI: 10.1002/viw.20220004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 01/08/2023] Open
Abstract
Acidosis, such as respiratory acidosis and metabolic acidosis, can be induced by coronavirus disease 2019 (COVID-19) infection and is associated with increased mortality in critically ill COVID-19 patients. It remains unclear whether acidosis further promotes SARS-CoV-2 infection in patients, making virus removal difficult. For antacid therapy, sodium bicarbonate poses great risks caused by sodium overload, bicarbonate side effects, and hypocalcemia. Therefore, new antacid antidote is urgently needed. Our study showed that an acidosis-related pH of 6.8 increases SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2) expression on the cell membrane by regulating intracellular microfilament polymerization, promoting SARS-CoV-2 pseudovirus infection. Based on this, we synthesized polyglutamic acid-PEG materials, used complexation of calcium ions and carboxyl groups to form the core, and adopted biomineralization methods to form a calcium carbonate nanoparticles (CaCO3-NPs) nanoantidote to neutralize excess hydrogen ions (H+), and restored the pH from 6.8 to approximately 7.4 (normal blood pH). CaCO3-NPs effectively prevented the heightened SARS-CoV-2 infection efficiency due to pH 6.8. Our study reveals that acidosis-related pH promotes SARS-CoV-2 infection, which suggests the existence of a positive feedback loop in which SARS-CoV-2 infection-induced acidosis enhances SARS-CoV-2 infection. Therefore, antacid therapy for acidosis COVID-19 patients is necessary. CaCO3-NPs may become an effective antacid nanoantidote superior to sodium bicarbonate.
Collapse
Affiliation(s)
- Qidong Liu
- Department of Anesthesiology and Perioperative MedicineShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopedic Department, Tongji Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
| | - Huitong Ruan
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Zhihao Sheng
- Department of AnesthesiologyShanghai First Maternity and Infant Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
| | - Xiaoru Sun
- Department of Anesthesiology and Perioperative MedicineShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
| | - Siguang Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopedic Department, Tongji Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Cheng Li
- Department of Anesthesiology and Perioperative MedicineShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
| |
Collapse
|
57
|
Ma X, Li SJ, Liu Y, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Bioengineered nanogels for cancer immunotherapy. Chem Soc Rev 2022; 51:5136-5174. [PMID: 35666131 DOI: 10.1039/d2cs00247g] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed increasingly rapid advances in nanocarrier-based biomedicine aimed at improving treatment paradigms for cancer. Nanogels serve as multipurpose and constructed vectors formed via intramolecular cross-linking to generate drug delivery systems, which is attributed predominantly to their satisfactory biocompatibility, bio-responsiveness, high stability, and low toxicity. Recently, immunotherapy has experienced unprecedented growth and has become the preferred strategy for cancer treatment, and mainly involves the mobilisation of the immune system and an enhanced anti-tumour immunity of the tumour microenvironment. Despite the inspiring success, immunotherapeutic strategies are limited due to the low response rates and immune-related adverse events. Like other nanomedicines, nanogels are comparably limited by lower focal enrichment rates upon introduction into the organism via injection. Because nanogels are three-dimensional cross-linked aqueous materials that exhibit similar properties to natural tissues and are structurally stable, they can comfortably cope with shear forces and serum proteins in the bloodstream, and the longer circulation life increases the chance of nanogel accumulation in the tumour, conferring deep tumour penetration. The large specific surface area can reduce or eliminate off-target effects by introducing stimuli-responsive functional groups, allowing multiple physical and chemical modifications for specific purposes to improve targeting to specific immune cell subpopulations or immune organs, increasing the bioavailability of the drug, and conferring a low immune-related adverse events on nanogel therapies. The slow release upon reaching the tumour site facilitates long-term awakening of the host's immune system, ultimately achieving enhanced therapeutic effects. As an effective candidate for cancer immunotherapy, nanogel-based immunotherapy has been widely used. In this review, we mainly summarize the recent advances of nanogel-based immunotherapy to deliver immunomodulatory small molecule drugs, antibodies, genes and cytokines, to target antigen presenting cells, form cancer vaccines, and enable chimeric antigen receptor (CAR)-T cell therapy. Future challenges as well as expected and feasible prospects for clinical treatment are also highlighted.
Collapse
Affiliation(s)
- Xianbin Ma
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Shu-Jin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Yuantong Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Tian Zhang
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Peng Xue
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Yuejun Kang
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Zhigang Xu
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| |
Collapse
|
58
|
Huynh V, Tatari N, Marple A, Savage N, McKenna D, Venugopal C, Singh SK, Wylie R. Real-time evaluation of a hydrogel delivery vehicle for cancer immunotherapeutics within embedded spheroid cultures. J Control Release 2022; 348:386-396. [PMID: 35644288 DOI: 10.1016/j.jconrel.2022.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/02/2022] [Accepted: 05/22/2022] [Indexed: 11/19/2022]
Abstract
Many protein immunotherapeutics are hindered by transport barriers that prevent the obtainment of minimum effective concentrations (MECs) in solid tumors. Local delivery vehicles with tunable release (infusion) rates for immunotherapeutics are being developed to achieve local and sustained release. To expedite their discovery and translation, in vitro models can identify promising delivery vehicles and immunotherapies that benefit from sustained release by evaluating cancer spheroid killing in real-time. Using displacement affinity release (DAR) within a hydrogel, we tuned the release of a CD133 targeting dual antigen T cell engager (DATE) without the need for further DATE or hydrogel modifications, yielding an injectable vehicle that acts as a tunable infusion pump. To quantify bioactivity benefits, a 3D embedded cancer spheroid model was developed for the evaluation of sustained protein release and combination therapies on T cell mediated spheroid killing. Using automated brightfield and fluorescent microscopy, the size of red fluorescent protein (iRFP670) expressing spheroids were tracked to quantify spheroid growth or killing over time as a function of controlled delivery. We demonstrate that sustained DATE release enhanced T cell mediated killing of embedded glioblastoma spheroids at longer timepoints, killing was further enhanced with the addition of anti-PD1 antibody (αPD1). The multi-cellular embedded spheroid model with automated microscopy demonstrated the benefit of extended bispecific release on T cell mediated killing, which will expedite the identification and translation of delivery vehicles such as DAR for immunotherapeutics.
Collapse
Affiliation(s)
- Vincent Huynh
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Nazanin Tatari
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - April Marple
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Neil Savage
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Dillon McKenna
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Chitra Venugopal
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Sheila K Singh
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Ryan Wylie
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada; School of Biomedical Engineering, McMaster University, Hamilton, Ontario L8S 4M1, Canada.
| |
Collapse
|
59
|
Shang Q, Dong Y, Su Y, Leslie F, Sun M, Wang F. Local scaffold-assisted delivery of immunotherapeutic agents for improved cancer immunotherapy. Adv Drug Deliv Rev 2022; 185:114308. [PMID: 35472398 DOI: 10.1016/j.addr.2022.114308] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 12/18/2022]
Abstract
Cancer immunotherapy, which reprograms a patient's own immune system to eradicate cancer cells, has been demonstrated as a promising therapeutic strategy clinically. Immune checkpoint blockade (ICB) therapies, cytokine therapies, cancer vaccines, and chimeric antigen receptor (CAR) T cell therapies utilize immunotherapy techniques to relieve tumor immune suppression and/or activate cellular immune responses to suppress tumor growth, metastasis and recurrence. However, systemic administration is often hampered by limited drug efficacy and adverse side effects due to nonspecific tissue distribution of immunotherapeutic agents. Advancements in local scaffold-based delivery systems facilitate a controlled release of therapeutic agents into specific tissue sites through creating a local drug reservoir, providing a potent strategy to overcome previous immunotherapy limitations by improving site-specific efficacy and minimizing systemic toxicity. In this review, we summarized recent advances in local scaffold-assisted delivery of immunotherapeutic agents to reeducate the immune system, aiming to amplify anticancer efficacy and minimize immune-related adverse events. Additionally, the challenges and future perspectives of local scaffold-assisted cancer immunotherapy for clinical translation and applications are discussed.
Collapse
Affiliation(s)
- Qi Shang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Yabing Dong
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Yun Su
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21231, United States
| | - Faith Leslie
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, United States
| | - Mingjiao Sun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, United States; Department of Ophthalmology, School of Medicine, The Johns Hopkins University, Baltimore, MD 21231, United States
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
60
|
Yan J, Liu X, Wu F, Ge C, Ye H, Chen X, Wei Y, Zhou R, Duan S, Zhu R, Zheng Y, Yin L. Platelet Pharmacytes for the Hierarchical Amplification of Antitumor Immunity in Response to Self-Generated Immune Signals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109517. [PMID: 35388551 DOI: 10.1002/adma.202109517] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/02/2022] [Indexed: 05/24/2023]
Abstract
Systemic immunosuppression mediated by tumor-derived exosomes is an important cause for the resistance of immune checkpoint blockade (ICB) therapy. Herein, self-adaptive platelet (PLT) pharmacytes are engineered to mediate cascaded delivery of exosome-inhibiting siRNA and anti-PD-L1 (aPDL1) toward synergized antitumor immunity. In the pharmacytes, polycationic nanocomplexes (NCs) assembled from Rab27 siRNA (siRab) and a membrane-penetrating polypeptide are encapsulated inside the open canalicular system of PLTs, and cytotoxic T lymphocytes (CTLs)-responsive aPDL1 nanogels (NGs) are covalently backpacked on the PLT surface. Upon systemic administration, the pharmacytes enable prolonged blood circulation and active accumulation to tumors, wherein PLTs are activated to liberate siRab NCs, which efficiently transfect tumor cells, silence Rab27a, and inhibit exosome secretion. The immunosuppression is thus relieved, leading to the activation, proliferation, and tumoral infiltration of cytotoxic T cells, which trigger latent aPDL1 release. As such, the competitive aPDL1 exhaustion by PD-L1-expressing exosomes is minimized to sensitize ICB. Synergistically, siRab and aPDL1 induce strong antitumor immunological response and memory against syngeneic murine melanoma. This study reports a bioinspired mechanism to resolve the blood circulation/cell internalization contradiction of polycationic siRNA delivery systems, and renders an enlightened approach for the spatiotemporal enhancement of antitumor immunity.
Collapse
Affiliation(s)
- Jing Yan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123, China
| | - Xun Liu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Fan Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123, China
| | - Chenglong Ge
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123, China
| | - Huan Ye
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123, China
| | - Xingye Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yuansong Wei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123, China
| | - Renxiang Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123, China
| | - Shanzhou Duan
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Rongying Zhu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yiran Zheng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123, China
| |
Collapse
|
61
|
Qi J, Ding T, Liu T, Xia X, Wu S, Liu J, Chen Q, Zhang D, Zhao H. Inosine‐Based Supramolecular Hydrogel for Highly Efficient PD‐L1 Blockade Therapy via Mediating CD8
+
T Cells. ADVANCED FUNCTIONAL MATERIALS 2022. [DOI: 10.1002/adfm.202204273] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Jiajia Qi
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Tingting Ding
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Tiannan Liu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Xin Xia
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Shihong Wu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Jiang Liu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Dunfang Zhang
- Department of Biotherapy State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management Med‐X Center for Materials West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| |
Collapse
|
62
|
Li J, Zhang P, Zhou M, Liu C, Huang Y, Li L. Trauma-Responsive Scaffold Synchronizing Oncolysis Immunization and Inflammation Alleviation for Post-Operative Suppression of Cancer Metastasis. ACS NANO 2022; 16:6064-6079. [PMID: 35344338 DOI: 10.1021/acsnano.1c11562] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor surgery can create an inflammatory trauma to aggravate residual tumor "seed" to colonize pre-metastatic niches (PMNs) "soil" at secondary sites, thereby promoting post-operative metastasis. However, two-pronged strategies for post-surgical elimination of asynchronous "seeds" and "soil" at different regions are currently lacking. Here, we have designed a hydrogel that can be injected into a resection cavity, where it immediately forms a scaffold and gradually degrades responding to enriched reactive oxygen species at adjacent trauma for local delivery and on-demand release of autologous cancer cells succumbing to oncolysis (ACCO) and anti-inflammatory agent. The autologous cell source self-provides a whole array of tumor-associated antigens, and the oncolysis orchestration of a subcellular cascade confers a self-adjuvanting property, together guaranteeing high immunogenicity of the ACCO vaccine that enables specific antitumor immunization. In parallel, inflammation alleviation exerted bidirectional functions to reshape the local immune landscape and resuscitate ACCO, leading to the eradication of residual tumor "seeds" while simultaneously intercepting the "seed-soil" crosstalk to normalize distant lung leading to regression of pre-existing PMN "soil". As a result, regional and metastatic recurrence were completely thwarted. Together, this framework synchronizing oncolysis immunization and inflammation alleviation provides an effective option for post-operative suppression of metastasis.
Collapse
Affiliation(s)
- Junlin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ping Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Minglu Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chendong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
63
|
Dong J, Huang Y, Zhou Z, Sun M. Breaking Immunosuppressive Barriers by Engineered Nanoplatforms for Turning Cold Tumor to Hot. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jingwen Dong
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Ying Huang
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Zhanwei Zhou
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 P. R. China
| |
Collapse
|
64
|
Kim S, Lee J, Im S, Kim WJ. Injectable immunogel based on polymerized phenylboronic acid and mannan for cancer immunotherapy. J Control Release 2022; 345:138-146. [PMID: 35271910 DOI: 10.1016/j.jconrel.2022.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/17/2022] [Accepted: 03/03/2022] [Indexed: 11/24/2022]
Abstract
The recent development and prospects of cancer immunotherapy have led to diversification of the types of therapeutic agents used. By simultaneously administering various agents, a more effective therapeutic effect can be expected due to the synergistic effects of multiple therapeutics. In particular, if a substance with adjuvanticity and tumor antigen is delivered at the same time, enhanced cancer immunotherapy can be achieved through high cross-presentation and antigen-presenting cell (APC) maturation. To this end, we developed a polymerized phenylboronic acid (pPBA)-based immunogel for the simultaneous delivery of mannan, which has adjuvanticity and tumor antigen. The immunogel was formed by simple mixing of the polysaccharide mannan with pPBA through the formation of phenylboronic ester between the diol of mannose monomers and phenylboronic acids of pPBA. The immunogel was slowly degraded by hydrolysis to release the loaded tumor antigen. In addition, the released mannan played a key role in both APC maturation in vitro and the upregulation of cross-presentation. Finally, the pPBA-mannan immunogel exhibited a significant anticancer effect in the 4 T1 cell-inoculated mouse model, implying the potential of a codelivery system of antigens and adjuvants for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Seonil Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Junseok Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; OmniaMed Co, Ltd., Pohang 37673, Republic of Korea
| | - Sooseok Im
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; OmniaMed Co, Ltd., Pohang 37673, Republic of Korea
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; OmniaMed Co, Ltd., Pohang 37673, Republic of Korea.
| |
Collapse
|
65
|
Liu Y, Geng Y, Yue B, Lo PC, Huang J, Jin H. Injectable Hydrogel as a Unique Platform for Antitumor Therapy Targeting Immunosuppressive Tumor Microenvironment. Front Immunol 2022; 12:832942. [PMID: 35111169 PMCID: PMC8801935 DOI: 10.3389/fimmu.2021.832942] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy can boost the immune response of patients to eliminate tumor cells and suppress tumor metastasis and recurrence. However, immunotherapy resistance and the occurrence of severe immune-related adverse effects are clinical challenges that remain to be addressed. The tumor microenvironment plays a crucial role in the therapeutic efficacy of cancer immunotherapy. Injectable hydrogels have emerged as powerful drug delivery platforms offering good biocompatibility and biodegradability, minimal invasion, convenient synthesis, versatility, high drug-loading capacity, controlled drug release, and low toxicity. In this review, we summarize the application of injectable hydrogels as a unique platform for targeting the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Yushuai Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Geng
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Beilei Yue
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, SAR China
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
66
|
Chen M, Juengpanich S, Li S, Topatana W, Lu Z, Zheng Q, Cao J, Hu J, Chan E, Hou L, Chen J, Chen F, Liu Y, Jiansirisomboon S, Gu Z, Tongpeng S, Cai X. Bortezomib-Encapsulated Dual Responsive Copolymeric Nanoparticles for Gallbladder Cancer Targeted Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103895. [PMID: 35068071 PMCID: PMC8895115 DOI: 10.1002/advs.202103895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 12/13/2021] [Indexed: 05/09/2023]
Abstract
Gallbladder cancer (GBC) is a rare but the most malignant type of biliary tract tumor. It is usually diagnosed at an advanced stage and conventional treatments are unsatisfactory. As a proteasome inhibitor, bortezomib (BTZ) exhibits excellent antitumor ability in GBC. However, the long-term treatment efficacy is limited by its resistance, poor stability, and high toxicity. Herein, BTZ-encapsulated pH-responsive copolymeric nanoparticles with estrone (ES-NP(BTZ; Ce6) ) for GBC-specific targeted therapy is reported. Due to the high estrogen receptor expression in GBC, ES-NP(BTZ; Ce6) can rapidly enter the cells and accumulate near the nucleus via ES-mediated endocytosis. Under acidic tumor microenvironment (TME) and 808 nm laser irradiation, BTZ is released and ROS is generated by Ce6 to destroy the "bounce-back" response pathway proteins, such as DDI2 and p97, which can effectively inhibit proteasomes and increase apoptosis. Compared to the traditional treatment using BTZ monotherapy, ES-NP(BTZ; Ce6) can significantly impede disease progression at lower BTZ concentrations and improve its resistance. Moreover, ES-NP(BTZ; Ce6) demonstrates similar antitumor abilities in patient-derived xenograft animal models and five other types of solid tumor cells, revealing its potential as a broad-spectrum antitumor formulation.
Collapse
Affiliation(s)
- Mingyu Chen
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
- School of MedicineZhejiang UniversityHangzhou310058China
| | - Sarun Juengpanich
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
- School of MedicineZhejiang UniversityHangzhou310058China
| | - Shijie Li
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
- School of MedicineZhejiang UniversityHangzhou310058China
| | - Win Topatana
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
- School of MedicineZhejiang UniversityHangzhou310058China
| | - Ziyi Lu
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Qiang Zheng
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
| | - Jiasheng Cao
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
| | - Jiahao Hu
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
| | - Esther Chan
- School of Physical and Mathematical SciencesNanyang Technological UniversitySingapore637371Singapore
| | - Lidan Hou
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
| | - Jiang Chen
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
| | - Fang Chen
- Department of ChemistryZhejiang UniversityHangzhou310027China
| | - Yu Liu
- College of Life SciencesZhejiang UniversityHangzhou310058China
| | - Sukanda Jiansirisomboon
- School of Ceramic EngineeringInstitute of EngineeringSuranaree University of TechnologyNakhon Ratchasima30000Thailand
| | - Zhen Gu
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Suparat Tongpeng
- School of Ceramic EngineeringInstitute of EngineeringSuranaree University of TechnologyNakhon Ratchasima30000Thailand
| | - Xiujun Cai
- Department of General SurgerySir Run‐Run Shaw HospitalZhejiang UniversityHangzhou310016China
- School of MedicineZhejiang UniversityHangzhou310058China
| |
Collapse
|
67
|
Immunogenic hydrogel toolkit disturbing residual tumor “seeds” and pre-metastatic “soil” for inhibition of postoperative tumor recurrence and metastasis. Acta Pharm Sin B 2022; 12:3383-3397. [PMID: 35967277 PMCID: PMC9366231 DOI: 10.1016/j.apsb.2022.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/18/2022] [Accepted: 02/06/2022] [Indexed: 11/21/2022] Open
Abstract
Tumor recurrence and metastasis is the leading cause of mortality for postoperative breast cancer patients. However, chemotherapy intervention after surgery is often unsatisfactory, because residual microtumors are difficult to target and require frequent administration. Here, an all-in-one and once-for-all drug depot based on in situ-formed hydrogel was applied to fit the irregular surgical trauma, and enable direct contact with residual tumors and sustained drug release. Our immunological analysis after resection of orthotopic breast tumor revealed that postsurgical activation of CXCR4–CXCL12 signal exacerbated the immunosuppression and correlated with adaptive upregulation of PD-L1 in recurrent tumors. Thus, a multifunctional hydrogel toolkit was developed integrating strategies of CXCR4 inhibition, immunogenicity activation and PD-L1 blockade. Our results showed that the hydrogel toolkit not only exerted local effect on inhibiting residual tumor cell “seeds” but also resulted in abscopal effect on disturbing pre-metastatic “soil”. Furthermore, vaccine-like effect and durable antitumor memory were generated, which resisted a secondary tumor rechallenge in 100% cured mice. Strikingly, one single dose of such modality was able to eradicate recurrent tumors, completely prevent pulmonary metastasis and minimize off-target toxicity, thus providing an effective option for postoperative intervention.
Collapse
|
68
|
Pandit P, Brahmkhatri V. Structural Basis of Targeted Imaging and Therapy in Cancer Explorations with the Epigenetic Drugs. Subcell Biochem 2022; 100:503-521. [PMID: 36301504 DOI: 10.1007/978-3-031-07634-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Origin of cancer is strongly related to the unusual epigenetic regulation of gene function as indicated by recent reports. The covalent modifications to DNA or histones without affecting genomes that finally lead to phenotypical changes in cells or organisms are referred as "Epigenetics." The possibility to reprogram the epigenetics in the cancer epigenome is the most important target for cancer treatment and drug resistance. The development of epigenetic drugs holds a great potential for the current cancer therapeutic approaches. Nevertheless, targeting cancer epigenetic pathways is still exciting due to the lack of selective and effective small molecule compounds or drug molecules. Therefore, the current book chapter highlights epigenetic pathways for cancer and potential small molecule inhibitors and epidrugs targeting DNA methyltransferase, histone modification, and more new therapies with nanomaterials and imaging to improve the effectiveness of cancer treatment. The structural aspects on discovery of novel small molecules or drugs targeting epigenetic pathways in cancer exploration as promising strategies will be also discussed.
Collapse
Affiliation(s)
- Parimal Pandit
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bengaluru, Karnataka, India
| | - Varsha Brahmkhatri
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bengaluru, Karnataka, India.
| |
Collapse
|
69
|
Han W, Duan X, Ni K, Li Y, Chan C, Lin W. Co-delivery of dihydroartemisinin and pyropheophorbide-iron elicits ferroptosis to potentiate cancer immunotherapy. Biomaterials 2022; 280:121315. [PMID: 34920370 PMCID: PMC8724418 DOI: 10.1016/j.biomaterials.2021.121315] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/19/2021] [Accepted: 12/07/2021] [Indexed: 01/03/2023]
Abstract
Dihydroartemisinin (DHA) has shown cytotoxicity against various tumor cells in vitro in an iron-dependent manner, but its in vivo antitumor efficacy is compromised by its rapid degradation and clearance. Here we show the induction of ferroptosis by DHA in an immunogenic fashion and the maximization of in vivo antitumor efficacy of DHA by co-delivering a cholesterol derivative of DHA (Chol-DHA) and Pyropheophorbide-iron (Pyro-Fe) in ZnP@DHA/Pyro-Fe core-shell nanoparticles. ZnP@DHA/Pyro-Fe particles stabilize DHA against hydrolysis and prolong blood circulation of Chol-DHA and Pyro-Fe for their enhanced uptake in tumors. Co-delivery of an exogenous iron complex and DHA induces more ROS production and causes significant tumor inhibition in vivo. By increasing tumor immunogenicity, the combination of DHA and Pyro-Fe sensitizes non-immunogenic colorectal tumors to anti-PD-L1 checkpoint blockade immunotherapy. These findings suggest the potential of using nanotechnology to repurpose DHA and other drugs with excellent safety profiles for combination with immune checkpoint blockade to treat cancers.
Collapse
Affiliation(s)
- Wenbo Han
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiaopin Duan
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Kaiyuan Ni
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Youyou Li
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Christina Chan
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA; Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, IL, 60637, USA.
| |
Collapse
|
70
|
Zhao Y, Liu S, Shi Z, Zhu H, Li M, Yu Q. Pathogen infection-responsive nanoplatform targeting macrophage endoplasmic reticulum for treating life-threatening systemic infection. NANO RESEARCH 2022; 15:6243-6255. [PMID: 35382032 PMCID: PMC8972645 DOI: 10.1007/s12274-022-4211-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 05/03/2023]
Abstract
UNLABELLED Systemic infections caused by life-threatening pathogens represent one of the main factors leading to clinical death. In this study, we developed a pathogen infection-responsive and macrophage endoplasmic reticulum-targeting nanoplatform to alleviate systemic infections. The nanoplatform is composed of large-pore mesoporous silica nanoparticles (MSNs) grafted by an endoplasmic reticulum-targeting peptide, and a pathogen infection-responsive cap containing the reactive oxygen species-cleavable boronobenzyl acid linker and bovine serum albumin. The capped MSNs exhibited the capacity to high-efficiently load the antimicrobial peptide melittin, and to rapidly release the cargo triggered by H2O2 or the pathogen-macrophage interaction system, but had no obvious toxicity to macrophages. During the interaction with pathogenic Candida albicans cells and macrophages, the melittin-loading nanoplatform MSNE+MEL+TPB strongly inhibited pathogen growth, survived macrophages, and suppressed endoplasmic reticulum stress together with pro-inflammatory cytokine secretion. In a systemic infection model, the nanoplatform efficiently prevented kidney dysfunction, alleviated inflammatory symptoms, and protected the mice from death. This study developed a macrophage organelle-targeting nanoplatform for treatment of life-threatening systemic infections. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (N2 adsorption curves of the initial synthesized MSNs, FT-IR spectra of MSN, and MSNE, MEL release from the FITC-MEL-loading MSNE + TPB induced by different concentration of H2O2, viability of NIH3T3 cells, and DC2.4 cells after treatment of free MEL or the used nanoparticles, effect of MEL on C. albicans growth and macrophage death during the interaction between C. albicans and macrophages, effect of MEL and the nanoparticles on S. aureus growth and macrophage death during the interaction between S. aureus and macrophages, quantification of GRP78 (a) and activated Caspase-3, flow cytometry analysis of kidney non-macrophages with the Alexa Fluor 594 signal, survival curve of the infected mice treated by MEL or MSNE + MEL, kidney burden, blood urea levels and serum TNF-α levels in the infected mice) is available in the online version of this article at 10.1007/s12274-022-4211-z.
Collapse
Affiliation(s)
- Yan Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Shuo Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, 300071 China
- College of Environmental Science and Engineering, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, Nankai University, Tianjin, 300350 China
| | - Zhishang Shi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Hangqi Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| |
Collapse
|
71
|
Kim D, Wu Y, Shim G, Oh YK. Genome-Editing-Mediated Restructuring of Tumor Immune Microenvironment for Prevention of Metastasis. ACS NANO 2021; 15:17635-17656. [PMID: 34723493 DOI: 10.1021/acsnano.1c05420] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Modulating the tumor immune microenvironment to activate immune cells has been investigated to convert cold to hot tumors. Here, we report that metal-lipid hybrid nanoparticle (MLN)-mediated gene editing of transforming growth factor-β (TGF-β) can restructure the tumor microenvironment to an "immune activated" state for subsequent immunotherapy. MLNs with cationic lipids and elemental metallic Au inside were designed to deliver plasmid DNA encoding TGF-β single guide RNA and Cas9 protein (pC9sTgf) and to convert near-infrared light (NIR) to heat. Upon NIR irradiation, MLNs induced photothermal anticancer effects and calreticulin exposure on B16F10 cancer cells. Lipoplexes of pC9sTgf and MLN (pC9sTgf@MLN) provided gene editing of B16F10 cells and in vivo tumor tissues. In mice treated with pC9sTgf@MLNs and NIR irradiation, the tumor microenvironment showed increases in mature dendritic cells, cytotoxic T cells, and interferon-γ expression. In B16F10 tumor-bearing mice, intratumoral injection of pC9sTgf@MLNs and NIR irradiation resulted in ablation of primary tumors. Application of pC9sTgf@MLNs and NIR irradiation prevented the growth of secondarily challenged B16F10 cells at distant sites and B16F10 lung metastasis. Combined TGF-β gene editing and phototherapy is herein supported as a modality for restructuring the tumor immune microenvironment and preventing tumor recurrence.
Collapse
Affiliation(s)
- Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Gayong Shim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| |
Collapse
|
72
|
Locally Injectable Hydrogels for Tumor Immunotherapy. Gels 2021; 7:gels7040224. [PMID: 34842684 PMCID: PMC8628785 DOI: 10.3390/gels7040224] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022] Open
Abstract
Hydrogel-based local delivery systems provide a good delivery platform for cancer immunotherapy. Injectable hydrogels can directly deliver antitumor drugs to the tumor site to reduce systemic toxicity and achieve low-dose amplification immunotherapy. Therefore, it may overcome the problems of low drug utilization rate and the systemic side effects in cancer immunotherapy through systemic immune drugs, and it provides simple operation and little invasion at the same time. This study aimed to review the research progress of injectable hydrogels in tumor immunotherapy in recent years. Moreover, the local delivery of multiple drugs using injectable hydrogels in tumors is introduced to achieve single immunotherapy, combined chemo-immunotherapy, combined radio-immunotherapy, and photo-immunotherapy. Finally, the application of hydrogels in tumor immunotherapy is summarized, and the challenges and prospects for injectable hydrogels in tumor immunotherapy are proposed.
Collapse
|
73
|
Yang G, Lu SB, Li C, Chen F, Ni JS, Zha M, Li Y, Gao J, Kang T, Liu C, Li K. Type I macrophage activator photosensitizer against hypoxic tumors. Chem Sci 2021; 12:14773-14780. [PMID: 34820093 PMCID: PMC8597846 DOI: 10.1039/d1sc04124j] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Photodynamic immunotherapy has emerged as a promising strategy to treat cancer. However, the hypoxic nature of most solid tumors and notoriously immunosuppressive tumor microenvironment could greatly compromise the efficacy of photodynamic immunotherapy. To address this challenge, we rationally synthesized a type I photosensitizer of TPA-DCR nanoparticles (NPs) with aggregation-enhanced reactive oxygen species generation via an oxygen-independent pathway. We demonstrated that the free radicals produced by TPA-DCR NPs could reprogram M0 and M2 macrophages into an anti-tumor state, which is not restricted by the hypoxic conditions. The activated M1 macrophages could further induce the immunogenic cell death of cancer cells by secreting pro-inflammatory cytokines and phagocytosis. In addition, in vivo anti-tumor experiments revealed that the TPA-DCR NPs could further trigger tumor immune response by re-educating tumor-associated macrophages toward M1 phenotype and promoting T cell infiltration. Overall, this work demonstrates the design of type I organic photosensitizers and mechanistic investigation of their superior anti-tumor efficacy. The results will benefit the exploration of advanced strategies to regulate the tumor microenvironment for effective photodynamic immunotherapy against hypoxic tumors.
Collapse
Affiliation(s)
- Guang Yang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Song-Bo Lu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Chong Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Feng Chen
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Jen-Shyang Ni
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Menglei Zha
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Yaxi Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Ji Gao
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Tianyi Kang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Chao Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Kai Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| |
Collapse
|
74
|
Qin X, He L, Feng C, Fan D, Liang W, Wang Q, Fang J. Injectable Micelle-Incorporated Hydrogels for the Localized Chemo-Immunotherapy of Breast Tumors. ACS APPLIED MATERIALS & INTERFACES 2021; 13:46270-46281. [PMID: 34550685 DOI: 10.1021/acsami.1c11563] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Although immune checkpoint blockade (ICB) holds potential for the treatment of various tumors, a considerable proportion of patients show a limited response to ICB therapy due to the low immunogenicity of a variety of tumors. It has been shown that some chemotherapeutics can turn low-immunogenic tumors into immunogenic phenotypes by inducing a cascade of immune responses. In this paper, we synthesized an injectable micelle-incorporated hydrogel, which was able to sequentially release the chemotherapeutic gemcitabine (GEM) and the hydrophobic indoleamine 2, 3-dioxygenase inhibitor, d-1-methyltryptophan (d-1MT) at tumor sites. The hydrogel was formed via the thiol-ene click reaction between the thiolated chondroitin sulfate and the micelle formed by amphiphilic methacrylated Pluronic F127, in which hydrophobic d-1MT was encapsulated in the core of the F127 micelles and the hydrophilic GEM was dispersed in the hydrogel network. The successive release of chemotherapeutics and immune checkpoint inhibitors at tumor tissues will first promote the infiltration of cytotoxic T lymphocytes and subsequently induce a robust antitumor immune response, ultimately exerting a synergetic therapeutic efficacy. In a 4T1 tumor-bearing mice model, our results showed that the combination of chemotherapy and immunotherapy through the micelle-incorporated hydrogel triggered an effective antitumor immune response and inhibited tumor metastasis to the lung. Our results highlight the potential of the injectable micelle-incorporated hydrogel for the localized chemo-immunotherapy in the treatment of breast tumors.
Collapse
Affiliation(s)
- Xianyan Qin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Liming He
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Chenglan Feng
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Donghao Fan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenlang Liang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiyu Fang
- Advanced Materials Processing and Analysis and Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida 32816, United States
| |
Collapse
|
75
|
Correa S, Grosskopf AK, Lopez Hernandez H, Chan D, Yu AC, Stapleton LM, Appel EA. Translational Applications of Hydrogels. Chem Rev 2021; 121:11385-11457. [PMID: 33938724 PMCID: PMC8461619 DOI: 10.1021/acs.chemrev.0c01177] [Citation(s) in RCA: 461] [Impact Index Per Article: 115.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Advances in hydrogel technology have unlocked unique and valuable capabilities that are being applied to a diverse set of translational applications. Hydrogels perform functions relevant to a range of biomedical purposes-they can deliver drugs or cells, regenerate hard and soft tissues, adhere to wet tissues, prevent bleeding, provide contrast during imaging, protect tissues or organs during radiotherapy, and improve the biocompatibility of medical implants. These capabilities make hydrogels useful for many distinct and pressing diseases and medical conditions and even for less conventional areas such as environmental engineering. In this review, we cover the major capabilities of hydrogels, with a focus on the novel benefits of injectable hydrogels, and how they relate to translational applications in medicine and the environment. We pay close attention to how the development of contemporary hydrogels requires extensive interdisciplinary collaboration to accomplish highly specific and complex biological tasks that range from cancer immunotherapy to tissue engineering to vaccination. We complement our discussion of preclinical and clinical development of hydrogels with mechanical design considerations needed for scaling injectable hydrogel technologies for clinical application. We anticipate that readers will gain a more complete picture of the expansive possibilities for hydrogels to make practical and impactful differences across numerous fields and biomedical applications.
Collapse
Affiliation(s)
- Santiago Correa
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Abigail K. Grosskopf
- Chemical
Engineering, Stanford University, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Doreen Chan
- Chemistry, Stanford University, Stanford, California 94305, United States
| | - Anthony C. Yu
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | | | - Eric A. Appel
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
- Bioengineering, Stanford University, Stanford, California 94305, United States
- Pediatric
Endocrinology, Stanford University School
of Medicine, Stanford, California 94305, United States
- ChEM-H Institute, Stanford
University, Stanford, California 94305, United States
- Woods
Institute for the Environment, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
76
|
Qiu W, Liang M, Gao Y, Yang X, Zhang X, Zhang X, Xue P, Kang Y, Xu Z. Polyamino acid calcified nanohybrids induce immunogenic cell death for augmented chemotherapy and chemo-photodynamic synergistic therapy. Theranostics 2021; 11:9652-9666. [PMID: 34646391 PMCID: PMC8490510 DOI: 10.7150/thno.64354] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/28/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Monotherapy for cancer treatment is limited by unstable efficacy and uncontrollable toxic side effects, while the multifunctional nanoplatform with complex preparation process cannot avoid the potential toxicity of each functional component. Methods: We exploited tumor-specific activated polyamino acid calcified nanoparticles (CHC NPs) as new-type oxidative stress amplification of anticancer drugs via building a safe and biodegradable multifunctional nanoplatform. Giving priority to chemotherapy, and synergizing chemodynamic therapy (CDT) with photodynamic therapy (PDT), this strategy was to achieve enhanced chemotherapy, simultaneously inducing immunogenic cell death and inhibiting tumor cell invasion. Results: Based on amorphous calcium carbonate, pH-responsive nanocarrier was prepared with classical chemotherapeutic drug 10-hydroxycamplothecin (HCPT) and photosensitizer Chlorin e6 (Ce6) to realize multifunctional nanotheranostics. Conclusion: Inventive calcified nanohybrids, where topoisomerase inhibited by HCPT to prevent DNA synthesis, the generation of •OH induced via Fenton reaction, along with a large amount of 1O2 produced by Ce6, might be a promising strategy for anti-tumor combination therapy in clinical translation.
Collapse
Affiliation(s)
- Wei Qiu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
| | - Mengyun Liang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
| | - Yuan Gao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
| | - Xuelian Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
| | - Xingyao Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
| | - Xiaoli Zhang
- Pediatric Research Institute, Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, Guangdong 518038, P. R. China
| | - Peng Xue
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
| | - Yuejun Kang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China
- Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, P. R. China
| |
Collapse
|
77
|
Paradiso F, Serpelloni S, Francis LW, Taraballi F. Mechanical Studies of the Third Dimension in Cancer: From 2D to 3D Model. Int J Mol Sci 2021; 22:10098. [PMID: 34576261 PMCID: PMC8472581 DOI: 10.3390/ijms221810098] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/11/2022] Open
Abstract
From the development of self-aggregating, scaffold-free multicellular spheroids to the inclusion of scaffold systems, 3D models have progressively increased in complexity to better mimic native tissues. The inclusion of a third dimension in cancer models allows researchers to zoom out from a significant but limited cancer cell research approach to a wider investigation of the tumor microenvironment. This model can include multiple cell types and many elements from the extracellular matrix (ECM), which provides mechanical support for the tissue, mediates cell-microenvironment interactions, and plays a key role in cancer cell invasion. Both biochemical and biophysical signals from the extracellular space strongly influence cell fate, the epigenetic landscape, and gene expression. Specifically, a detailed mechanistic understanding of tumor cell-ECM interactions, especially during cancer invasion, is lacking. In this review, we focus on the latest achievements in the study of ECM biomechanics and mechanosensing in cancer on 3D scaffold-based and scaffold-free models, focusing on each platform's level of complexity, up-to-date mechanical tests performed, limitations, and potential for further improvements.
Collapse
Affiliation(s)
- Francesca Paradiso
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Stefano Serpelloni
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| | - Lewis W. Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| |
Collapse
|
78
|
Chen G, Chen Z, Wang Z, Obenchain R, Wen D, Li H, Wirz RE, Gu Z. Portable air-fed cold atmospheric plasma device for postsurgical cancer treatment. SCIENCE ADVANCES 2021; 7:eabg5686. [PMID: 34516919 PMCID: PMC8442862 DOI: 10.1126/sciadv.abg5686] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Surgery represents the major option for treating most solid tumors. Despite continuous improvements in surgical techniques, cancer recurrence after surgical resection remains the most common cause of treatment failure. Here, we report cold atmospheric plasma (CAP)–mediated postsurgical cancer treatment, using a portable air-fed CAP (aCAP) device. The aCAP device we developed uses the local ambient air as the source gas to generate cold plasma discharge with only joule energy level electrical input, thus providing a device that is simple and highly tunable for a wide range of biomedical applications. We demonstrate that local aCAP treatment on residual tumor cells at the surgical cavities effectively induces cancer immunogenic cell death in situ and evokes strong T cell–mediated immune responses to combat the residual tumor cells. In both 4T1 breast tumor and B16F10 melanoma models, aCAP treatment after incomplete tumor resection contributes to inhibiting tumor growth and prolonging survival.
Collapse
Affiliation(s)
- Guojun Chen
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering and the Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Zhitong Chen
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- National Innovation Center for Advanced Medical Devices, Shenzhen 518000, China
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Richard Obenchain
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Di Wen
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hongjun Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Richard E. Wirz
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Corresponding author. (Z.G.); (R.E.W.)
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Zhejiang Laboratory of Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Corresponding author. (Z.G.); (R.E.W.)
| |
Collapse
|
79
|
Zhang N, Wang D, Jing X, Yang T, Yang H, Meng L. pH/ROS Dual-Responsive Polymer-Drug-Based Nanocarriers: Click-Reaction Preparation and Fluorescence Imaging-Guided Chemotherapy and Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2021; 4:6294-6303. [PMID: 35006916 DOI: 10.1021/acsabm.1c00569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the past decades, polymer-drug conjugates of anticancer agents have gained much attention due to their enhanced aqueous solubility, improved pharmacokinetics, and better drug utilization than their conventional insoluble counterparts. Several polymer-drug conjugates have entered the third phase of clinical trials yet suffer from inherent deficiencies, including uncontrolled drug release and unclear degradation mechanisms. In this study, a pH/reactive oxygen species (ROS) dual-responsive PEG-doxorubicin (DOX) conjugate (denoted as TPD) was synthesized through acyl alkynyl-amine click reaction by PEG dipropiolate (PEGB), amine-terminated thioketal (TKL), and doxorubicin (DOX). Due to the generated ene-amine and thioketal in the backbone, the prepared amphiphilic TPD not only has a high drug loading ratio for photosensitizer chlorin e6 (Ce6) but also has the sensitivity to the acidic tumor microenvironment (TME) and ROS. Considering the complex conditions of TME, the prepared TPD@Ce6 nanoparticles (NPs) might respond to the relatively low pH and release Ce6 initially, and upon laser radiation, Ce6 produces abundant singlet oxygen (1O2) to achieve a programmable accelerated release of DOX and more Ce6 at the tumor site. In addition, the NIR fluorescence of DOX could monitor drug delivery and controlled release. The developed TPD@Ce6 NPs can realize the targeted tumor in combination therapy with negligible cytotoxicity on normal tissues.
Collapse
Affiliation(s)
- Ning Zhang
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Jiaotong University, Xi'an 710049, China
| | - Daquan Wang
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xunan Jing
- Talent Highland, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Tingting Yang
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hongbo Yang
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Jiaotong University, Xi'an 710049, China
| | - Lingjie Meng
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Jiaotong University, Xi'an 710049, China.,Instrumental Analysis Center, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
80
|
Lin J, Huang L, Xiang R, Ou H, Li X, Chen A, Liu Z. Blood compatibility evaluations of CaCO 3particles. Biomed Mater 2021; 16. [PMID: 34340221 DOI: 10.1088/1748-605x/ac19bf] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/02/2021] [Indexed: 11/12/2022]
Abstract
CaCO3particles, due to their unique properties such as biodegradation, pH-sensitivity, and porous surface, have been widely used as carrier materials for delivering drugs, genes, vaccines, and other bioactive molecules. In these applications, CaCO3particles are often administered intravenously. In this sense, the interaction between CaCO3particles and blood components plays a key role in their delivery efficacy and biosafety, though the hemocompatibility of CaCO3particles has not been evaluated until now. Deficiency in the biosafety information has delayed the clinical use of CaCO3particles in delivery systems. In this work, we investigated the biosafety of CaCO3particles, focusing on theirin vitroandin vivoeffects on key blood components (red blood cells, platelets, etc) and coagulation functions. We foundin vitrothat high concentrations of CaCO3particles can cause the aggregation and hemolysis of red blood cells, with platelet activation and coagulation prolongation.In vivo, we found that intravenously injected CaCO3particles at 50 mg kg-1significantly disturbed the red blood cells, and platelet-related blood routine indexes, but did not induce visible abnormalities in the tissue structures of the key organs. Overall, these effects may be due to the enormous adsorption capability of the porous surface of CaCO3particles. 0.1 mg ml-1of the CaCO3particles exhibit excellent compatibility for their practical applications. These results would be expected to greatly promote thein vivoapplications and clinical use of CaCO3particles in biomedicine.
Collapse
Affiliation(s)
- Jiansheng Lin
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha 410208, People's Republic of China
| | - Linghong Huang
- Department of Biomedical Engineering, Jinan University, Guangzhou 510632, People's Republic of China
| | - Rong Xiang
- Pediatrics Department of Changsha Hospital for Maternal and Child Health Care, Changsha 410007, People's Republic of China
| | - Haibo Ou
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha 410208, People's Republic of China
| | - Xinhua Li
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha 410208, People's Republic of China
| | - An Chen
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha 410208, People's Republic of China
| | - Zonghua Liu
- Department of Biomedical Engineering, Jinan University, Guangzhou 510632, People's Republic of China
| |
Collapse
|
81
|
Liu M, Cao Z, Zhang R, Chen Y, Yang X. Injectable Supramolecular Hydrogel for Locoregional Immune Checkpoint Blockade and Enhanced Cancer Chemo-Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:33874-33884. [PMID: 34275267 DOI: 10.1021/acsami.1c08285] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunotherapy has revolutionized the therapeutic modalities of cancer treatment but is severely limited by a low objective response rate and the risk of immune-related side effects. Herein, an injectable supramolecular hydrogel is developed for local delivery of the DPPA-1 peptide (a d-peptide antagonist with a high binding affinity to programmed cell death-ligand 1 (PD-L1)) and doxorubicin (DOX). On the one hand, DOX could kill tumor cells directly and also induce immunogenic cell death to provoke the antitumor immune response. On the other hand, the DPPA-1 peptide could locoregionally block the PD-1/PD-L1 pathway to potentiate T-cell-mediated immune responses and minimize side effects. Eventually, by local injection of this supramolecular hydrogel, the synergistic cancer therapeutic effect was evaluated, showing promise in improving the objective response rate of immunotherapy and minimizing its systemic side effects.
Collapse
Affiliation(s)
- Mengting Liu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Ziyang Cao
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Runlin Zhang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, P. R. China
| | - Yunhua Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, P. R. China
| | - Xianzhu Yang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
82
|
Shrestha B, Wang L, Brey EM, Uribe GR, Tang L. Smart Nanoparticles for Chemo-Based Combinational Therapy. Pharmaceutics 2021; 13:853. [PMID: 34201333 PMCID: PMC8227511 DOI: 10.3390/pharmaceutics13060853] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/27/2022] Open
Abstract
Cancer is a heterogeneous and complex disease. Traditional cancer therapy is associated with low therapeutic index, acquired resistance, and various adverse effects. With the increasing understanding of cancer biology and technology advancements, more strategies have been exploited to optimize the therapeutic outcomes. The rapid development and application of nanomedicine have motivated this progress. Combinational regimen, for instance, has become an indispensable approach for effective cancer treatment, including the combination of chemotherapeutic agents, chemo-energy, chemo-gene, chemo-small molecules, and chemo-immunology. Additionally, smart nanoplatforms that respond to external stimuli (such as light, temperature, ultrasound, and magnetic field), and/or to internal stimuli (such as changes in pH, enzymes, hypoxia, and redox) have been extensively investigated to improve precision therapy. Smart nanoplatforms for combinational therapy have demonstrated the potential to be the next generation cancer treatment regimen. This review aims to highlight the recent advances in smart combinational therapy.
Collapse
Affiliation(s)
| | | | | | - Gabriela Romero Uribe
- Department of Biomedical and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (B.S.); (L.W.); (E.M.B.)
| | - Liang Tang
- Department of Biomedical and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (B.S.); (L.W.); (E.M.B.)
| |
Collapse
|
83
|
Ye Q, Wang Y, Shen S, Xu C, Wang J. Biomaterials-Based Delivery of Therapeutic Antibodies for Cancer Therapy. Adv Healthc Mater 2021; 10:e2002139. [PMID: 33870637 DOI: 10.1002/adhm.202002139] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/05/2021] [Indexed: 12/19/2022]
Abstract
Considerable breakthroughs in the treatment of malignant tumors using antibody drugs, especially immunomodulating monoclonal antibodies (mAbs), have been made in the past decade. Despite technological advancements in antibody design and manufacture, multiple challenges face antibody-mediated cancer therapy, such as instability in vivo, poor tumor penetration, limited response rate, and undesirable off-target cytotoxicity. In recent years, an increasing number of biomaterials-based delivery systems have been reported to enhance the antitumor efficacy of antibody drugs. This review summarizes the advances and breakthroughs in integrating biomaterials with therapeutic antibodies for enhanced cancer therapy. A brief introduction to the principal mechanism of antibody-based cancer therapy is first established, and then various antibody immobilization strategies are provided. Finally, the current state-of-the-art in biomaterials-based antibody delivery systems and their applications in cancer treatment are summarized, highlighting how the delivery systems augment the therapeutic efficacy of antibody drugs. The outlook and perspective on biomaterials-based delivery of antitumor antibodies are also discussed.
Collapse
Affiliation(s)
- Qian‐Ni Ye
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
| | - Yue Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Shenzhen Bay Laboratory Shenzhen 518132 P. R. China
| | - Song Shen
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Cong‐Fei Xu
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction South China University of Technology Guangzhou 510006 P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction South China University of Technology Guangzhou 510006 P. R. China
| |
Collapse
|
84
|
Zhang Q, Zhang J, Song J, Liu Y, Ren X, Zhao Y. Protein-Based Nanomedicine for Therapeutic Benefits of Cancer. ACS NANO 2021; 15:8001-8038. [PMID: 33900074 DOI: 10.1021/acsnano.1c00476] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Proteins, a type of natural biopolymer that possess many prominent merits, have been widely utilized to engineer nanomedicine for fighting against cancer. Motivated by their ever-increasing attention in the scientific community, this review aims to provide a comprehensive showcase on the current landscape of protein-based nanomedicine for cancer therapy. On the basis of role differences of proteins in nanomedicine, protein-based nanomedicine engineered with protein therapeutics, protein carriers, enzymes, and composite proteins is introduced. The cancer therapeutic benefits of the protein-based nanomedicine are also discussed, including small-molecular therapeutics-mediated therapy, macromolecular therapeutics-mediated therapy, radiation-mediated therapy, reactive oxygen species-mediated therapy, and thermal effect-mediated therapy. Lastly, future developments and potential challenges of protein-based nanomedicine are elucidated toward clinical translation. It is believed that protein-based nanomedicine will play a vital role in the battle against cancer. We hope that this review will inspire extensive research interests from diverse disciplines to further push the developments of protein-based nanomedicine in the biomedical frontier, contributing to ever-greater medical advances.
Collapse
Affiliation(s)
- Qiuhong Zhang
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Junmin Zhang
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jun Song
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yizhen Liu
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiangzhong Ren
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| |
Collapse
|
85
|
Hu B, Gao M, Boakye-Yiadom KO, Ho W, Yu W, Xu X, Zhang XQ. An intrinsically bioactive hydrogel with on-demand drug release behaviors for diabetic wound healing. Bioact Mater 2021; 6:4592-4606. [PMID: 34095619 PMCID: PMC8141414 DOI: 10.1016/j.bioactmat.2021.04.040] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/14/2022] Open
Abstract
Prolonged, intense inflammation and excessive oxidative stress hinder diabetic wounds from healing normally, leading to disorders downstream including the postponement of re-epithelialization and extracellular matrix (ECM) formation. Herein, we report a hyaluronic acid (HA) and chitosan based hydrogel (OHA-CMC) with inherent antibacterial and hemostatic activities fabricated via Schiff base reaction. By encapsulating nanotechnologically-modified curcumin (CNP) and epidermal growth factor (EGF) into the hydrogel, OHA-CMC/CNP/EGF exhibited extraordinary antioxidant, anti-inflammatory, and migration-promoting effects in vitro. Meanwhile, OHA-CMC/CNP/EGF presented on-demand drug release in synchrony with the phases of the wound healing process. Specifically, curcumin was rapidly and constantly released to alleviate inflammation and oxidative stress in the early phase of wound healing, while a more gradual and sustained release of EGF supported late proliferation and ECM remodeling. In a diabetic full-thickness skin defect model, OHA-CMC/CNP/EGF dramatically improved wound healing with ideal re-epithelialization, granulation tissue formation, and skin appendage regeneration, highlighting the enormous therapeutic potential this biomaterial holds as a diabetic wound dressing. OHA-CMC hydrogel showed excellent inherent antibacterial and hemostatic activities. OHA-CMC co-delivered curcumin and EGF with on-demand drug release that met the repair requirements of each healing stage. OHA-CMC/CNP/EGF showed potent antioxidant and anti-inflammation activities, and was capable of promoting cell migration. OHA-CMC/CNP/EGF significantly accelerated diabetic wound healing.
Collapse
Affiliation(s)
- Bin Hu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| | - Mingzhu Gao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| | - Kofi Oti Boakye-Yiadom
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| | - William Ho
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Wei Yu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| | - Xiaoyang Xu
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Xue-Qing Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| |
Collapse
|
86
|
Ji X, Guo D, Ma J, Yin M, Yu Y, Liu C, Zhou Y, Sun J, Li Q, Chen N, Fan C, Song H. Epigenetic Remodeling Hydrogel Patches for Multidrug-Resistant Triple-Negative Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2100949. [PMID: 33792093 DOI: 10.1002/adma.202100949] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Indexed: 05/14/2023]
Abstract
The induced expansion of tumor-initiating cells (T-ICs) upon repeated exposure of tumors to chemotherapeutic drugs forms a major cause for chemoresistance and cancer metastasis. Here, a tumor-microenvironment-responsive hydrogel patch is designed to modulate the plasticity of T-ICs in triple-negative breast cancer (TNBC), which is insensitive to hormone- and HER2-targeting. The on-site formation of the hydrogel network patches tumors in a chemoresistant TNBC murine model and senses intratumoral reactive oxygen species for linker cleavage and payload release. Patch-mediated inhibition of the histone demethylase lysine-specific demethylase 1 (LSD1) epigenetically regulates the switch of T-ICs from self-renewal to differentiation, rehabilitating their chemosensitivity. Moreover, the hydrogel patch enhances tumor immunogenicity and increases T-cell infiltration via epigenetic activation of innate immunity. A single-dose of the hydrogel patch harboring LSD1 inhibitor and chemotherapy agent efficiently suppresses tumor growth, postsurgical relapse, and metastasis. The superior efficacy against multidrug resistance further reveals the broad applicability of epigenetic remodeling hydrogel patches.
Collapse
Affiliation(s)
- Xiaoyuan Ji
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Daoxia Guo
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Min Yin
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Yun Yu
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chang Liu
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanfeng Zhou
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinli Sun
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Nan Chen
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyun Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
87
|
Hou XL, Dai X, Yang J, Zhang B, Zhao DH, Li CQ, Yin ZY, Zhao YD, Liu B. Injectable polypeptide-engineered hydrogel depot for amplifying the anti-tumor immune effect induced by chemo-photothermal therapy. J Mater Chem B 2021; 8:8623-8633. [PMID: 32821893 DOI: 10.1039/d0tb01370f] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The immunosuppressive tumor microenvironment has caused great obstacles to tumor immunotherapy, especially where less tumor-associated antigens are released from tumor sites. Herein, a Ag2S QD/DOX/Bestatin@PC10ARGD genetically engineered polypeptide hydrogel PC10ARGD as a sustained-release material was developed for mammary carcinoma treatment. A near-infrared silver sulfide (Ag2S) QD as a photosensitizer was encapsulated into the hydrophobic cavity formed by the self-assembly of the polypeptide nanogel (PC10ARGD) for photothermal therapy. The water-soluble drug DOX and Bestatin were integrated into the PC10ARGD hydrogel. The photothermal effect could trigger the sustained release of the DOX, which could be applied to initiate in situ vaccination. Bestatin as an immune-adjuvant drug could amplify the body's immune function. The results of in vivo therapy tests exhibited that the Ag2S QD/DOX/Bestatin@PC10ARGD hydrogel with laser irradiation could activate anti-tumor immune effects that inhibit the growth of primary tumors and distal lung metastatic nodules. Meanwhile, a safer lower-temperature with multiple laser irradiation treatment strategy exhibited more effective tumor-killing performance (84.4% tumor inhibition rate) and promoted the penetration of immune cells into the tumor tissue. The CD8+ and CD4+ cytotoxic T cells ratio was increased by 5.3 and 10 times, respectively, thus exhibiting a good prognostic signal. The multifunctional polypeptide hydrogel as a green manufacturing and engineering material is promising to serve as a cancer vaccine for anticancer applications.
Collapse
Affiliation(s)
- Xiao-Lin Hou
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China.
| | - Xiang Dai
- Eugenic Genetics Laboratory, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, Hubei, P. R. China
| | - Jie Yang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China.
| | - Bin Zhang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China.
| | - Dong-Hui Zhao
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China.
| | - Chao-Qing Li
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China.
| | - Zhong-Yuan Yin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China.
| | - Yuan-di Zhao
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China. and Key Laboratory of Biomedical Photonics (HUST), Ministry of Education, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China
| | - Bo Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, P. R. China.
| |
Collapse
|
88
|
Abstract
INTRODUCTION Compared with traditional cancer treatment methods, tumor-targeted immunotherapy can combine targeted therapy and immunotherapy with long-lasting responses to achieve synergistic therapy, which brings hope to the complete cure of cancer. AREAS COVERED This review summarizes the newest and most up-to-date advances in tumor-targeted immunotherapy, including tumor-associated macrophages (TAMs) targeted immunotherapy, regulatory T (Treg) cells targeted immunotherapy, tumor-associated fibroblasts (TAFs) targeted immunotherapy and immune checkpoints targeted immunotherapy. EXPERT OPINION Immunotherapy can restore anti-tumor immunity in the tumor microenvironment and produce a lasting immune surveillance effect. Smart multifunctional nano delivery system can effectively combine targeted therapy with immunotherapy, which has attracted extensive attention. With the deepening of research, more and more tumor-targeted immunotherapy enter into the clinical trial phases, especially antibodies and inhibitors. Tumor-targeted immunotherapy is a promising approach for conquering cancer and bringing hope for human health.
Collapse
Affiliation(s)
- Yuelin Fang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Aihua Yu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
89
|
Li J, Fang Y, Zhang Y, Wang H, Yang Z, Ding D. Supramolecular Self-Assembly-Facilitated Aggregation of Tumor-Specific Transmembrane Receptors for Signaling Activation and Converting Immunologically Cold to Hot Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2008518. [PMID: 33734518 DOI: 10.1002/adma.202008518] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/04/2021] [Indexed: 05/05/2023]
Abstract
Supramolecular self-assembling peptide systems are attracting increasing interest in the field of cancer theranostics. Additionally, transformation of the immunologically cold tumor microenvironment into hot is of great importance for obtaining high antitumor responses for most immunotherapies. However, as far as it is known, there are nearly no studies on self-assembling peptides reported to be able to convert cold to hot tumors. Herein, a self-assembling peptide-based cancer theranostic agent (named DBT-2FFGYSA) is designed and synthesized, which can target tumor-specific transmembrane Eph receptor A2 (EphA2) receptors selectively and make the receptors form large aggregates. Such aggregate formation promotes the cross-phosphorylations among EphA2 receptors, leading to signal transduction of antitumor pathway. As a consequence, DBT-2FFGYSA can not only visualize EphA2 receptors in a fluorescence turn-on manner, but also specifically suppress the EphA2 receptor-overexpressed cancer cell proliferation and tumor growth. What is more, DBT-2FFGYSA also serves as an effective agent to convert immunologically cold tumors to hot by inducing the immunogenic cell death of EphA2 receptor-overexpressed cancer cells and recruiting massive tumor-infiltrating T cells. This study, thus, introduces a new category of agents capable of converting cold to hot tumors by pure supramolecular self-assembly without any aid of known anticancer drugs.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuan Fang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yufan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Huaimin Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang, 310024, China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| |
Collapse
|
90
|
Wen Y, Liu Y, Chen C, Chi J, Zhong L, Zhao Y, Zhao Y. Metformin loaded porous particles with bio-microenvironment responsiveness for promoting tumor immunotherapy. Biomater Sci 2021; 9:2082-2089. [PMID: 33475656 DOI: 10.1039/d0bm01931c] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PD1/PD-L1 antibody blockade-based immunotherapy has been widely recognized in the field of cancer treatment; however, only a small number of cancer patients have been shown to respond well due to the PD1/PD-L1 antibody hydrolysis induced substandard immunotherapeutic efficacy and the low immunogenicity and immunosuppressive tumor microenvironment of the patients. Here, we present a novel tumor microenvironment (TME) responsive particle delivery system with a metformin-loaded chitosan (CS) inverse opal core and a manganese dioxide (MnO2) shell (denoted as CS-metformin@MnO2 particles) for inhibiting the PD-1/PD-L1 signaling pathway and promoting tumor immunotherapy. Benefiting from the interconnected porous structure of the inverse opal, metformin can be easily extensively loaded into the CS particles. With the coating of the TME responsive MnO2 shells, the particle delivery system was imparted with an intelligent "trigger" to prevent premature leaking of the drug until it reaches the tumor tissue. We have demonstrated that CS-metformin@MnO2 particles were able to promote the apoptosis of tumor cells through immunotherapeutic means both in vivo and in vitro. Specifically, the viability of tumor cells in the drug carrier-treated group was nearly 20% less than in the untreated group. In addition, the CS particles could serve as scaffolds for the regeneration of normal tissues and promote post-surgical wound healing due to their biocompatibility and antibacterial ability. These results make CS-metformin@MnO2 particles an excellent delivery system in tumor immunotherapy and post-surgical wound healing applications.
Collapse
Affiliation(s)
- Yuanyuan Wen
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | | | | | | | | | | | | |
Collapse
|
91
|
In Situ Crosslinked Hydrogel Depot for Sustained Antibody Release Improves Immune Checkpoint Blockade Cancer Immunotherapy. NANOMATERIALS 2021; 11:nano11020471. [PMID: 33673289 PMCID: PMC7918828 DOI: 10.3390/nano11020471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/06/2021] [Accepted: 02/09/2021] [Indexed: 01/22/2023]
Abstract
The therapeutic inhibition of immune checkpoints, including cytotoxic T lymphocyte-associated protein (CTLA)-4 and programmed cell death 1 (PD-1), through the use of function blocking antibodies can confer improved clinical outcomes by invigorating CD8+ T cell-mediated anticancer immunity. However, low rates of patient responses and the high rate of immune-related adverse events remain significant challenges to broadening the benefit of this therapeutic class, termed immune checkpoint blockade (ICB). To overcome these significant limitations, controlled delivery and release strategies offer unique advantages relevant to this therapeutic class, which is typically administered systemically (e.g., intravenously), but more recently, has been shown to be highly efficacious using locoregional routes of administration. As such, in this paper, we describe an in situ crosslinked hydrogel for the sustained release of antibodies blocking CTLA-4 and PD-1 signaling from a locoregional injection proximal to the tumor site. This formulation results in efficient and durable anticancer effects with a reduced systemic toxicity compared to the bolus delivery of free antibody using an equivalent injection route. This formulation and strategy thus represent an approach for achieving the efficient and safe delivery of antibodies for ICB cancer immunotherapy.
Collapse
|
92
|
Li Y, Zhang X, Liu X, Pan W, Li N, Tang B. Intelligent stimuli-responsive nano immunomodulators for cancer immunotherapy. Chem Sci 2021; 12:3130-3145. [PMID: 34164080 PMCID: PMC8179382 DOI: 10.1039/d0sc06557a] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer immunotherapy is a revolutionary treatment method in oncology, which uses a human's own immune system against cancer. Many immunomodulators that trigger an immune response have been developed and applied in cancer immunotherapy. However, there is the risk of causing an excessive immune response upon directly injecting common immunomodulators into the human body to trigger an immune response. Therefore, the development of intelligent stimuli-responsive immunomodulators to elicit controlled immune responses in cancer immunotherapy is of great significance. Nanotechnology offers the possibility of designing smart nanomedicine to amplify the antitumor response in a safe and effective manner. Progress relating to intelligent stimuli-responsive nano immunomodulators for cancer immunotherapy is highlighted as a new creative direction in the field. Considering the clinical demand for cancer immunotherapy, we put forward some suggestions for constructing new intelligent stimuli-responsive nano immunomodulators, which will advance the development of cancer immunotherapy.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Xia Zhang
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Xiaohan Liu
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| |
Collapse
|
93
|
Yang G, Ni JS, Li Y, Zha M, Tu Y, Li K. Acceptor Engineering for Optimized ROS Generation Facilitates Reprogramming Macrophages to M1 Phenotype in Photodynamic Immunotherapy. Angew Chem Int Ed Engl 2021; 60:5386-5393. [PMID: 33236483 DOI: 10.1002/anie.202013228] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/01/2020] [Indexed: 12/17/2022]
Abstract
Reprogramming tumor-associated macrophages to an antitumor M1 phenotype by photodynamic therapy is a promising strategy to overcome the immunosuppression of tumor microenvironment for boosted immunotherapy. However, it remains unclear how the reactive oxygen species (ROS) generated from type I and II mechanisms, relate to the macrophage polarization efficacy. Herein, we design and synthesize three donor-acceptor structured photosensitizers with varied ROS-generating efficiencies. Surprisingly, we discovered that the extracellular ROS generated from type I mechanism are mainly responsible for reprogramming the macrophages from a pro-tumor type (M2) to an anti-tumor state (M1). In vivo experiments prove that the photosensitizer can trigger photodynamic immunotherapy for effective suppression of the tumor growth, while the therapeutic outcome is abolished with depleted macrophages. Overall, our strategy highlights the designing guideline of macrophage-activatable photosensitizers.
Collapse
Affiliation(s)
- Guang Yang
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Jen-Shyang Ni
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Yaxi Li
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Menglei Zha
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Yao Tu
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Kai Li
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| |
Collapse
|
94
|
Yang G, Ni J, Li Y, Zha M, Tu Y, Li K. Acceptor Engineering for Optimized ROS Generation Facilitates Reprogramming Macrophages to M1 Phenotype in Photodynamic Immunotherapy. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Guang Yang
- Department of Biomedical Engineering Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Jen‐Shyang Ni
- Department of Biomedical Engineering Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Yaxi Li
- Department of Biomedical Engineering Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Menglei Zha
- Department of Biomedical Engineering Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Yao Tu
- Department of Biomedical Engineering Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Kai Li
- Department of Biomedical Engineering Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| |
Collapse
|
95
|
Cancer Immunotherapy and Application of Nanoparticles in Cancers Immunotherapy as the Delivery of Immunotherapeutic Agents and as the Immunomodulators. Cancers (Basel) 2020; 12:cancers12123773. [PMID: 33333816 PMCID: PMC7765190 DOI: 10.3390/cancers12123773] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Cancer becomes one of the major public health problems globally and the burden is expected to be increasing. Currently, both the medical and research communities have attempted an approach to nonconventional cancer therapies that can limit damage or loss of healthy tissues and be able to fully eradicate the cancer cells. In the last few decades, cancer immunotherapy becomes an important tactic for cancer treatment. Immunotherapy of cancer must activate the host’s anti-tumor response by enhancing the innate immune system and the effector cell number, while, minimizing the host’s suppressor mechanisms. However, many immunotherapies are still limited by poor therapeutic targeting and unwanted side effects. Hence, a deeper understanding of tumor immunology and antitumor immune responses is essential for further improvement of cancer immunotherapy. In addition, effective delivery systems are required to deliver immunotherapeutic agents to the site of interest (such as: to Tumor microenvironments, to Antigen-Presenting Cells, and to the other immune systems) to enhance their efficacy by minimizing off-targeted and unwanted cytotoxicity. Abstract In the last few decades, cancer immunotherapy becomes an important tactic for cancer treatment. However, some immunotherapy shows certain limitations including poor therapeutic targeting and unwanted side effects that hinder its use in clinics. Recently, several researchers are exploring an alternative methodology to overcome the above limitations. One of the emerging tracks in this field area is nano-immunotherapy which has gone through rapid progress and revealed considerable potentials to solve limitations related to immunotherapy. Targeted and stimuli-sensitive biocompatible nanoparticles (NPs) can be synthesized to deliver immunotherapeutic agents in their native conformations to the site of interest to enhance their antitumor activity and to enhance the survival rate of cancer patients. In this review, we have discussed cancer immunotherapy and the application of NPs in cancer immunotherapy, as a carrier of immunotherapeutic agents and as a direct immunomodulator.
Collapse
|
96
|
Li Y, Zhang X, Liu X, Pan W, Li N, Tang B. Designing and Engineering of Nanocarriers for Bioapplication in Cancer Immunotherapy. ACS APPLIED BIO MATERIALS 2020; 3:8321-8337. [DOI: 10.1021/acsabm.0c01272] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Xia Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Xiaohan Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| |
Collapse
|
97
|
Yang B, Gao J, Pei Q, Xu H, Yu H. Engineering Prodrug Nanomedicine for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002365. [PMID: 33304763 PMCID: PMC7709995 DOI: 10.1002/advs.202002365] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/16/2020] [Indexed: 12/11/2022]
Abstract
Immunotherapy has shifted the clinical paradigm of cancer management. However, despite promising initial progress, immunotherapeutic approaches to cancer still suffer from relatively low response rates and the possibility of severe side effects, likely due to the low inherent immunogenicity of tumor cells, the immunosuppressive tumor microenvironment, and significant inter- and intratumoral heterogeneity. Recently, nanoformulations of prodrugs have been explored as a means to enhance cancer immunotherapy by simultaneously eliciting antitumor immune responses and reversing local immunosuppression. Prodrug nanomedicines, which integrate engineering advances in chemistry, oncoimmunology, and material science, are rationally designed through chemically modifying small molecule drugs, peptides, or antibodies to yield increased bioavailability and spatiotemporal control of drug release and activation at the target sites. Such strategies can help reduce adverse effects and enable codelivery of multiple immune modulators to yield synergistic cancer immunotherapy. In this review article, recent advances and translational challenges facing prodrug nanomedicines for cancer immunotherapy are overviewed. Last, key considerations are outlined for future efforts to advance prodrug nanomedicines aimed to improve antitumor immune responses and combat immune tolerogenic microenvironments.
Collapse
Affiliation(s)
- Bin Yang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Qing Pei
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Huixiong Xu
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
98
|
Han X, Li H, Zhou D, Chen Z, Gu Z. Local and Targeted Delivery of Immune Checkpoint Blockade Therapeutics. Acc Chem Res 2020; 53:2521-2533. [PMID: 33073988 DOI: 10.1021/acs.accounts.0c00339] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immune checkpoint blockade (ICB) therapy elicits antitumor response by inhibiting immune suppressor components, including programmed cell death protein 1 and its ligand (PD-1/PD-L1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4). Despite improved therapeutic efficacy, the clinical response rate is still unsatisfactory as revealed by the fact that only a minority of patients experience durable benefits. Additionally, "off-target" effects after systemic administration remain challenging for ICB treatment. To this end, the local and targeted delivery of ICB agents instead could be a potential solution to maximize the therapeutic outcomes while minimizing the side effects.In this Account, our recent studies directed at the development of different strategies for the local and targeted delivery of ICB agents are discussed. For example, transdermal microneedle patches loaded with anti-programmed death-1 antibody (aPD1) and anti-CTLA4 were developed to facilitate sustained release of ICB agents at the diseased sites. Triggered release could also be achieved by various stimuli within the tumor microenvironment, including low pH and abnormally expressed enzymes. Recently, the combination of an anti-programmed death-ligand 1 antibody (aPD-L1) loaded hollow-structured microneedle patch with cold atmospheric plasma (CAP) therapy was also reported. Microneedles provided microchannels to facilitate the transdermal transport of CAP and further induce immunogenic tumor cell death, which could be synergized by the local release of aPD-L1. In addition, in situ formed injectable or sprayable hydrogels were tailored to deliver immunomodulatory antibodies to the surgical bed to inhibit tumor recurrence after primary tumor resection. In paralell, inspired by the unique targeting ability of platelets toward the inflammatory sites, we engineered natural platelets decorated with aPD-L1 for targeted delivery after tumor resection to inhibit tumor recurrence. We further constructed a cell-cell combination delivery platform based on conjugates of platelets and hematopoietic stem cells (HSCs) for leukemia treatment. With the homing ability of HSCs to the bone marrow, the HSC-platelet-aPD1 assembly could effectively deliver aPD1 in an acute myeloid leukemia mouse model. Besides living cells, we also leveraged HEK293T-derived vesicles with PD1 receptors on their surfaces to disrupt the PD-1/PD-L1 immune inhibitory pathway. Moreover, the inner space of the vesicles allowed the packaging of an indoleamine 2,3-dioxygenase inhibitor, further reinforcing the therapeutic efficacy. A similar approach has also been demonstrated by genetically engineering platelets overexpressing PD1 receptor for postsurgical treatment. We hope the local and targeted ICB agent delivery methods introduced in this collection would further inspire the development of advanced drug delivery strategies to improve the efficiency of cancer treatment while alleviating side effects.
Collapse
Affiliation(s)
- Xiao Han
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Hongjun Li
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Daojia Zhou
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Zhaowei Chen
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, California 90095, United States
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
99
|
Min S, Jeon YS, Jung HJ, Khatua C, Li N, Bae G, Choi H, Hong H, Shin JE, Ko MJ, Ko HS, Jun I, Fu HE, Kim SH, Thangam R, Song JJ, Dravid VP, Kim YK, Kang H. Independent Tuning of Nano-Ligand Frequency and Sequences Regulates the Adhesion and Differentiation of Stem Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2004300. [PMID: 32820574 DOI: 10.1002/adma.202004300] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Indexed: 06/11/2023]
Abstract
The native extracellular matrix (ECM) can exhibit heterogeneous nano-sequences periodically displaying ligands to regulate complex cell-material interactions in vivo. Herein, an ECM-emulating heterogeneous barcoding system, including ligand-bearing Au and ligand-free Fe nano-segments, is developed to independently present tunable frequency and sequences in nano-segments of cell-adhesive RGD ligand. Specifically, similar exposed surface areas of total Fe and Au nano-segments are designed. Fe segments are used for substrate coupling of nanobarcodes and as ligand-free nano-segments and Au segments for ligand coating while maintaining both nanoscale (local) and macroscale (total) ligand density constant in all groups. Low nano-ligand frequency in the same sequences and terminally sequenced nano-ligands at the same frequency independently facilitate focal adhesion and mechanosensing of stem cells, which are collectively effective both in vitro and in vivo, thereby inducing stem cell differentiation. The Fe/RGD-Au nanobarcode implants exhibit high stability and no local and systemic toxicity in various tissues and organs in vivo. This work sheds novel insight into designing biomaterials with heterogeneous nano-ligand sequences at terminal sides and/or low frequency to facilitate cellular adhesion. Tuning the electrodeposition conditions can allow synthesis of unlimited combinations of ligand nano-sequences and frequencies, magnetic elements, and bioactive ligands to remotely regulate numerous host cells in vivo.
Collapse
Affiliation(s)
- Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yoo Sang Jeon
- Research Institute of Engineering and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- International Institute for Nanotechnology, Evanston, IL, 60208, USA
- NUANCE Center, Northwestern University, Evanston, IL, 60208, USA
| | - Chandra Khatua
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Na Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyojun Choi
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jeong Eun Shin
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Min Jun Ko
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Han Seok Ko
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Indong Jun
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Hong En Fu
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seung Hyun Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Vinayak P Dravid
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- International Institute for Nanotechnology, Evanston, IL, 60208, USA
- NUANCE Center, Northwestern University, Evanston, IL, 60208, USA
| | - Young Keun Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
100
|
Wang M, Zhou B, Wang L, Zhou F, Smith N, Saunders D, Towner RA, Song J, Qu J, Chen WR. Biodegradable pH-responsive amorphous calcium carbonate nanoparticles as immunoadjuvants for multimodal imaging and enhanced photoimmunotherapy. J Mater Chem B 2020; 8:8261-8270. [PMID: 32812632 PMCID: PMC7530098 DOI: 10.1039/d0tb01453b] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Development of bioresponsive theranostic nanoparticles to enhance cancer diagnostics and control cancer metastasis is highly desirable. In this study, we developed such a bioresponsive theranostic nanoparticle for synergistic photoimmunotherapy. In particular, these nanoparticles were constructed by embedding indocyanine green (ICG) into Mn2+-doped amorphous calcium carbonate (ACC(Mn)) nanoparticles, followed by loading of the Toll-like-receptor-7 agonist imiquimod (IMQ). The IMQ@ACC(Mn)-ICG/PEG nanoparticles respond to the acidic pH of the tumor microenvironment (TME) and co-deliver ICG and IMQ into the tumor. Selective phototherapy was achieved upon activation using a near-infrared laser. In the presence of IMQ and arising from phototherapeutically treated tumor cells, tumor-associated antigens give rise to a strong antitumor immune response. Reversal of the immunosuppressive TME via H+ scavenging of the tumor through ACC nanoparticles effectively inhibits tumor metastases. Moreover, the combination of ICG and Mn2+ also serves as an advanced contrast agent for cancer multimode imaging. Overall, these bioresponsive nanoparticles provide a promising approach for cancer theranostics with promising potential for future clinical translation.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Benqing Zhou
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Lu Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, USA.
| | - Feifan Zhou
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Jun Song
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, USA.
| |
Collapse
|