1
|
Wang X, Tian N, He L, Yuan Z, Han L. Emerging Applications of Pickering Emulsions in Pharmaceutical Formulations: A Comprehensive Review. Int J Nanomedicine 2025; 20:5923-5947. [PMID: 40356863 PMCID: PMC12067652 DOI: 10.2147/ijn.s514928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Over the past two decades, particle-stabilized Pickering emulsions (PEs) have emerged as a versatile platform in pharmaceutical formulations, demonstrating distinct advantages over surfactant-based systems through enhanced stability, reduced toxicity, and tunable interfacial properties. These systems exhibit unique drug delivery potential through their precisely controllable architecture, particularly in achieving spatiotemporal drug release patterns, tissue-specific targeting, and enhanced therapeutic payload encapsulation. In this review, the characteristics of PEs are first detailed, followed by an introduction to the main preparation methods and the key parameters for controlling the type, droplet size, and stability of PEs. The third section categorizes and discusses the advantages and disadvantages of various solid particles as emulsifiers. Lastly, emphasis is placed on the application of PEs in the pharmaceutical field, including functionalized designs and various administration routes to enlighten the rational design of PEs for effective drug delivery.
Collapse
Affiliation(s)
- Xingyue Wang
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Na Tian
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Zhixiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
2
|
Wang Y, Hu X, Wang J, Zhang Y, Guo P, Lv Y, Ma G, Wei W, Wang S. Versatile PLGA-Based Drug Delivery Systems for Tumor Immunotherapy. SMALL METHODS 2025; 9:e2401623. [PMID: 39924767 DOI: 10.1002/smtd.202401623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/07/2025] [Indexed: 02/11/2025]
Abstract
Tumor immunotherapy, which utilizes the immune system to fight cancer, represents a revolutionary method for cancer treatment. Poly (lactic-co-glycolic acid) (PLGA) copolymer has emerged as a promising material for tumor immunotherapy due to its biocompatibility, biodegradability, and versatility in drug delivery. By tuning the size, shape, and surface properties of PLGA-based systems, researchers have improved their ability to align with the requirements for diverse tumor immunotherapy modalities. In this review, the basic properties of the PLGA materials are first introduced and further the principal forms of the PLGA systems for controlled release are summarized and delivery applications are targeted. In addition, recent advances in the use of PLGA delivery systems are highlighted to enhance antitumor immune responses in terms of tumor vaccines, immunogenic cell death-mediated immune responses, tumor microenvironment modulation, and combination immunotherapies. Finally, prospects for the future research and clinical translation of PLGA materials are proposed.
Collapse
Affiliation(s)
- Yishu Wang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaoming Hu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghui Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, P. R. China
| | - Yu Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlin Lv
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wei Wei
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shuang Wang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
3
|
Chen X, Ding W, Jiang Y, Shi W, Qiu Y, Zhao H, Luo X. Emerging Strategies for Local Delivery of Immune Checkpoint Inhibitors to Potentiate Cancer Immunotherapy: Current Status and Future Prospects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59682-59696. [PMID: 39436983 DOI: 10.1021/acsami.4c12603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Cancer constitutes a significant threat to patients' lives worldwide. Immunotherapy, particularly immune checkpoint inhibitors (ICIs) that boost antitumor immunity by targeting immune checkpoint components, has emerged as a promising strategy for its treatment in recent years. However, the objective response rates of the ICIs are unsatisfactory. As the primary route, systemic administration of ICIs is often accompanied by immune-related adverse events. Local delivery of ICIs serves as a potential therapeutic strategy that can improve the efficacy while simultaneously reducing side effects through precise drug release at the tumor site. Initial validation of direct local application of ICIs for tumors in clinical trials has indicated reduced side effects and improved efficacy, while low bioavailability remains a challenge. Furthermore, research on various carriers, including nanoparticles, microneedles, hydrogels, combined platforms, and implantable devices for local release of ICIs has exhibited applying potential in treating murine tumors, among which combined platforms such as combined hydrogel system hold the highest promise due to their encompassment of the advantages of multiple carriers. These carriers, by incorporating ICIs and other therapeutics, could manage cancers more potently, which needs to be confirmed in clinical trials after the refinement of their biocompatibility. This review summarizes the latest research advancements regarding local administration of ICIs, with a particular focus on the carriers for local delivery as well as the combination therapies, thus providing novel insights and research guidance for scholars to enhance the efficacy of locally delivered ICIs on managing multiple cancers in the future.
Collapse
Affiliation(s)
- Xin 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, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wei 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, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuchen Jiang
- 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, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenjin Shi
- 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, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yan Qiu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, 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, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaobo Luo
- 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, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
4
|
Grindel AL, Fretellier N, Soares M, Bouzakher N, Millot Maysounabe V, Santus R, Bawa O, Wintrebert M, Couquelet C, Robert P, Emile JF, Capron C. Antitumoral effect of local injection of TLR-9 agonist emulsified in Lipiodol with systemic anti-PD-1 in a murine model of colorectal carcinoma. Front Immunol 2024; 14:1272246. [PMID: 38292484 PMCID: PMC10825566 DOI: 10.3389/fimmu.2023.1272246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Local treatments of cancer, including transarterial chemoembolization, could enhance responses to systemic immune checkpoint inhibitors such as anti-PD-1 antibodies. Lipiodol, a radiopaque oil, is widely used for transarterial chemoembolization as a tumor-targeting drug carrier and could be used in emulsion with immunomodulators. This study aimed at evaluating the antitumoral effect of intra-tumoral injection of Lipiodol-immunomodulator emulsions combined with systemic anti-PD-1 therapy in a murine model of colorectal carcinoma. Method Mice (male BALB/c) with anti-PD-1-resistant subcutaneous CT26 tumors were injected with immunomodulators, emulsified or not with Lipiodol (N=10-12/group). Results The TLR-9 agonist CpG displayed antitumor effects, while Poly I:C and QS21 did not. The Lipiodol-CpG emulsion appeared to be stable and maintained CpG within tumors for a longer time. Repeated intra-tumoral injections, combined with anti-PD-1, induced responses towards the tumor as well as to a distant metastatic-like nodule. This treatment was associated with an increase in proliferative CD8+ T cells and of IFN-γ expression, a decrease in proliferative regulatory T cells but also, surprisingly, an increase in myeloid derived suppressor cells. Conclusions Local administration of CpG emulsified with Lipiodol led to an effective antitumoral effect when combined to systemic anti-PD-1 therapy. Lipiodol, apart from its radiopaque properties, is an efficient drug-delivery system. The formulated oil-in-water emulsion allows efficient loading and control release of CpG, which induces favorable immune modifications in this murine tumor model.
Collapse
Affiliation(s)
- Anne-Laure Grindel
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
| | | | - Miguel Soares
- Guerbet, Research and Innovation Division, Aulnay-sous-Bois, France
| | - Nabiha Bouzakher
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
| | | | - Robin Santus
- Guerbet, Research and Innovation Division, Aulnay-sous-Bois, France
| | - Olivia Bawa
- INSERM US23 Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse (AMMICA), Villejuif, France
| | | | | | - Philippe Robert
- Guerbet, Research and Innovation Division, Aulnay-sous-Bois, France
| | - Jean-Francois Emile
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
- Département d’anatomie Pathologique et de Cytologie, Hôpital Ambroise-Paré, Boulogne-Billancourt, France
| | - Claude Capron
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
- Immunology and Hematology Department, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| |
Collapse
|
5
|
Korábková E, Kašpárková V, Vašíček O, Víchová Z, Káčerová S, Valášková K, Urbánková L, Vícha J, Münster L, Skopalová K, Humpolíček P. Pickering emulsions as an effective route for the preparation of bioactive composites: A study of nanocellulose/polyaniline particles with immunomodulatory effect. Carbohydr Polym 2024; 323:121429. [PMID: 37940298 DOI: 10.1016/j.carbpol.2023.121429] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/04/2023] [Accepted: 09/21/2023] [Indexed: 11/10/2023]
Abstract
Several studies have reported on application of cellulose particles for stabilizing Pickering emulsions (PE). Here we employ an original approach that involves using these particles as a part of advanced composite colloids made of conducting polymer polyaniline (PANI) and cellulose nanocrystals (CNC) or nanofibrils (CNF). PANI/cellulose particles were prepared using oxidative polymerization of aniline in situ in the presence of CNC or CNF. The type and amount of celluloses (CNC vs CNF) and concentration of precursors (aniline monomer and oxidant) used in the reaction determined properties of the colloidal particles, such as size, morphology and content of PANI. The particles demonstrated intriguing biological characteristics, including no cytotoxicity, antibacterial activity against Staphylococcus aureus and Escherichia coli, antioxidant activity and related immunomodulatory activity. For the first time, such composites were used to successfully stabilize oil-in-water PE with undecane or capric/caprylic triglyceride oils. The properties of the emulsions were determined by the PANI/cellulose particles and oil used. The key finding of the study is the demonstrated ability of PANI/cellulose particles to stabilize PE, as well as the excellent antioxidant activity and ROS scavenging action originating from PANI presence, indicating potential of such systems for use in biomedicine, particularly for wound healing.
Collapse
Affiliation(s)
- Eva Korábková
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic
| | - Věra Kašpárková
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic; Department of Fat, Surfactant and Cosmetics Technology, Faculty of Technology, Tomas Bata University in Zlín, nám. T. G. Masaryka 5555, 760 01 Zlín, Czech Republic
| | - Ondřej Vašíček
- Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, 612 65 Brno, Czech Republic.
| | - Zdenka Víchová
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic
| | - Simona Káčerová
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic
| | - Kristýna Valášková
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic
| | - Lucie Urbánková
- Department of Fat, Surfactant and Cosmetics Technology, Faculty of Technology, Tomas Bata University in Zlín, nám. T. G. Masaryka 5555, 760 01 Zlín, Czech Republic
| | - Jan Vícha
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic
| | - Lukáš Münster
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic
| | - Kateřina Skopalová
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic
| | - Petr Humpolíček
- Centre of Polymer Systems, Tomas Bata University in Zlin, nám. T.G.Masaryka 5555, 760 01 Zlin, Czech Republic; Department of Fat, Surfactant and Cosmetics Technology, Faculty of Technology, Tomas Bata University in Zlín, nám. T. G. Masaryka 5555, 760 01 Zlín, Czech Republic.
| |
Collapse
|
6
|
Meyblum L, Chevaleyre C, Susini S, Jego B, Deschamps F, Kereselidze D, Bonnet B, Marabelle A, de Baere T, Lebon V, Tselikas L, Truillet C. Local and distant response to intratumoral immunotherapy assessed by immunoPET in mice. J Immunother Cancer 2023; 11:e007433. [PMID: 37949616 PMCID: PMC10649793 DOI: 10.1136/jitc-2023-007433] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Despite the promising efficacy of immune checkpoint blockers (ICB), tumor resistance and immune-related adverse events hinder their success in cancer treatment. To address these challenges, intratumoral delivery of immunotherapies has emerged as a potential solution, aiming to mitigate side effects through reduced systemic exposure while increasing effectiveness by enhancing local bioavailability. However, a comprehensive understanding of the local and systemic distribution of ICBs following intratumoral administration, as well as their impact on distant tumors, remains crucial for optimizing their therapeutic potential.To comprehensively investigate the distribution patterns following the intratumoral and intravenous administration of radiolabeled anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and to assess its corresponding efficacy in both injected and non-injected tumors, we conducted an immunoPET imaging study. METHODS CT26 and MC38 syngeneic colorectal tumor cells were implanted subcutaneously on both flanks of Balb/c and C57Bl/6 mice, respectively. Hamster anti-mouse CTLA-4 antibody (9H10) labeled with zirconium-89 ([89Zr]9H10) was intratumorally or intravenously administered. Whole-body distribution of the antibody was monitored by immunoPET imaging (n=12 CT26 Balb/c mice, n=10 MC38 C57Bl/6 mice). Tumorous responses to injected doses (1-10 mg/kg) were correlated with specific uptake of [89Zr]9H10 (n=24). Impacts on the tumor microenvironment were assessed by immunofluorescence and flow cytometry. RESULTS Half of the dose was cleared into the blood 1 hour after intratumoral administration. Despite this, 7 days post-injection, 6-8% of the dose remained in the intratumoral-injected tumors. CT26 tumors with prolonged ICB exposure demonstrated complete responses. Seven days post-injection, the contralateral non-injected tumor uptake of the ICB was comparable to the one achieved through intravenous administration (7.5±1.7% ID.cm-3 and 7.6±2.1% ID.cm-3, respectively) at the same dose in the CT26 model. This observation was confirmed in the MC38 model. Consistent intratumoral pharmacodynamic effects were observed in both intratumoral and intravenous treatment groups, as evidenced by a notable increase in CD8+T cells within the CT26 tumors following treatment. CONCLUSIONS ImmunoPET-derived pharmacokinetics supports intratumoral injection of ICBs to decrease systemic exposure while maintaining efficacy compared with intravenous. Intratumoral-ICBs lead to high local drug exposure while maintaining significant therapeutic exposure in non-injected tumors. This immunoPET approach is applicable for clinical practice to support evidence-based drug development.
Collapse
Affiliation(s)
- Louis Meyblum
- Université Paris-Saclay, CEA, CNRS, INSERM UMR1281, Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay (BioMaps), Orsay, France
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Service de Radiologie Interventionnelle, Gustave Roussy, Villejuif, France
| | - Céline Chevaleyre
- Université Paris-Saclay, CEA, CNRS, INSERM UMR1281, Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay (BioMaps), Orsay, France
| | - Sandrine Susini
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
- BIOTHERIS, Centre d'Investigation Clinique, INSERM U1428, Villejuif, France
| | - Benoit Jego
- Université Paris-Saclay, CEA, CNRS, INSERM UMR1281, Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay (BioMaps), Orsay, France
| | - Frederic Deschamps
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Service de Radiologie Interventionnelle, Gustave Roussy, Villejuif, France
- BIOTHERIS, Centre d'Investigation Clinique, INSERM U1428, Villejuif, France
| | - Dimitri Kereselidze
- Université Paris-Saclay, CEA, CNRS, INSERM UMR1281, Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay (BioMaps), Orsay, France
| | - Baptiste Bonnet
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Service de Radiologie Interventionnelle, Gustave Roussy, Villejuif, France
- BIOTHERIS, Centre d'Investigation Clinique, INSERM U1428, Villejuif, France
| | - Aurelien Marabelle
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
- BIOTHERIS, Centre d'Investigation Clinique, INSERM U1428, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris Saclay, Saint Aubin, France
| | - Thierry de Baere
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Service de Radiologie Interventionnelle, Gustave Roussy, Villejuif, France
- BIOTHERIS, Centre d'Investigation Clinique, INSERM U1428, Villejuif, France
- Université Paris Saclay, Saint Aubin, France
| | - Vincent Lebon
- Université Paris-Saclay, CEA, CNRS, INSERM UMR1281, Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay (BioMaps), Orsay, France
| | - Lambros Tselikas
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Service de Radiologie Interventionnelle, Gustave Roussy, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
- BIOTHERIS, Centre d'Investigation Clinique, INSERM U1428, Villejuif, France
- Université Paris Saclay, Saint Aubin, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, INSERM UMR1281, Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay (BioMaps), Orsay, France
| |
Collapse
|
7
|
Zhang Q, Zhao Q, Zhu B, Chen R, Zhou Y, Pei X, Zhou H, An H, Tan Y, Chen C. Acetalized starch-based nanoparticles stabilized acid-sensitive Pickering emulsion as a potential antitumor drug carrier. Int J Biol Macromol 2023:125393. [PMID: 37331543 DOI: 10.1016/j.ijbiomac.2023.125393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/25/2023] [Accepted: 06/12/2023] [Indexed: 06/20/2023]
Abstract
Pickering emulsions are attracting increased attention owing to their therapeutic applications. However, the slow-release property of Pickering emulsions and the in vivo solid particle accumulation caused by the solid particle stabilizer film limit their applications in therapeutic delivery. In this study, drug-loaded, acid-sensitive Pickering emulsions were prepared using acetal-modified starch-based nanoparticles as stabilizers. The acetalized starch-based nanoparticles (Ace-SNPs) not only act as a solid-particle emulsifier to stabilize Pickering emulsions but also exhibit acid sensitivity and degradability, conducive to the destabilization of Pickering emulsions to release the drug and reduce the effect of particle accumulation in an acidic therapeutic environment. In vitro drug release profiles show that 50 % of curcumin was released in 12 h in an acidic medium (pH 5.4), whereas only 14 % of curcumin was released in 12 h at higher pH (7.4), indicating that the Ace-SNP stabilized Pickering emulsion possess good acid-responsive release characteristics in acidic environments. Moreover, acetalized starch-based nanoparticles and their degradation products showed good biocompatibility, and the resulting curcumin-loaded Pickering emulsions exhibited significant anticancer activity. These features suggest that the acetalized starch-based nanoparticle-stabilized Pickering emulsion has the potential for application as an antitumor drug carrier to enhance therapeutic effects.
Collapse
Affiliation(s)
- Qimeng Zhang
- School of Petrochemical Engineering, Liaoning Petrochemical University, Fushun 113001, China; Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Qifan Zhao
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Bingbing Zhu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325000, China
| | - Rong Chen
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Yating Zhou
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Xiaopeng Pei
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| | - Hua Zhou
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Huiyong An
- School of Petrochemical Engineering, Liaoning Petrochemical University, Fushun 113001, China.
| | - Ying Tan
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Chengshui Chen
- Department of Puelmonary and Critical Care Medicine, Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China; Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
8
|
Leading Edge: Intratumor Delivery of Monoclonal Antibodies for the Treatment of Solid Tumors. Int J Mol Sci 2023; 24:ijms24032676. [PMID: 36768997 PMCID: PMC9917067 DOI: 10.3390/ijms24032676] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/02/2023] Open
Abstract
Immunotherapies based on immune checkpoint blockade have shown remarkable clinical outcomes and durable responses in patients with many tumor types. Nevertheless, these therapies lack efficacy in most cancer patients, even causing severe adverse events in a small subset of patients, such as inflammatory disorders and hyper-progressive disease. To diminish the risk of developing serious toxicities, intratumor delivery of monoclonal antibodies could be a solution. Encouraging results have been shown in both preclinical and clinical studies. Thus, intratumor immunotherapy as a new strategy may retain efficacy while increasing safety. This approach is still an exploratory frontier in cancer research and opens up new possibilities for next-generation personalized medicine. Local intratumor delivery can be achieved through many means, but an attractive approach is the use of gene therapy vectors expressing mAbs inside the tumor mass. Here, we summarize basic, translational, and clinical results of intratumor mAb delivery, together with descriptions of non-viral and viral strategies for mAb delivery in preclinical and clinical development. Currently, this is an expanding research subject that will surely play a key role in the future of oncology.
Collapse
|
9
|
Meng L, Wei Y, Xiao Y. Chemo-immunoablation of solid tumors: A new concept in tumor ablation. Front Immunol 2023; 13:1057535. [PMID: 36713427 PMCID: PMC9878389 DOI: 10.3389/fimmu.2022.1057535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
Chemical ablation was designed to inject chemical agents directly into solid tumors to kill cells and is currently only used clinically for the palliative treatment of tumors. The application and combination of different drugs, from anhydrous ethanol, and glacial acetic acid to epi-amycin, have been clinically tested for a long time. The effectiveness is unsatisfactory due to chemical agents' poor diffusion and concentration. Immunotherapy is considered a prospective oncologic therapeutic. Still, the clinical applications were limited by the low response rate of patients to immune drugs and the immune-related adverse effects caused by high doses. The advent of intratumoral immunotherapy has well addressed these issues. However, the efficacy of intratumoral immunotherapy alone is uncertain, as suggested by the results of preclinical and clinical studies. In this study, we will focus on the research of immunosuppressive tumor microenvironment with chemoablation and intratumoral immunotherapy, the synergistic effect between chemotherapeutic drugs and immunotherapy. We propose a new concept of intratumoral chemo-immunoablation. The concept opens a new perspective for tumor treatment from direct killing of tumor cells while, enhancing systemic anti-tumor immune response, and significantly reducing adverse effects of drugs.
Collapse
Affiliation(s)
- Liangliang Meng
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Radiology, Chinese PAP Hospital of Beijing, Beijing, China
| | - Yingtian Wei
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yueyong Xiao
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,*Correspondence: Yueyong Xiao,
| |
Collapse
|
10
|
Som A, Rosenboom JG, Chandler A, Sheth RA, Wehrenberg-Klee E. Image-guided intratumoral immunotherapy: Developing a clinically practical technology. Adv Drug Deliv Rev 2022; 189:114505. [PMID: 36007674 PMCID: PMC10456124 DOI: 10.1016/j.addr.2022.114505] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 07/14/2022] [Accepted: 08/17/2022] [Indexed: 02/07/2023]
Abstract
Immunotherapy has revolutionized the contemporary oncology landscape, with durable responses possible across a range of cancer types. However, the majority of cancer patients do not respond to immunotherapy due to numerous immunosuppressive barriers. Efforts to overcome these barriers and increase systemic immunotherapy efficacy have sparked interest in the local intratumoral delivery of immune stimulants to activate the local immune response and subsequently drive systemic tumor immunity. While clinical evaluation of many therapeutic candidates is ongoing, development is hindered by a lack of imaging confirmation of local delivery, insufficient intratumoral drug distribution, and a need for repeated injections. The use of polymeric drug delivery systems, which have been widely used as platforms for both image guidance and controlled drug release, holds promise for delivery of intratumoral immunoadjuvants and the development of an in situ cancer vaccine for patients with metastatic cancer. In this review, we explore the current state of the field for intratumoral delivery and methods for optimizing controlled drug release, as well as practical considerations for drug delivery design to be optimized for clinical image guided delivery particularly by CT and ultrasound.
Collapse
Affiliation(s)
- Avik Som
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, United States
| | - Jan-Georg Rosenboom
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, United States; Department of Gastroenterology, Brigham and Women's Hospital, United States
| | - Alana Chandler
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, United States; Department of Gastroenterology, Brigham and Women's Hospital, United States
| | - Rahul A Sheth
- Department of Interventional Radiology, M.D. Anderson Cancer Center, United States
| | - Eric Wehrenberg-Klee
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, United States.
| |
Collapse
|
11
|
Humeau J, Le Naour J, Galluzzi L, Kroemer G, Pol JG. Trial watch: intratumoral immunotherapy. Oncoimmunology 2021; 10:1984677. [PMID: 34676147 PMCID: PMC8526014 DOI: 10.1080/2162402x.2021.1984677] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
While chemotherapy and radiotherapy remain the first-line approaches for the management of most unresectable tumors, immunotherapy has emerged in the past two decades as a game-changing treatment, notably with the clinical success of immune checkpoint inhibitors. Immunotherapies aim at (re)activating anticancer immune responses which occur in two main steps: (1) the activation and expansion of tumor-specific T cells following cross-presentation of tumor antigens by specialized myeloid cells (priming phase); and (2) the immunological clearance of malignant cells by these antitumor T lymphocytes (effector phase). Therapeutic vaccines, adjuvants, monoclonal antibodies, cytokines, immunogenic cell death-inducing agents including oncolytic viruses, anthracycline-based chemotherapy and radiotherapy, as well as adoptive cell transfer, all act at different levels of this cascade to (re)instate cancer immunosurveillance. Intratumoral delivery of these immunotherapeutics is being tested in clinical trials to promote superior antitumor immune activity in the context of limited systemic toxicity.
Collapse
Affiliation(s)
- Juliette Humeau
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Julie Le Naour
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Institut Universitaire de France, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Jonathan G. Pol
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| |
Collapse
|
12
|
Melero I, Castanon E, Alvarez M, Champiat S, Marabelle A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat Rev Clin Oncol 2021; 18:558-576. [PMID: 34006998 PMCID: PMC8130796 DOI: 10.1038/s41571-021-00507-y] [Citation(s) in RCA: 296] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2021] [Indexed: 02/04/2023]
Abstract
Immune-checkpoint inhibitors and chimeric antigen receptor (CAR) T cells are revolutionizing oncology and haematology practice. With these and other immunotherapies, however, systemic biodistribution raises safety issues, potentially requiring the use of suboptimal doses or even precluding their clinical development. Delivering or attracting immune cells or immunomodulatory factors directly to the tumour and/or draining lymph nodes might overcome these problems. Hence, intratumoural delivery and tumour tissue-targeted compounds are attractive options to increase the in situ bioavailability and, thus, the efficacy of immunotherapies. In mouse models, intratumoural administration of immunostimulatory monoclonal antibodies, pattern recognition receptor agonists, genetically engineered viruses, bacteria, cytokines or immune cells can exert powerful effects not only against the injected tumours but also often against uninjected lesions (abscopal or anenestic effects). Alternatively, or additionally, biotechnology strategies are being used to achieve higher functional concentrations of immune mediators in tumour tissues, either by targeting locally overexpressed moieties or engineering 'unmaskable' agents to be activated by elements enriched within tumour tissues. Clinical trials evaluating these strategies are ongoing, but their development faces issues relating to the administration methodology, pharmacokinetic parameters, pharmacodynamic end points, and immunobiological and clinical response assessments. Herein, we discuss these approaches in the context of their historical development and describe the current landscape of intratumoural or tumour tissue-targeted immunotherapies.
Collapse
Affiliation(s)
- Ignacio Melero
- Department of Immunology, Clínica Universidad de Navarra, Pamplona, Spain.
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
- Program for Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Eduardo Castanon
- Department of Immunology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Maite Alvarez
- Program for Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Université Paris Saclay, Gustave Roussy, Villejuif, France
- INSERM U1015, Gustave Roussy, Villejuif, France
- Biotherapies for In Situ Antitumor Immunization (BIOTHERIS), Centre d'Investigation Clinique INSERM CICBT1428, Villejuif, France
| | - Aurelien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Université Paris Saclay, Gustave Roussy, Villejuif, France.
- INSERM U1015, Gustave Roussy, Villejuif, France.
- Biotherapies for In Situ Antitumor Immunization (BIOTHERIS), Centre d'Investigation Clinique INSERM CICBT1428, Villejuif, France.
| |
Collapse
|
13
|
Gorbet MJ, Singh A, Mao C, Fiering S, Ranjan A. Using nanoparticles for in situ vaccination against cancer: mechanisms and immunotherapy benefits. Int J Hyperthermia 2021; 37:18-33. [PMID: 33426995 DOI: 10.1080/02656736.2020.1802519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy to treat cancer is now an established clinical approach. Immunotherapy can be applied systemically, as done with checkpoint blockade antibodies, but it can also be injected directly into identified tumors, in a strategy of in situ vaccination (ISV). ISV is designed to stimulate a strong local antitumor immune response involving both innate and adaptive immune cells, and through this generate a systemic antitumor immune response against metastatic tumors. A variety of ISVs have been utilized to generate an immunostimulatory tumor microenvironment (TME). These include attenuated microorganisms, recombinant proteins, small molecules, physical disruptors of TME (alternating magnetic and focused ultrasound heating, photothermal therapy, and radiotherapy), and more recently nanoparticles (NPs). NPs are attractive and unique since they can load multiple drugs or other reagents to influence immune and cancer cell functions in the TME, affording a unique opportunity to stimulate antitumor immunity. Here, we describe the NP-ISV therapeutic mechanisms, review chemically synthesized NPs (i.e., liposomes, polymeric, chitosan-based, inorganic NPs, etc.), biologically derived NPs (virus and bacteria-based NPs), and energy-activated NP-ISVs in the context of their use as local ISV. Data suggests that NP-ISVs can enhance outcomes of immunotherapeutic regimens including those utilizing tumor hyperthermia and checkpoint blockade therapies.
Collapse
Affiliation(s)
| | - Akansha Singh
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center at Dartmouth and Dartmouth Hitchcock, Lebanon, NH, USA
| | - Ashish Ranjan
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
14
|
Hashemzadeh N, Dolatkhah M, Adibkia K, Aghanejad A, Barzegar-Jalali M, Omidi Y, Barar J. Recent advances in breast cancer immunotherapy: The promising impact of nanomedicines. Life Sci 2021; 271:119110. [PMID: 33513401 DOI: 10.1016/j.lfs.2021.119110] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/29/2022]
Abstract
Breast cancer (BC) is one of the prevalent cancers among women. Generally, the treatment of BC is mostly based on several prominent strategies, including chemotherapy, surgery, endocrine therapy, molecular targeted therapy, and radiation. Owing to the growing knowledge about the complexity of BC pathobiology, immunotherapy as a promising treatment modality has substantially improved the patients' care in the clinic. Immunotherapy is used to harness the patient's immune system to recognize and battle devious cancer cells. As a novel therapy approach, this emerging strategy targets the key molecular entities of tumor tissue. To achieve maximal therapeutic impacts, the dynamic interplay between cancer and immune cells needs to be fully comprehended. The key molecular machinery of solid tumors can be targeted by nanoscale immunomedicines. While discussing the potential biomarkers involved in the initiation and progression of BC, we aimed to provide comprehensive insights into the immunotherapy and articulate the recent advances in terms of the therapeutic strategies used to control this disease, including immune checkpoint inhibitors, vaccines, chimeric antigen receptor T cells therapy, and nanomedicines.
Collapse
Affiliation(s)
- Nastaran Hashemzadeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mitra Dolatkhah
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Barzegar-Jalali
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Tselikas L, Champiat S, Sheth RA, Yevich S, Ammari S, Deschamps F, Farhane S, Roux C, Susini S, Mouraud S, Delpla A, Raoult T, Robert C, Massard C, Barlesi F, Soria JC, Marabelle A, de Baere T. Interventional Radiology for Local Immunotherapy in Oncology. Clin Cancer Res 2021; 27:2698-2705. [PMID: 33419781 DOI: 10.1158/1078-0432.ccr-19-4073] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/24/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022]
Abstract
Human intratumoral immunotherapy (HIT-IT) is under rapid development, with promising preliminary results and high expectations for current phase III trials. While outcomes remain paramount for patients and the referring oncologists, the technical aspects of drug injection are critical to the interventional radiologist to ensure optimal and reproducible outcomes. The technical considerations for HIT-IT affect the safety, efficacy, and further development of this treatment option. Image-guided access to the tumor allows the therapeutic index of a treatment to be enhanced by increasing the intratumoral drug concentration while minimizing its systemic exposure and associated on-target off-tumor adverse events. Direct access to the tumor also enables the acquisition of cancer tissue for sequential sampling to better understand the pharmacodynamics of the injected immunotherapy and its efficacy through correlation of immune responses, pathologic responses, and imaging tumor response. The aim of this article is to share the technical insights of HIT-IT, with particular consideration for patient selection, lesion assessment, image guidance, and technical injection options. In addition, the organization of a standard patient workflow is discussed, so as to optimize HIT-IT outcome and the patient experience.
Collapse
Affiliation(s)
- Lambros Tselikas
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France. .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Rahul A Sheth
- Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steve Yevich
- Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samy Ammari
- Radiology Department, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Frederic Deschamps
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Siham Farhane
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Charles Roux
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Sandrine Susini
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Severine Mouraud
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Alexandre Delpla
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France
| | - Thibault Raoult
- Service de Promotion des Etudes Cliniques (SPEC), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Fabrice Barlesi
- Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Jean-Charles Soria
- Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| | - Aurélien Marabelle
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry de Baere
- Département d'Anesthésie, Chirurgie et Interventionnel (DACI), Gustave Roussy, Villejuif, France.,Gustave Roussy Cancer Campus, Université Paris Saclay, Saint-Aubin, France
| |
Collapse
|
16
|
Dovedi SJ, Elder MJ, Yang C, Sitnikova SI, Irving L, Hansen A, Hair J, Jones DC, Hasani S, Wang B, Im SA, Tran B, Subramaniam DS, Gainer SD, Vashisht K, Lewis A, Jin X, Kentner S, Mulgrew K, Wang Y, Overstreet MG, Dodgson J, Wu Y, Palazon A, Morrow M, Rainey GJ, Browne GJ, Neal F, Murray TV, Toloczko AD, Dall'Acqua W, Achour I, Freeman DJ, Wilkinson RW, Mazor Y. Design and Efficacy of a Monovalent Bispecific PD-1/CTLA4 Antibody That Enhances CTLA4 Blockade on PD-1 + Activated T Cells. Cancer Discov 2021; 11:1100-1117. [PMID: 33419761 DOI: 10.1158/2159-8290.cd-20-1445] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/04/2020] [Accepted: 12/17/2020] [Indexed: 11/16/2022]
Abstract
The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1+ activated T cells. We show that MEDI5752 preferentially saturates CTLA4 on PD-1+ T cells versus PD-1- T cells, reducing the dose required to elicit IL2 secretion. Unlike conventional PD-1/CTLA4 mAbs, MEDI5752 leads to the rapid internalization and degradation of PD-1. Moreover, we show that MEDI5752 preferentially localizes and accumulates in tumors providing enhanced activity when compared with a combination of mAbs targeting PD-1 and CTLA4 in vivo. Following treatment with MEDI5752, robust partial responses were observed in two patients with advanced solid tumors. MEDI5752 represents a novel immunotherapy engineered to preferentially inhibit CTLA4 on PD-1+ T cells. SIGNIFICANCE: The unique characteristics of MEDI5752 represent a novel immunotherapy engineered to direct CTLA4 inhibition to PD-1+ T cells with the potential for differentiated activity when compared with current conventional mAb combination strategies targeting PD-1 and CTLA4. This molecule therefore represents a step forward in the rational design of cancer immunotherapy.See related commentary by Burton and Tawbi, p. 1008.This article is highlighted in the In This Issue feature, p. 995.
Collapse
Affiliation(s)
- Simon J Dovedi
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
| | | | - Chunning Yang
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | | | - Lorraine Irving
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Anna Hansen
- Translational Science and Experimental Medicine, Respiratory and Immunology (RI), Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - James Hair
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Des C Jones
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Sumati Hasani
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Bo Wang
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Seock-Ah Im
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul National University School of Medicine, Seoul, Korea
| | - Ben Tran
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | | | - Kapil Vashisht
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Arthur Lewis
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Xiaofang Jin
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Stacy Kentner
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kathy Mulgrew
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Yaya Wang
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | - James Dodgson
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Yanli Wu
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Asis Palazon
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Gareth J Browne
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Frances Neal
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Thomas V Murray
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Aleksandra D Toloczko
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - William Dall'Acqua
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Ikbel Achour
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Yariv Mazor
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland.
| |
Collapse
|
17
|
Baère TD. Immunotherapy and Interventional Radiology (IR): Time to Change the Paradigm—Keynote Lecture PAIRS 2021. THE ARAB JOURNAL OF INTERVENTIONAL RADIOLOGY 2021. [DOI: 10.1055/s-0041-1731604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Thierry de Baère
- Interventional Radiology, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Villejuif, France
| |
Collapse
|