1
|
Zou JX, Chang MR, Kuznetsov NA, Kee JX, Babak MV, Ang WH. Metal-based immunogenic cell death inducers for cancer immunotherapy. Chem Sci 2025; 16:6160-6187. [PMID: 40160356 PMCID: PMC11949249 DOI: 10.1039/d4sc08495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunogenic cell death (ICD) has attracted enormous attention over the past decade due to its unique characteristics in cancer cell death and its role in activating innate and adaptive immune responses against tumours. Many efforts have been dedicated to screening, identifying and discovering ICD inducers, resulting in the validation of several based on metal complexes. In this review, we provide a comprehensive summary of current metal-based ICD inducers, their molecular mechanisms for triggering ICD initiation and subsequent protective antitumour immune responses, along with considerations for validating ICD both in vitro and in vivo. We also aim to offer insights into the future development of metal complexes with enhanced ICD-inducing properties and their applications in potentiating antitumour immunity.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Meng Rui Chang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Nikita A Kuznetsov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Jia Xuan Kee
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- NUS Graduate School - Integrative Science and Engineering Programme (ISEP), National University of Singapore 21 Lower Kent Ridge Rd Singapore 119077 Singapore
| |
Collapse
|
2
|
Han M, Zhou S, Liao Z, Zishan C, Yi X, Wu C, Zhang D, He Y, Leong KW, Zhong Y. Bimetallic peroxide-based nanotherapeutics for immunometabolic intervention and induction of immunogenic cell death to augment cancer immunotherapy. Biomaterials 2025; 315:122934. [PMID: 39509856 DOI: 10.1016/j.biomaterials.2024.122934] [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: 07/19/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
Immunotherapy has transformed cancer treatment, but its efficacy is often limited by the immunosuppressive characteristics of the tumor microenvironment (TME), which are predominantly influenced by the metabolism of cancer cells. Among these metabolic pathways, the indoleamine 2,3-dioxygenase (IDO) pathway is particularly crucial, as it significantly contributes to TME suppression and influences immune cell activity. Additionally, inducing immunogenic cell death (ICD) in tumor cells can reverse the immunosuppressive TME, thereby enhancing the efficacy of immunotherapy. Herein, we develop CGDMRR, a novel bimetallic peroxide-based nanodrug based on copper-cerium peroxide nanoparticles. These nanotherapeutics are engineered to mitigate tumor hypoxia and deliver therapeutics such as 1-methyltryptophan (1MT), glucose oxidase (GOx), and doxorubicin (Dox) in a targeted manner. The design aims to alleviate tumor hypoxia, reduce the immunosuppressive effects of the IDO pathway, and promote ICD. CGDMRR effectively inhibits the growth of 4T1 tumors and elicits antitumor immune responses by leveraging immunometabolic interventions and therapies that induce ICD. Furthermore, when CGDMRR is combined with a clinically certified anti-PD-L1 antibody, its efficacy in inhibiting tumor growth is enhanced. This improved efficacy extends beyond unilateral tumor models, also affecting bilateral tumors and lung metastases, due to the activation of systemic antitumor immunity. This study underscores CGDMRR's potential to augment the efficacy of PD-L1 blockade in breast cancer immunotherapy.
Collapse
Affiliation(s)
- Min Han
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Shiying Zhou
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Zunde Liao
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Chen Zishan
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Xiangting Yi
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Chuanbin Wu
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Dongmei Zhang
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Yao He
- Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, United States.
| | - Yiling Zhong
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China; Department of Biomedical Engineering, Columbia University, New York, NY, 10027, United States.
| |
Collapse
|
3
|
Reinhold A, Glasow A, Nürnberger S, Weimann A, Telemann L, Stolzenburg JU, Neuhaus J, Berndt-Paetz M. Ionizing radiation and photodynamic therapy lead to multimodal tumor cell death, synergistic cytotoxicity and immune cell invasion in human bladder cancer organoids. Photodiagnosis Photodyn Ther 2025; 51:104459. [PMID: 39746560 DOI: 10.1016/j.pdpdt.2024.104459] [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: 10/09/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Photodynamic therapy (PDT) and radiotherapy using ionizing radiation (IR) are promising options for organ-preserving treatment of bladder cancer (BCa). A combination therapy (IR+PDT) could be beneficial for BCa treatment. PURPOSE For PDT, we used the near-infrared photosensitizer tetrahydroporphyrin-tetratosylate (THPTS) showing high therapeutic efficacy. Treatment responses were analyzed in BCa organoids. METHODS Organoids consisting of BCa cells lines, bladder fibroblasts and muscle cells were treated with IR (9 Gy) and/or PDT using THPTS (25, 50 μM; 20 J/cm2). Cytotoxicity was determined by microscopy, cell-based assays and histology. The cell death mode was analyzed by applying specific inhibitors followed by immunofluorescence or qPCR analyses of cell death markers. A matrix-based co-culture model was used to study T cell migration into the environment of treated organoids. RESULTS PDT and/or IR resulted in concentration-dependent reduction of metabolic activity, organoid diameter and integrity. Higher cytotoxicity of IR+PDT vs. monotherapies was observed after 72 h. Non-malignant organoids showed no cytotoxic effects. While apoptosis, necroptosis and ferroptosis were clearly involved in cell death of T-24 cells, cytotoxicity in RT-112 cells was probably provoked by apoptosis, ferroptosis and pyroptosis. IR+PDT resulted in significant migration of Jurkat cells into ECM-embedded organoids within 3 days after treatment. CONCLUSION Treatment with IR+PDT showed tumor-selective cytotoxicity with additive or synergistic effects in BCa organoids. Thereby, IR+PDT led to multimodal cell death depending on the cellular context. Migration of T cells into the organoid environment illustrates the immunogenic potential of IR+PDT. Therefore, it might be a promising approach for organ-preserving BCa treatment.
Collapse
Affiliation(s)
- Annabell Reinhold
- Department of Urology, Research Laboratories, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Annegret Glasow
- Department of Radiation Therapy, University of Leipzig, Stephanstraße 9a, 04103 Leipzig, Germany
| | - Sandra Nürnberger
- Department of Urology, Research Laboratories, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Annett Weimann
- Department of Urology, Research Laboratories, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Lucie Telemann
- Department of Urology, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
| | - Jens-Uwe Stolzenburg
- Department of Urology, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
| | - Jochen Neuhaus
- Department of Urology, Research Laboratories, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Mandy Berndt-Paetz
- Department of Urology, Research Laboratories, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany.
| |
Collapse
|
4
|
Huang G, Zhang L, Feng J, Wu D, Wu L, Pan W, Jiang Y, Chen M, Chen J, Shui P. Hypoxia-Responsive Covalent Organic Framework Nanoplatform for Breast-Cancer-Targeted Cocktail Immunotherapy via Triple Therapeutic Switch Mechanisms. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407553. [PMID: 39797461 DOI: 10.1002/smll.202407553] [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: 08/26/2024] [Revised: 12/23/2024] [Indexed: 01/13/2025]
Abstract
Covalent organic frameworks (COFs), known for their exceptional in situ encapsulation and precise release capabilities, are emerging as pioneering drug delivery systems. This study introduces a hypoxia-responsive COF designed to encapsulate the chemotherapy drug gambogic acid (GA) in situ. Bimetallic gold-palladium islands were grown on UiO-66-NH2 (UiO) to form UiO@Au-Pdislands (UAPi), which were encapsulated with GA through COF membrane formation, resulting in a core-shell structure (UAPiGC). Further modification with hyaluronic acid (HA) created UiO@Au-Pdislands@GA-COF@HA (UAPiGCH) for enhanced tumor targeting. In the hypoxic tumor microenvironment, the COF collapses, releasing GA and UAPi, initiating a triple therapeutic response: nanozyme-catalyzed therapy, near-infrared II (NIR-II) mild photothermal therapy (mild-PTT), and chemotherapy. UAPi exhibits catalase (CAT)-like and peroxidase (POD)-like activities, generating oxygen to alleviate hypoxia and reactive oxygen species (ROS) for tumor destruction. GA acts as a chemotherapeutic agent and inhibits heat shock protein 90 (HSP90), enhancing photothermal sensitivity. In vitro and in vivo studies confirm UAPiGCH's ability to induce pyroptosis, stimulate dendritic cell maturation, and boost T cell infiltration, demonstrating its potential as a precise therapeutic nanoplatform. This strategy integrates multiple therapies into a hypoxia-responsive system, offering promising applications in cancer treatment.
Collapse
Affiliation(s)
- Guoqin Huang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Lianying Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jiahao Feng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Dan Wu
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Libo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Weilun Pan
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yu Jiang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ming Chen
- The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Jinxiang Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| |
Collapse
|
5
|
Wu T, Fang L, Ruan Y, Shi M, Su D, Ma Y, Ma M, Wang B, Liao Y, Han S, Lu X, Zhang C, Liu C, Zhang Y. Tumor aggression-defense index-a novel indicator to predicts recurrence and survival in stage II-III colorectal cancer. J Transl Med 2025; 23:107. [PMID: 39844178 PMCID: PMC11755833 DOI: 10.1186/s12967-025-06141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Although the TNM staging system plays a critical role in guiding adjuvant chemotherapy for colorectal cancer (CRC), its precision for risk stratification in stage II and III CRC patients with proficient DNA mismatch repair (pMMR) remains limited. Therefore, precise predictive models and research on postoperative treatments are crucial for enhancing patient survival and improving quality of life. METHODS This retrospective study analyzed 1051 pMMR CRC patients who underwent radical resection and were randomly assigned to training (n = 736) and validation (n = 315) groups. Immunohistochemistry and hematoxylin and eosin staining were utilized to evaluate regulatory-Immunoscore (RIS), tertiary lymphoid structures (TLS), and tumor budding (TB). The Tumor Aggression-Defense Index (TADI) was derived through a multi-factor COX regression model. Subgroup analysis demonstrated potential of TADI in guiding personalized adjuvant therapy for stage II and III CRC. RESULTS Univariate and multivariate Cox analysis indicated that TADI was an independent prognostic indicator. Among stage II CRC, chemotherapy was significantly correlated with improved recurrence times in individuals with intermediate (95% CI 0.19-0.59, P < 0.001) and high (95% CI 0.36-0.95, P = 0.031) TADI. In stage III CRC receiving adjuvant chemotherapy, a duration of 3 months or longer was notably associated with a prolonged time to recurrence in those with high TADI (95% CI 0.40-0.98, P = 0.041) compared to durations of less than 3 months. CONCLUSION The TADI serves as an effective parameter for predicting the survival outcomes of stage I-III pMMR CRC patients and guiding precision treatment strategies.
Collapse
Affiliation(s)
- Tong Wu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Lin Fang
- Phase I Clinical Research Center, The Affiliated Hospital of Qingdao University in Shandong, Qingdao, China
| | - Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Mengde Shi
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Dan Su
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Yue Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Ming Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Bojun Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Yuanyu Liao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Shuling Han
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Xiaolin Lu
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chunhui Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China.
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China.
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China.
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
6
|
Gabizon AA, Gabizon-Peretz S, Modaresahmadi S, La-Beck NM. Thirty years from FDA approval of pegylated liposomal doxorubicin (Doxil/Caelyx): an updated analysis and future perspective. BMJ ONCOLOGY 2025; 4:e000573. [PMID: 39885941 PMCID: PMC11751825 DOI: 10.1136/bmjonc-2024-000573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/16/2024] [Indexed: 02/01/2025]
Abstract
In 2025, it will be 30 years since the initial clinical approval of pegylated liposomal doxorubicin (PLD) by the Food and Drug Administration. PLD predated the field of nanomedicine and became a model nanomedicine setting key pharmacological principles (prolonged circulation, slow drug release and the enhanced permeability and retention (EPR) effect) for clinical application of other nano-drugs in cancer therapy. The impressive reduction of cardiotoxicity conferred by PLD is the most valuable clinical asset. While PLD has gained a strong foothold in relapsed ovarian cancer and metastatic breast cancer, it has not been extensively tested in primary (neoadjuvant) and adjuvant therapy and has not fulfilled the expectations from the results in animal models efficacy-wise. This discrepancy may be due to the large dose gap between mice and humans and the apparent variability of the EPR effect in human cancer. PLD is a complex product and we are still in a learning curve regarding a number of factors such as its interaction with the complement system and its immune modulatory properties, as well as its integration in multimodality therapy that may potentiate its value and role in cancer therapy.
Collapse
Affiliation(s)
- Alberto A Gabizon
- The Leah and Jakub Susskind Nano-Oncology Research Laboratory, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
- Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Shira Gabizon-Peretz
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Central, Israel
- Tel Aviv University, Faculty of Medicine, Tel Aviv, Israel
| | - Shadan Modaresahmadi
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H Hodge School of Pharmacy, Abilene, Texas, USA
| | - Ninh M La-Beck
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H Hodge School of Pharmacy, Abilene, Texas, USA
| |
Collapse
|
7
|
Xia Q, Zhang J. Interaction Between Autophagy and the Inflammasome in Human Tumors: Implications for the Treatment of Human Cancers. Cell Biochem Funct 2025; 43:e70035. [PMID: 39722223 DOI: 10.1002/cbf.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Autophagy is a physiologically regulated cellular process orchestrated by autophagy-related genes (ATGs) that, depending on the tumor type and stage, can either promote or suppress tumor growth and progression. It can also modulate cancer stem cell maintenance and immune responses. Therefore, targeted manipulation of autophagy may inhibit tumor development by overcoming tumor-promoting mechanisms. The inflammasome is another multifunctional bioprocess that induces a form of pro-inflammatory programmed cell death, called pyroptosis. Dysregulation or overactivation of the inflammasome has been implicated in tumor pathogenesis and development. Additionally, autophagy can inhibit the NLRP3 inflammasome by removing inflammatory drivers. Recent research suggests that the NLRP3 inflammasome, in turn, affects autophagy. Understanding the complex interplay between autophagy and inflammasomes could lead to more precise and effective strategies for cancer treatments. In this review, we summarize the impact of autophagy and inflammasome dysregulation on tumor progression or suppression. We then highlight their targeting for cancer treatment as monotherapy or in combination with other therapies. Furthermore, we discuss the interaction between autophagy and tumor-promoting inflammation or the NLRP3 inflammasome. Finally, based on recent findings, we review the potential of this interaction for cancer treatment.
Collapse
Affiliation(s)
- Qing Xia
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingzhou Zhang
- Peking Union Medical College, Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Han C, Xiao S, Xing Z, Xu X, Wang M, Han X, Adeli M, Qiu L, Ye L, Cheng C. NADPH Oxidases-Inspired Reactive Oxygen Biocatalysts with Electron-Rich Pt Sites to Potently Amplify Immune Checkpoint Blockade Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407644. [PMID: 39400421 DOI: 10.1002/adma.202407644] [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: 05/29/2024] [Revised: 07/19/2024] [Indexed: 10/15/2024]
Abstract
Clinical immune checkpoint blockade (ICB)-based immunotherapy of malignant tumors only elicits durable responses in a minority of patients, primarily due to the highly immunosuppressive tumor microenvironment. Although inducing immunogenic cell death (ICD) through reactive oxygen biocatalyst represents an attractive therapeutic strategy to amplify ICB, currently reported biocatalysts encounter insurmountable challenges in achieving high ROS-generating activity to induce potent ICD. Here, inspired by the natural catalytic characteristics of NADPH oxidases, the design of efficient, robust, and electron-rich Pt-based redox centers on the non-stoichiometric W18O49 substrates (Pt─WOx) to serve as bioinspired reactive oxygen biocatalysts to potently activate the ICD, which eventually enhance cancer immune responses and amplifies the ICB-based immunotherapy is reported. These studies demonstrate that the Pt─WOx exhibits rapid electron transfer capability and can promote the formation of electron-rich and low oxophilic Pt redox centers for superior reactive oxygen biocatalysis, which enables the Pt─WOx-based inducers to trigger endoplasmic reticulum stress directly and stimulate immune responses potently for amplifying the anti-PD-L1-based ICB therapy. This bioinspired design provides a straightforward strategy to engineer efficient, robust, and electron-rich reactive oxygen biocatalysts and also opens up a new avenue to create efficient ICD inducers for primary/metastatic tumor treatments.
Collapse
Affiliation(s)
- Chuyi Han
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Sutong Xiao
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Zhenyu Xing
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xiaohui Xu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xianglong Han
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mohsen Adeli
- Institute of Chemistry and Biochemistry, Freie Universitat Berlin, Takustr. 3, 14195, Berlin, Germany
- Department of Organic Chemistry, Faculty of Chemistry, Lorestan University, Khorramabad, 68137-17133, Iran
| | - Li Qiu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling Ye
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chong Cheng
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
9
|
Zhang X, Deng X, Tan J, Liu H, Zhang H, Li C, Li Q, Zhou J, Xiao Z, Li J. Idarubicin-loaded degradable hydrogel for TACE therapy enhances anti-tumor immunity in hepatocellular carcinoma. Mater Today Bio 2024; 29:101343. [PMID: 39687797 PMCID: PMC11647502 DOI: 10.1016/j.mtbio.2024.101343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/22/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a common and deadly cancer, often diagnosed at advanced stages, limiting surgical options. Transcatheter arterial chemoembolization (TACE) is a primary treatment for inoperable and involves the use of drug-eluting microspheres to slowly release chemotherapy drugs. However, patient responses to TACE vary, with some experiencing tumor progression and recurrence. Traditional TACE uses agents like oil-based drug emulsions and polyvinyl alcohol particles, which can permanently block blood vessels and increase tumor hypoxia. Additionally, TACE can suppress the immune system by reducing immune cell numbers and function, contributing to poor treatment outcomes. New approaches, like TACE using degradable starch microspheres and hydrogel-based materials, offer the potential to create different tumor environments that could improve both safety and efficacy. In our research, we developed a composite hydrogel (IF@Gel) made of Poloxamer-407 gel and Fe3O4 nanoparticles, loaded with idarubicin, to use as an embolic material for TACE in a rat model of orthotopic HCC. We observed promising therapeutic effects and investigated the impact on the tumor immune microenvironment, focusing on the role of immunogenic cell death (ICD). The composite hydrogel demonstrated excellent potential as an embolic material for TACE, and IF@Gel-based TACE demonstrated significant efficacy in rat HCC. Furthermore, our findings highlight the potential synergistic effects of ICD with anti-PD-L1 therapy, providing new insights into HCC treatment strategies. This study aims to provide improved treatment options for HCC and to deepen our understanding of the mechanisms of TACE and tumor environment regulation.
Collapse
Affiliation(s)
- Xiaokai Zhang
- Department of Hepatobiliopancreatic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450003, China
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiujiao Deng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Jizhou Tan
- Department of Stomatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Haikuan Liu
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Hong Zhang
- Department of Interventional Radiology and Vascular Surgery, The Sixth Affiliated Hospital of Jinan University, Dongguan 523067, China
| | - Chengzhi Li
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Qingjun Li
- Department of Hepatobiliopancreatic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450003, China
| | - Jinxue Zhou
- Department of Hepatobiliopancreatic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450003, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
10
|
Li Y, Wang J, Li Y, Luo Z, Peng T, Zou T. Nanomaterials based on hollow gold nanospheres for cancer therapy. Regen Biomater 2024; 11:rbae126. [PMID: 39664940 PMCID: PMC11631698 DOI: 10.1093/rb/rbae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 12/13/2024] Open
Abstract
Gold nanoparticles have recently been exploited as versatile nanocarriers in diagnostic and therapeutic drug delivery for cancer nanomedicine, owing to their biocompatibility, low biotoxicity, surface modifiability and plasma optical properties. A variety of gold nanoparticles have emerged for drug delivery, mainly including gold nanorods, gold nanocages, gold nanostars, gold solid nanospheres and hollow gold nanospheres (HGNs). Among these, HGNs have widely been studied for their higher photothermal conversion efficiency, wider spectral absorption range and stronger surface-enhanced Raman scattering compared with solid gold nanospheres. Therefore, nowadays, researchers prefer to use HGNs to other metal nanocarriers, which can not only play the role of controlled-release drugs but also act as photothermal agents for tumor therapy and diagnosis, due to their properties of surface modification. Combined with the Au-S bond on the surface of HGNs, the targeted preparation is loaded to achieve precise drug delivery. With the assistance of the photothermal characteristics of HGNs themselves, the efficacy of loaded drugs in HGNs is enhanced. In addition, HGNs also have vital values in the field of bioimaging, which serve as photothermal imaging agents and Raman scattering-guided preparations due to their surface-enhanced Raman scattering properties to assist researchers in achieving the purpose of tumor diagnosis. In this review, we summarize the synthesis methods of HGNs and the recent application of HGNs-based nanomaterials in the field of cancer diagnosis and therapy. In addition, the issues to be addressed were pointed out for a bright prospect of HGNs-based nanomaterials.
Collapse
Affiliation(s)
- You Li
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P.R. China
| | - Jing Wang
- Laboratory for Genetic Engineering of Antibodies and Functional Proteins, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Ying Li
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P.R. China
| | - Ziqiang Luo
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P.R. China
| | - Tao Peng
- GEM (Wuhan) Urban Mining Industrial Group Co., Ltd, Wuhan 430415, P.R. China
| | - Tao Zou
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P.R. China
| |
Collapse
|
11
|
Gong N, Alameh MG, El-Mayta R, Xue L, Weissman D, Mitchell MJ. Enhancing in situ cancer vaccines using delivery technologies. Nat Rev Drug Discov 2024; 23:607-625. [PMID: 38951662 DOI: 10.1038/s41573-024-00974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/03/2024]
Abstract
In situ cancer vaccination refers to any approach that exploits tumour antigens available at a tumour site to induce tumour-specific adaptive immune responses. These approaches hold great promise for the treatment of many solid tumours, with numerous candidate drugs under preclinical or clinical evaluation and several products already approved. However, there are challenges in the development of effective in situ cancer vaccines. For example, inadequate release of tumour antigens from tumour cells limits antigen uptake by immune cells; insufficient antigen processing by antigen-presenting cells restricts the generation of antigen-specific T cell responses; and the suppressive immune microenvironment of the tumour leads to exhaustion and death of effector cells. Rationally designed delivery technologies such as lipid nanoparticles, hydrogels, scaffolds and polymeric nanoparticles are uniquely suited to overcome these challenges through the targeted delivery of therapeutics to tumour cells, immune cells or the extracellular matrix. Here, we discuss delivery technologies that have the potential to reduce various clinical barriers for in situ cancer vaccines. We also provide our perspective on this emerging field that lies at the interface of cancer vaccine biology and delivery technologies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, Center for BioAnalytical Chemistry, Hefei National Research Center for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Rakan El-Mayta
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Neefjes J, Gurova K, Sarthy J, Szabó G, Henikoff S. Chromatin as an old and new anticancer target. Trends Cancer 2024; 10:696-707. [PMID: 38825423 PMCID: PMC11479676 DOI: 10.1016/j.trecan.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024]
Abstract
Recent genome-wide analyses identified chromatin modifiers as one of the most frequently mutated classes of genes across all cancers. However, chemotherapies developed for cancers involving DNA damage remain the standard of care for chromatin-deranged malignancies. In this review we address this conundrum by establishing the concept of 'chromatin damage': the non-genetic damage to protein-DNA interactions induced by certain small molecules. We highlight anthracyclines, a class of chemotherapeutic agents ubiquitously applied in oncology, as an example of overlooked chromatin-targeting agents. We discuss our current understanding of this phenomenon and explore emerging chromatin-damaging agents as a basis for further studies to maximize their impact in modern cancer treatment.
Collapse
Affiliation(s)
- Jacques Neefjes
- Department of Cell and Chemical Biology and Oncode Institute, LUMC, Einthovenweg 20, 2333, ZC, Leiden, The Netherlands
| | - Katerina Gurova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| | - Jay Sarthy
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, 1920 Terry Ave, Seattle, WA 98109, USA
| | - Gábor Szabó
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, Egyetem tér 1, 4032, Hungary
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
13
|
Mentucci FM, Romero Nuñez EA, Ercole A, Silvetti V, Dal Col J, Lamberti MJ. Impact of Genomic Mutation on Melanoma Immune Microenvironment and IFN-1 Pathway-Driven Therapeutic Responses. Cancers (Basel) 2024; 16:2568. [PMID: 39061208 PMCID: PMC11274745 DOI: 10.3390/cancers16142568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The BRAFV600E mutation, found in approximately 50% of melanoma cases, plays a crucial role in the activation of the MAPK/ERK signaling pathway, which promotes tumor cell proliferation. This study aimed to evaluate its impact on the melanoma immune microenvironment and therapeutic responses, particularly focusing on immunogenic cell death (ICD), a pivotal cytotoxic process triggering anti-tumor immune responses. Through comprehensive in silico analysis of the Cancer Genome Atlas data, we explored the association between the BRAFV600E mutation, immune subtype dynamics, and tumor mutation burden (TMB). Our findings revealed that the mutation correlated with a lower TMB, indicating a reduced generation of immunogenic neoantigens. Investigation into immune subtypes reveals an exacerbation of immunosuppression mechanisms in BRAFV600E-mutated tumors. To assess the response to ICD inducers, including doxorubicin and Me-ALA-based photodynamic therapy (PDT), compared to the non-ICD inducer cisplatin, we used distinct melanoma cell lines with wild-type BRAF (SK-MEL-2) and BRAFV600E mutation (SK-MEL-28, A375). We demonstrated a differential response to PDT between the WT and BRAFV600E cell lines. Further transcriptomic analysis revealed upregulation of IFNAR1, IFNAR2, and CXCL10 genes associated with the BRAFV600E mutation, suggesting their involvement in ICD. Using a gene reporter assay, we showed that PDT robustly activated the IFN-1 pathway through cGAS-STING signaling. Collectively, our results underscore the complex interplay between the BRAFV600E mutation and immune responses, suggesting a putative correlation between tumors carrying the mutation and their responsiveness to therapies inducing the IFN-1 pathway, such as the ICD inducer PDT, possibly mediated by the elevated expression of IFNAR1/2 receptors.
Collapse
Affiliation(s)
- Fátima María Mentucci
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Elisa Ayelén Romero Nuñez
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Agustina Ercole
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Valentina Silvetti
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy;
| | - María Julia Lamberti
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| |
Collapse
|
14
|
Elzoghby AO, Samir O, Emam HE, Soliman A, Abdelgalil RM, Elmorshedy YM, Elkhodairy KA, Nasr ML. Engineering nanomedicines for immunogenic eradication of cancer cells: Recent trends and synergistic approaches. Acta Pharm Sin B 2024; 14:2475-2504. [PMID: 38828160 PMCID: PMC11143780 DOI: 10.1016/j.apsb.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/07/2024] [Accepted: 03/09/2024] [Indexed: 06/05/2024] Open
Abstract
Resistance to cancer immunotherapy is mainly attributed to poor tumor immunogenicity as well as the immunosuppressive tumor microenvironment (TME) leading to failure of immune response. Numerous therapeutic strategies including chemotherapy, radiotherapy, photodynamic, photothermal, magnetic, chemodynamic, sonodynamic and oncolytic therapy, have been developed to induce immunogenic cell death (ICD) of cancer cells and thereby elicit immunogenicity and boost the antitumor immune response. However, many challenges hamper the clinical application of ICD inducers resulting in modest immunogenic response. Here, we outline the current state of using nanomedicines for boosting ICD of cancer cells. Moreover, synergistic approaches used in combination with ICD inducing nanomedicines for remodeling the TME via targeting immune checkpoints, phagocytosis, macrophage polarization, tumor hypoxia, autophagy and stromal modulation to enhance immunogenicity of dying cancer cells were analyzed. We further highlight the emerging trends of using nanomaterials for triggering amplified ICD-mediated antitumor immune responses. Endoplasmic reticulum localized ICD, focused ultrasound hyperthermia, cell membrane camouflaged nanomedicines, amplified reactive oxygen species (ROS) generation, metallo-immunotherapy, ion modulators and engineered bacteria are among the most innovative approaches. Various challenges, merits and demerits of ICD inducer nanomedicines were also discussed with shedding light on the future role of this technology in improving the outcomes of cancer immunotherapy.
Collapse
Affiliation(s)
- Ahmed O. Elzoghby
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Omar Samir
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Hagar E. Emam
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Ahmed Soliman
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Riham M. Abdelgalil
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Yomna M. Elmorshedy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Kadria A. Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mahmoud L. Nasr
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| |
Collapse
|
15
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Naumenko VA, Chekhonin VP. The need for paradigm shift: prognostic significance and implications of standard therapy-related systemic immunosuppression in glioblastoma for immunotherapy and oncolytic virotherapy. Front Immunol 2024; 15:1326757. [PMID: 38390330 PMCID: PMC10881776 DOI: 10.3389/fimmu.2024.1326757] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Despite significant advances in our knowledge regarding the genetics and molecular biology of gliomas over the past two decades and hundreds of clinical trials, no effective therapeutic approach has been identified for adult patients with newly diagnosed glioblastoma, and overall survival remains dismal. Great hopes are now placed on combination immunotherapy. In clinical trials, immunotherapeutics are generally tested after standard therapy (radiation, temozolomide, and steroid dexamethasone) or concurrently with temozolomide and/or steroids. Only a minor subset of patients with progressive/recurrent glioblastoma have benefited from immunotherapies. In this review, we comprehensively discuss standard therapy-related systemic immunosuppression and lymphopenia, their prognostic significance, and the implications for immunotherapy/oncolytic virotherapy. The effectiveness of immunotherapy and oncolytic virotherapy (viro-immunotherapy) critically depends on the activity of the host immune cells. The absolute counts, ratios, and functional states of different circulating and tumor-infiltrating immune cell subsets determine the net immune fitness of patients with cancer and may have various effects on tumor progression, therapeutic response, and survival outcomes. Although different immunosuppressive mechanisms operate in patients with glioblastoma/gliomas at presentation, the immunological competence of patients may be significantly compromised by standard therapy, exacerbating tumor-related systemic immunosuppression. Standard therapy affects diverse immune cell subsets, including dendritic, CD4+, CD8+, natural killer (NK), NKT, macrophage, neutrophil, and myeloid-derived suppressor cell (MDSC). Systemic immunosuppression and lymphopenia limit the immune system's ability to target glioblastoma. Changes in the standard therapy are required to increase the success of immunotherapies. Steroid use, high neutrophil-to-lymphocyte ratio (NLR), and low post-treatment total lymphocyte count (TLC) are significant prognostic factors for shorter survival in patients with glioblastoma in retrospective studies; however, these clinically relevant variables are rarely reported and correlated with response and survival in immunotherapy studies (e.g., immune checkpoint inhibitors, vaccines, and oncolytic viruses). Our analysis should help in the development of a more rational clinical trial design and decision-making regarding the treatment to potentially improve the efficacy of immunotherapy or oncolytic virotherapy.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Victor A. Naumenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
16
|
Lasser SA, Ozbay Kurt FG, Arkhypov I, Utikal J, Umansky V. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat Rev Clin Oncol 2024; 21:147-164. [PMID: 38191922 DOI: 10.1038/s41571-023-00846-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
Anticancer agents continue to dominate the list of newly approved drugs, approximately half of which are immunotherapies. This trend illustrates the considerable promise of cancer treatments that modulate the immune system. However, the immune system is complex and dynamic, and can have both tumour-suppressive and tumour-promoting effects. Understanding the full range of immune modulation in cancer is crucial to identifying more effective treatment strategies. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid cells that develop in association with chronic inflammation, which is a hallmark of cancer. Indeed, MDSCs accumulate in the tumour microenvironment, where they strongly inhibit anticancer functions of T cells and natural killer cells and exert a variety of other tumour-promoting effects. Emerging evidence indicates that MDSCs also contribute to resistance to cancer treatments, particularly immunotherapies. Conversely, treatment approaches designed to eliminate cancer cells can have important additional effects on MDSC function, which can be either positive or negative. In this Review, we discuss the interplay between MDSCs and various other cell types found in tumours as well as the mechanisms by which MDSCs promote tumour progression. We also discuss the relevance and implications of MDSCs for cancer therapy.
Collapse
Affiliation(s)
- Samantha A Lasser
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Feyza G Ozbay Kurt
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Ihor Arkhypov
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Jochen Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany.
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany.
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| |
Collapse
|
17
|
Montico B, Nigro A, Lamberti MJ, Martorelli D, Mastorci K, Ravo M, Giurato G, Steffan A, Dolcetti R, Casolaro V, Dal Col J. Phospholipid scramblase 1 is involved in immunogenic cell death and contributes to dendritic cell-based vaccine efficiency to elicit antitumor immune response in vitro. Cytotherapy 2024; 26:145-156. [PMID: 38099895 DOI: 10.1016/j.jcyt.2023.11.014] [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: 06/22/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND AIMS Whole tumor cell lysates (TCLs) obtained from cancer cells previously killed by treatments able to promote immunogenic cell death (ICD) can be efficiently used as a source of tumor-associated antigens for the development of highly efficient dendritic cell (DC)-based vaccines. Herein, the potential role of the interferon (IFN)-inducible protein phospholipid scramblase 1 (PLSCR1) in influencing immunogenic features of dying cancer cells and in enhancing DC-based vaccine efficiency was investigated. METHODS PLSCR1 expression was evaluated in different mantle-cell lymphoma (MCL) cell lines following ICD induction by 9-cis-retinoic acid (RA)/IFN-α combination, and commercial kinase inhibitor was used to identify the signaling pathway involved in its upregulation. A Mino cell line ectopically expressing PLSCR1 was generated to investigate the potential involvement of this protein in modulating ICD features. Whole TCLs obtained from Mino overexpressing PLSCR1 were used for DC loading, and loaded DCs were employed for generation of tumor antigen-specific cytotoxic T lymphocytes. RESULTS The ICD inducer RA/IFN-α combination promoted PLSCR1 expression through STAT1 activation. PLSCR1 upregulation favored pro-apoptotic effects of RA/IFN-α treatment and enhanced the exposure of calreticulin on cell surface. Moreover, DCs loaded with TCLs obtained from Mino ectopically expressing PLSCR1 elicited in vitro greater T-cell-mediated antitumor responses compared with DCs loaded with TCLs derived from Mino infected with empty vector or the parental cell line. Conversely, PLSCR1 knock-down inhibited the stimulating activity of DCs loaded with RA/IFN-α-treated TCLs to elicit cyclin D1 peptide-specific cytotoxic T lymphocytes. CONCLUSIONS Our results indicate that PLSCR1 improved ICD-associated calreticulin exposure induced by RA/IFN-α and was clearly involved in DC-based vaccine efficiency as well, suggesting a potential contribution in the control of pathways associated to DC activation, possibly including those involved in antigen uptake and concomitant antitumor immune response activation.
Collapse
Affiliation(s)
- Barbara Montico
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| | - Annunziata Nigro
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy.
| | - Maria Julia Lamberti
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina.
| | - Debora Martorelli
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| | - Katy Mastorci
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| | - Maria Ravo
- Genomix4Life Srl, Baronissi, Salerno, Italy.
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Salerno, Italy.
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| | - Riccardo Dolcetti
- Centre for Cancer Immunotherapy, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia; Faculty of Medicine, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia.
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy.
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy.
| |
Collapse
|
18
|
Shi Y, Wu Y, Li F, Jiang K, Fang X, Wang Y, Song X, Wang R, Chen L, Zheng J, Wu C, Qin Y, Liu X, Liu S. Investigating the Immunogenic Cell Death-Dependent Subtypes and Prognostic Signature of Triple-Negative Breast Cancer. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:34-45. [PMID: 38605910 PMCID: PMC11003942 DOI: 10.1007/s43657-023-00133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 04/13/2024]
Abstract
Recently, immunotherapy has emerged as a promising and effective method for treating triple-negative breast cancer (TNBC). However, challenges still persist. Immunogenic cell death (ICD) is considered a prospective treatment and potential combinational treatment strategy as it induces an anti-tumor immune response by presenting the antigenic epitopes of dead cells. Nevertheless, the ICD process in TNBC and its impact on disease progression and the response to immunotherapy are not well understood. In this study, we observed dysregulation of the ICD process and verified the altered expression of prognostic ICD genes in TNBC through quantitative real-time polymerase chain reaction (qRT-PCR) analysis. To investigate the potential role of the ICD process in TNBC progression, we determined the ICD-dependent subtypes, and two were identified. Analysis of their distinct tumor immune microenvironment (TIME) and cancer hallmark features revealed that Cluster 1 and 2 corresponded to the immune "cold" and "hot" phenotypes, respectively. In addition, we constructed the prognostic signature ICD score of TNBC patients and demonstrated its clinical independence and generalizability. The ICD score could also serve as a potential biomarker for immune checkpoint blockade and may aid in the identification of targeted effective agents for individualized clinical strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-023-00133-x.
Collapse
Affiliation(s)
- Youyang Shi
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Yuanyuan Wu
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Feifei Li
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Kexin Jiang
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Xiaofang Fang
- First Clinical College, Shandong University of Traditional Chinese Medicine, No. 16369, Jingshi Road, Lixia District, Jinan, 250011 Shandong China
| | - Yu Wang
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Xiaoyun Song
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Rui Wang
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Lixin Chen
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Jinzhou Zheng
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Chunyu Wu
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Yuenong Qin
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| | - Xiaofei Liu
- First Clinical College, Shandong University of Traditional Chinese Medicine, No. 16369, Jingshi Road, Lixia District, Jinan, 250011 Shandong China
| | - Sheng Liu
- Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Wanping South Road, 725, Shanghai, 200033 China
| |
Collapse
|
19
|
Kwon S, Meng F, Tamam H, Gadalla HH, Wang J, Dong B, Hopf Jannasch AS, Ratliff TL, Yeo Y. Systemic Delivery of Paclitaxel by Find-Me Nanoparticles Activates Antitumor Immunity and Eliminates Tumors. ACS NANO 2024; 18:3681-3698. [PMID: 38227965 PMCID: PMC11025439 DOI: 10.1021/acsnano.3c11445] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Local delivery of immune-activating agents has shown promise in overcoming an immunosuppressive tumor microenvironment (TME) and stimulating antitumor immune responses in tumors. However, systemic therapy is ultimately needed to treat tumors that are not readily locatable or accessible. To enable systemic delivery of immune-activating agents, we employ poly(lactic-co-glycolide) (PLGA) nanoparticles (NPs) with a track record in systemic application. The surface of PLGA NPs is decorated with adenosine triphosphate (ATP), a damage-associated molecular pattern to recruit antigen-presenting cells (APCs). The ATP-conjugated PLGA NPs (NPpD-ATP) are loaded with paclitaxel (PTX), a chemotherapeutic agent inducing immunogenic cell death to generate tumor antigens in situ. We show that the NPpD-ATP retains ATP activity in hostile TME and provides a stable "find-me" signal to recruit APCs. Therefore, the PTX-loaded NPpD-ATP helps populate antitumor immune cells in TME and attenuate the growth of CT26 and B16F10 tumors better than a mixture of PTX-loaded NPpD and ATP. Combined with anti-PD-1 antibody, PTX-loaded NPpD-ATP achieves complete regression of CT26 tumors followed by antitumor immune memory. This study demonstrates the feasibility of systemic immunotherapy using a PLGA NP formulation that delivers ICD-inducing chemotherapy and an immunostimulatory signal.
Collapse
Affiliation(s)
- Soonbum Kwon
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Fanfei Meng
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Hassan Tamam
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Hytham H. Gadalla
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Jianping Wang
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Boyang Dong
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Amber S. Hopf Jannasch
- Metabolite Profiling Facility, Bindley Bioscience Center, Purdue University, 1203 Mitch Daniels Blvd., West Lafayette, IN 47907, USA
| | - Timothy L. Ratliff
- Purdue University Institute for Cancer Research, 201 South University Street, West Lafayette, IN, 47907, USA
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, IN, 47907, USA
| | - Yoon Yeo
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Purdue University Institute for Cancer Research, 201 South University Street, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, West Lafayette, IN 47907, USA
| |
Collapse
|
20
|
Wang R, Hu Q, Huang S, Fang Y, Kong X, Kaur P, Zhang J, Wang Y, Liu D, Wu H, Li Y, Ji J, Yang X, Ye L, Zhai G. Zwitterionic Injectable Hydrogel-Combined Chemo- and Immunotherapy Medicated by Monomolecular Micelles to Effectively Prevent the Recurrence of Tumor Post Operation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4071-4088. [PMID: 38194589 DOI: 10.1021/acsami.3c17017] [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: 01/11/2024]
Abstract
Surgical resection remains the most common method of tumor treatment; however, the high recurrence and metastasis after surgery need to be solved urgently. Herein, we report an injectable zwitterionic hydrogel based on "thiol-ene" click chemistry containing doxorubicin (DOX) and a macrophage membrane (MM)-coated 1-methyl-tryptophan (1-MT)-loaded polyamide-amine dendrimer (P-DOX/1MT) for preventing the postoperative recurrence of tumors. The results indicated that P-DOX/1MT@MM exhibited enhanced recognition and uptake of the dendrimer by tumor cells and induced the immunogenic cell death. In the mice tumor model, the P-DOX/1MT@MM-Gel exhibited high therapeutic efficiency, which could significantly reduce the recurrence of the tumor, including suppressing tumor growth, promoting dendritic cell maturation, and increasing tumor-infiltrating cytotoxic T lymphocytes. The mechanism analysis revealed that the hydrogel greatly reduces the side effects to normal tissues and significantly improves its therapeutic effect. 1MT in the hydrogel is released more rapidly, improving the tumor suppressor microenvironment and increasing the tumor cell sensitivity to DOX. Then, the DOX in the P-DOX/1MT@MM effectively eliminatedo the residual tumor cells and exerted enhanced toxicity. In conclusion, this novel injectable hydrogel that combines chemotherapy and immunotherapy has the property of sequential drug release and is a promising strategy for preventing the postoperative recurrence of tumors.
Collapse
Affiliation(s)
- Rong Wang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qiaoying Hu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Susu Huang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yuelin Fang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinru Kong
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Prabhleen Kaur
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Jicheng Zhang
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Yanqing Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dongzhu Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hang Wu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yingying Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaoye Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Ye
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Guangxi Zhai
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
21
|
Gao H, Ouyang D, Guan X, Xu J, Chen Q, Zeng L, Pang J, Zou Q, Qian K, Yi W. Immune characteristics and clinical significance of peripheral blood lymphocytes in breast cancer. BMC Cancer 2024; 24:50. [PMID: 38195475 PMCID: PMC10775541 DOI: 10.1186/s12885-024-11815-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/01/2024] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND In the context of breast cancer (BC), the correlation between lymphocytes and clinical outcomes, along with treatment response, has garnered attention. Despite this, few investigations have delved into the interplay among distinct peripheral blood lymphocyte (PBL) types, immune attributes, and their clinical implications within the BC landscape. METHODS The primary objective of this study was to scrutinize the baseline status of PBL subsets in patients with primary BC, track their dynamic changes throughout treatment, and ascertain their interrelation with prognosis. Flow cytometry was employed to analyse PBLs from a cohort of 74 BC patients. RESULTS Our analysis revealed that baseline levels of Treg and PD-L1 + T cells were lower in BC patients compared to the reference values. Notably, a disparity in baseline PD-L1 + T cell levels surfaced between patients who underwent adjuvant therapy and those subjected to neoadjuvant therapy (NAT). Furthermore, a meticulous evaluation of PBL subsets before and after treatment underscored discernible alterations in 324 + T cells and CD19 + CD32 + B cells over the course of therapy. Strikingly, heightened CD4 + T cell levels at baseline were linked to enhanced event-free survival (EFS) (p = 0.02) and a robust response to chemotherapy. CONCLUSIONS These results indicate that PBLs may serve as a significant marker to assess the immune status of BC patients, and therapy has the potential to modify patient immune profiles. In addition, peripheral blood CD4 + T cell levels may serve as promising biomarkers for diagnosis and prognosis in future studies of BC.
Collapse
Affiliation(s)
- Hongyu Gao
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
| | - Dengjie Ouyang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Xinyu Guan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
| | - Jiachi Xu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
| | - Qitong Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
| | - Liyun Zeng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
| | - Jian Pang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
| | - Qiongyan Zou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China
| | - Ke Qian
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China.
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China.
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 People's Road, Changsha, Hunan, 410011, P.R. China.
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, 410011, China.
| |
Collapse
|
22
|
Ramon J, Engelen Y, De Keersmaecker H, Goemaere I, Punj D, Mejía Morales J, Bonte C, Berx G, Hoste E, Stremersch S, Lentacker I, De Smedt SC, Raemdonck K, Braeckmans K. Laser-induced vapor nanobubbles for B16-F10 melanoma cell killing and intracellular delivery of chemotherapeutics. J Control Release 2024; 365:1019-1036. [PMID: 38065413 DOI: 10.1016/j.jconrel.2023.12.006] [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: 10/07/2022] [Revised: 11/23/2023] [Accepted: 12/02/2023] [Indexed: 12/25/2023]
Abstract
The most lethal form of skin cancer is cutaneous melanoma, a tumor that develops in the melanocytes, which are found in the epidermis. The treatment strategy of melanoma is dependent on the stage of the disease and often requires combined local and systemic treatment. Over the years, systemic treatment of melanoma has been revolutionized and shifted toward immunotherapeutic approaches. Phototherapies like photothermal therapy (PTT) have gained considerable attention in the field, mainly because of their straightforward applicability in melanoma skin cancer, combined with the fact that these strategies are able to induce immunogenic cell death (ICD), linked with a specific antitumor immune response. However, PTT comes with the risk of uncontrolled heating of the surrounding healthy tissue due to heat dissipation. Here, we used pulsed laser irradiation of endogenous melanin-containing melanosomes to induce cell killing of B16-F10 murine melanoma cells in a non-thermal manner. Pulsed laser irradiation of the B16-F10 cells resulted in the formation of water vapor nanobubbles (VNBs) around endogenous melanin-containing melanosomes, causing mechanical cell damage. We demonstrated that laser-induced VNBs are able to kill B16-F10 cells with high spatial resolution. When looking more deeply into the cell death mechanism, we found that a large part of the B16-F10 cells succumbed rapidly after pulsed laser irradiation, reaching maximum cell death already after 4 h. Practically all necrotic cells demonstrated exposure of phosphatidylserine on the plasma membrane and caspase-3/7 activity, indicative of regulated cell death. Furthermore, calreticulin, adenosine triphosphate (ATP) and high-mobility group box 1 (HMGB1), three key damage-associated molecular patterns (DAMPs) in ICD, were found to be exposed from B16-F10 cells upon pulsed laser irradiation to an extent that exceeded or was comparable to the bona fide ICD-inducer, doxorubicin. Finally, we could demonstrate that VNB formation from melanosomes induced plasma membrane permeabilization. This allowed for enhanced intracellular delivery of bleomycin, an ICD-inducing chemotherapeutic, which further boosted cell death with the potential to improve the systemic antitumor immune response.
Collapse
Affiliation(s)
- Jana Ramon
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| | - Yanou Engelen
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Herlinde De Keersmaecker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Light Microscopy Core Facility, Ghent University, 9000 Ghent, Belgium.
| | - Ilia Goemaere
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium.
| | - Deep Punj
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium.
| | - Julián Mejía Morales
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium.
| | - Cédric Bonte
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium.
| | - Geert Berx
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; VIB Center for Inflammation Research, 9052 Ghent, Belgium; Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| | - Esther Hoste
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| | - Stephan Stremersch
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
23
|
Huang X, Ren Q, Yang L, Cui D, Ma C, Zheng Y, Wu J. Immunogenic chemotherapy: great potential for improving response rates. Front Oncol 2023; 13:1308681. [PMID: 38125944 PMCID: PMC10732354 DOI: 10.3389/fonc.2023.1308681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
The activation of anti-tumor immunity is critical in treating cancers. Recent studies indicate that several chemotherapy agents can stimulate anti-tumor immunity by inducing immunogenic cell death and durably eradicate tumors. This suggests that immunogenic chemotherapy holds great potential for improving response rates. However, chemotherapy in practice has only had limited success in inducing long-term survival or cure of cancers when used either alone or in combination with immunotherapy. We think that this is because the importance of dose, schedule, and tumor model dependence of chemotherapy-activated anti-tumor immunity is under-appreciated. Here, we review immune modulation function of representative chemotherapy agents and propose a model of immunogenic chemotherapy-induced long-lasting responses that rely on synergetic interaction between killing tumor cells and inducing anti-tumor immunity. We comb through several chemotherapy treatment schedules, and identify the needs for chemotherapy dose and schedule optimization and combination therapy with immunotherapy when chemotherapy dosage or immune responsiveness is too low. We further review tumor cell intrinsic factors that affect the optimal chemotherapy dose and schedule. Lastly, we review the biomarkers indicating responsiveness to chemotherapy and/or immunotherapy treatments. A deep understanding of how chemotherapy activates anti-tumor immunity and how to monitor its responsiveness can lead to the development of more effective chemotherapy or chemo-immunotherapy, thereby improving the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Xiaojun Huang
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qinghuan Ren
- Alberta Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Leixiang Yang
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Di Cui
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenyang Ma
- Department of Internal Medicine of Traditional Chinese Medicine, The Second People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Yueliang Zheng
- Cancer Center, Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Junjie Wu
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Bikorimana J, El‐Hachem N, Moreau M, Lawson C, Tai L, Gonçalves M, Abusarah J, Beaudoin S, Stanga D, Plouffe S, Rafei M. Intratumoral administration of unconjugated Accum™ impairs the growth of pre-established solid lymphoma tumors. Cancer Sci 2023; 114:4499-4510. [PMID: 37776054 PMCID: PMC10728015 DOI: 10.1111/cas.15985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/25/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023] Open
Abstract
The Accum™ technology was initially designed to enhance the bioaccumulation of a given molecule in target cells. It does so by triggering endosomal membrane damages allowing endocytosed products to enter the cytosol, escaping the harsh environmental cues of the endosomal lumen. In an attempt to minimize manufacturing hurdles associated with Accum™ conjugation, we tested whether free Accum™ admixed with antigens could lead to outcomes similar to those obtained with conjugated products. Surprisingly, unconjugated Accum™ was found to promote cell death in vitro, an observation further confirmed on various murine tumor cell lines (EL4, CT-26, B16, and 4 T1). At the molecular level, unconjugated Accum™ triggers the production of reactive oxygen species and elicits immunogenic cell death while retaining its innate ability to cause endosomal damages. When administered as a monotherapy to animals with pre-established EL4 T-cell lymphoma, Accum™ controlled tumor growth in a dose-dependent manner, and its therapeutic effect relies on CD4 and CD8 T cells. Although unconjugated Accum™ synergizes with various immune checkpoint inhibitors (anti-CTLA4, anti-PD-1, or anti-CD47) at controlling tumor growth, its therapeutic potency could not be further enhanced when combined with all three tested immune checkpoint inhibitors at once due to its dependency on a specific dosing regimen. In sum, we report in this study an unprecedented new function for unconjugated Accum™ as a novel anticancer molecule. These results could pave the path for a new line of investigation aimed at exploring the pro-killing properties of additional Accum™ variants as a mean to develop second-generation anticancer therapeutics.
Collapse
Affiliation(s)
- Jean‐Pierre Bikorimana
- Department of Microbiology, Infectious Diseases and ImmunologyUniversité de MontréalMontréalQuebecCanada
| | - Nehme El‐Hachem
- Pediatric Hematology‐Oncology DivisionCentre Hospitalier Universitaire Sainte‐Justine Research CentreMontrealQuebecCanada
| | - Mathilde Moreau
- Department of Immunology and Cell Biology, Faculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecCanada
| | - Christine Lawson
- Department of Immunology and Cell Biology, Faculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecCanada
| | - Lee‐Hwa Tai
- Department of Immunology and Cell Biology, Faculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecCanada
| | - Marina Gonçalves
- Department of Molecular BiologyUniversité de MontréalMontréalQuebecCanada
| | - Jamilah Abusarah
- Department of Pharmacology and PhysiologyUniversité de MontréalMontrealQuebecCanada
| | - Simon Beaudoin
- Research and Development unitDefence Therapeutics Inc.MontrealQuebecCanada
| | - Daniela Stanga
- Research and Development unitDefence Therapeutics Inc.MontrealQuebecCanada
| | - Sebastien Plouffe
- Research and Development unitDefence Therapeutics Inc.MontrealQuebecCanada
| | - Moutih Rafei
- Department of Microbiology, Infectious Diseases and ImmunologyUniversité de MontréalMontréalQuebecCanada
- Department of Molecular BiologyUniversité de MontréalMontréalQuebecCanada
- Department of Pharmacology and PhysiologyUniversité de MontréalMontrealQuebecCanada
| |
Collapse
|
25
|
Wong RSJ, Ong RJM, Lim JSJ. Immune checkpoint inhibitors in breast cancer: development, mechanisms of resistance and potential management strategies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:768-787. [PMID: 38263984 PMCID: PMC10804393 DOI: 10.20517/cdr.2023.58] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/14/2023] [Accepted: 10/31/2023] [Indexed: 01/25/2024]
Abstract
The use of immune checkpoint inhibitors (ICIs) has increased exponentially in the past decade, although its progress specifically for breast cancer has been modest. The first U.S. Food and Drug Administration approval for ICI in breast cancer came in 2019, eight years after the first-ever approval of an ICI. At present, current indications for ICIs are relevant only to a subset of patients with triple-negative breast cancer, or those displaying high microsatellite instability or deficiency in the mismatch repair protein pathway. With an increasing understanding of the limitations of using ICIs, which stem from breast cancer being innately poorly immunogenic, as well as the presence of various intrinsic and acquired resistance pathways, ongoing trials are evaluating different combination therapies to overcome these barriers. In this review, we aim to describe the development timeline of ICIs and resistance mechanisms limiting their utility, and summarise the available approaches and ongoing trials relevant to overcoming each resistance mechanism.
Collapse
Affiliation(s)
- Rachel SJ Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Rebecca JM Ong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Joline SJ Lim
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
26
|
Chou W, Sun T, Peng N, Wang Z, Chen D, Qiu H, Zhao H. Photodynamic Therapy-Induced Anti-Tumor Immunity: Influence Factors and Synergistic Enhancement Strategies. Pharmaceutics 2023; 15:2617. [PMID: 38004595 PMCID: PMC10675361 DOI: 10.3390/pharmaceutics15112617] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/28/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Photodynamic therapy (PDT) is an approved therapeutic procedure that exerts cytotoxic activity towards tumor cells by activating photosensitizers (PSs) with light exposure to produce reactive oxygen species (ROS). Compared to traditional treatment strategies such as surgery, chemotherapy, and radiation therapy, PDT not only kills the primary tumors, but also effectively suppresses metastatic tumors by activating the immune response. However, the anti-tumor immune effects induced by PDT are influenced by several factors, including the localization of PSs in cells, PSs concentration, fluence rate of light, oxygen concentration, and the integrity of immune function. In this review, we systematically summarize the influence factors of anti-tumor immune effects mediated by PDT. Furthermore, an update on the combination of PDT and other immunotherapy strategies are provided. Finally, the future directions and challenges of anti-tumor immunity induced by PDT are discussed.
Collapse
Affiliation(s)
- Wenxin Chou
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (W.C.); (T.S.); (N.P.); (D.C.)
| | - Tianzhen Sun
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (W.C.); (T.S.); (N.P.); (D.C.)
| | - Nian Peng
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (W.C.); (T.S.); (N.P.); (D.C.)
| | - Zixuan Wang
- Department of Laser Medicine, the First Medical Center, PLA General Hospital, Beijing 100853, China;
| | - Defu Chen
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (W.C.); (T.S.); (N.P.); (D.C.)
| | - Haixia Qiu
- Department of Laser Medicine, the First Medical Center, PLA General Hospital, Beijing 100853, China;
| | - Hongyou Zhao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (W.C.); (T.S.); (N.P.); (D.C.)
| |
Collapse
|
27
|
Yao S, Han Y, Yang M, Jin K, Lan H. It's high-time to re-evaluate the value of induced-chemotherapy for reinforcing immunotherapy in colorectal cancer. Front Immunol 2023; 14:1241208. [PMID: 37920463 PMCID: PMC10619163 DOI: 10.3389/fimmu.2023.1241208] [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: 06/16/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
Immunotherapy has made significant advances in the treatment of colorectal cancer (CRC), revolutionizing the therapeutic landscape and highlighting the indispensable role of the tumor immune microenvironment. However, some CRCs have shown poor response to immunotherapy, prompting investigation into the underlying reasons. It has been discovered that certain chemotherapeutic agents possess immune-stimulatory properties, including the induction of immunogenic cell death (ICD), the generation and processing of non-mutated neoantigens (NM-neoAgs), and the B cell follicle-driven T cell response. Based on these findings, the concept of inducing chemotherapy has been introduced, and the combination of inducing chemotherapy and immunotherapy has become a standard treatment option for certain cancers. Clinical trials have confirmed the feasibility and safety of this approach in CRC, offering a promising method for improving the efficacy of immunotherapy. Nevertheless, there are still many challenges and difficulties ahead, and further research is required to optimize its use.
Collapse
Affiliation(s)
- Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yuejun Han
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Mengxiang Yang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Xu Z, Xu M, Wu X, Guo S, Tian Z, Zhu D, Yang J, Fu J, Li X, Song G, Liu Z, Song X. A Half-Sandwich Ruthenium(II) (N^N) Complex: Inducing Immunogenic Melanoma Cell Death in Vitro and in Vivo. ChemMedChem 2023; 18:e202300131. [PMID: 37226330 DOI: 10.1002/cmdc.202300131] [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: 03/06/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 05/26/2023]
Abstract
Efficacy of clinical chemotherapeutic agents depends not only on direct cytostatic and cytotoxic effects but also involves in eliciting (re)activation of tumour immune effects. One way to provoke long-lasting antitumour immunity is coined as immunogenic cell death (ICD), exploiting the host immune system against tumour cells as a "second hit". Although metal-based antitumour complexes hold promise as potential chemotherapeutic agents, ruthenium (Ru)-based ICD inducers remain sparse. Herein, we report a half-sandwich complex Ru(II) bearing aryl-bis(imino) acenaphthene chelating ligand with ICD inducing properties for melanoma in vitro and in vivo. Complex Ru(II) displays strong anti-proliferative potency and potential cell migration inhibition against melanoma cell lines. Importantly, complex Ru(II) drives the multiple biochemical hallmarks of ICD in melanoma cells, i. e., the elevated expression of calreticulin (CRT), high mobility group box 1 (HMGB1), Hsp70 and secretion of ATP, followed by the decreased expression of phosphorylation of Stat3. In vivo the inhibition of tumour growth in prophylactic tumour vaccination model further confirms that mice with complex Ru(II)-treated dying cells lead to activate adaptive immune responses and anti-tumour immunity by the activation of ICD in melanoma cells. Mechanisms of action studies show that complex Ru(II)-induced ICD could be associated with mitochondrial damage, ER stress and impairment of metabolic status in melanoma cells. We believe that the half-sandwich complex Ru(II) as an ICD inducer in this work will help to design new half-sandwich Ru-based organometallic complexes with immunomodulatory response in melanoma treatments.
Collapse
Affiliation(s)
- Zhishan Xu
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Mengke Xu
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Xueya Wu
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Sheng Guo
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Zhongwei Tian
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Di Zhu
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Jixuan Yang
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Jiyun Fu
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Xi Li
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Guozhen Song
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| | - Zhe Liu
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, Shandong, P. R. China
| | - Xiangfeng Song
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, 453000, Henan, P. R. China
| |
Collapse
|
29
|
Dumontet C, Reichert JM, Senter PD, Lambert JM, Beck A. Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov 2023; 22:641-661. [PMID: 37308581 DOI: 10.1038/s41573-023-00709-2] [Citation(s) in RCA: 278] [Impact Index Per Article: 139.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 06/14/2023]
Abstract
Antibody-drug conjugates (ADCs) combine the specificity of monoclonal antibodies with the potency of highly cytotoxic agents, potentially reducing the severity of side effects by preferentially targeting their payload to the tumour site. ADCs are being increasingly used in combination with other agents, including as first-line cancer therapies. As the technology to produce these complex therapeutics has matured, many more ADCs have been approved or are in late-phase clinical trials. The diversification of antigenic targets as well as bioactive payloads is rapidly broadening the scope of tumour indications for ADCs. Moreover, novel vector protein formats as well as warheads targeting the tumour microenvironment are expected to improve the intratumour distribution or activation of ADCs, and consequently their anticancer activity for difficult-to-treat tumour types. However, toxicity remains a key issue in the development of these agents, and better understanding and management of ADC-related toxicities will be essential for further optimization. This Review provides a broad overview of the recent advances and challenges in ADC development for cancer treatment.
Collapse
Affiliation(s)
- Charles Dumontet
- CRCL INSERM 1052/CNRS 5286, University of Lyon, Hospices Civils de Lyon, Lyon, France.
| | | | | | | | - Alain Beck
- Institut de Recherche Pierre Fabre, CIPF, Saint-Julien-en-Genevois, France
| |
Collapse
|
30
|
Wang L, Luo R, Onyshchenko K, Rao X, Wang M, Menz B, Gaedicke S, Grosu AL, Firat E, Niedermann G. Adding liposomal doxorubicin enhances the abscopal effect induced by radiation/αPD1 therapy depending on tumor cell mitochondrial DNA and cGAS/STING. J Immunother Cancer 2023; 11:e006235. [PMID: 37640480 PMCID: PMC10462948 DOI: 10.1136/jitc-2022-006235] [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] [Accepted: 07/21/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Localized radiotherapy (RT) can cause a T cell-mediated abscopal effect on non-irradiated tumor lesions, especially in combination with immune checkpoint blockade. However, this effect is still clinically rare and improvements are highly desirable. We investigated whether triple combination with a low dose of clinically approved liposomal doxorubicin (Doxil) could augment abscopal responses compared with RT/αPD-1 and Doxil/αPD-1. We also investigated whether the enhanced abscopal responses depended on the mitochondrial DNA (mtDNA)/cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING)/IFN-I pathway. MATERIALS/METHODS We used Doxil in combination with RT and αPD-1 in two tumor models (B16-CD133 melanoma and MC38 colon carcinoma) with mice bearing two tumors, only one of which was irradiated. Mechanistic studies on the role of the mtDNA/cGAS/STING/IFN-I axis were performed using inhibitors and knockout cells in vitro as well as in mice. RESULTS Addition of a single low dose of Doxil to RT and αPD-1 strongly enhanced the RT/αPD-1-induced abscopal effect in both models. Complete cures of non-irradiated tumors were mainly observed in triple-treated mice. Triple therapy induced more cross-presenting dendritic cells (DCs) and more tumor-specific CD8+ T cells than RT/αPD-1 and Doxil/αPD-1, particularly in non-irradiated tumors. Coincubation of Doxil-treated and/or RT-treated tumor cells with DCs enhanced DC antigen cross-presentation which is crucial for inducing CD8+ T cells. CD8+ T cell depletion or implantation of cGAS-deficient or STING-deficient tumor cells abolished the abscopal effect. Doxorubicin-induced/Doxil-induced IFNβ1 markedly depended on the cGAS/STING pathway. Doxorubicin-treated/Doxil-treated tumor cells depleted of mtDNA secreted less IFNβ1, of the related T cell-recruiting chemokine CXCL10, and ATP; coincubation with mtDNA-depleted tumor cells strongly reduced IFNβ1 secretion by DCs. Implantation of mtDNA-depleted tumor cells, particularly at the non-irradiated/abscopal site, substantially diminished the Doxil-enhanced abscopal effect and tumor infiltration by tumor-specific CD8+ T cells. CONCLUSIONS These data show that single low-dose Doxil can substantially enhance the RT/αPD-1-induced abscopal effect, with a strong increase in cross-presenting DCs and CD8+ tumor-specific T cells particularly in abscopal tumors compared with RT/αPD-1 and Doxil/αPD-1. Moreover, they indicate that the mtDNA/cGAS/STING/IFN-I axis is important for the immunogenic/immunomodulatory doxorubicin effects. Our findings may be helpful for the planning of clinical radiochemoimmunotherapy trials in (oligo)metastatic patients.
Collapse
Affiliation(s)
- Liqun Wang
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ren Luo
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kateryna Onyshchenko
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Xi Rao
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Meidan Wang
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Beatrice Menz
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
| | - Simone Gaedicke
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Firat
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
| | - Gabriele Niedermann
- Department of Radiation Oncology, University of Freiburg Faculty of Medicine, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
31
|
Yang X, Yang J, Gu X, Tao Y, Ji H, Miao X, Shen S, Zang H. (-)-Guaiol triggers immunogenic cell death and inhibits tumor growth in non-small cell lung cancer. Mol Cell Biochem 2023; 478:1611-1620. [PMID: 36441354 PMCID: PMC10209243 DOI: 10.1007/s11010-022-04613-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022]
Abstract
(-)-Guaiol is a sesquiterpenoid found in many traditional Chinese medicines with potent antitumor activity. However, its therapeutic effect and mechanism in non-small cell lung cancer (NSCLC) have not been fully elucidated. In this study, (-)-Guaiol was found to induce immunogenic cell death (ICD) in NSCLC in vitro. Using (-)-Guaiol in vivo, we found that (-)-Guaiol could suppress tumor growth, increase dendritic cell activation, and enhance T-cell infiltration. Vaccination experiments suggest that cellular immunoprophylaxis after (-)-Guaiol intervention can suppress tumor growth. Previous studies have found that (-)-Guaiol induces apoptosis and autophagy in NSCLC. Apoptosis and autophagy are closely related to ICD. To explore whether autophagy and apoptosis are involved in (-)-Guaiol-induced ICD, we used inhibitors of apoptosis and autophagy. The results showed that the release of damage-associated molecular patterns (DAMPs) was partly reversed after inhibition of apoptosis and autophagy. In conclusion, these results suggested that the (-)-Guaiol triggers immunogenic cell death and inhibits tumor growth in NSCLC.
Collapse
Affiliation(s)
- Xiaohui Yang
- Department of Oncology, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226000 China
| | - Junling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu Province China
| | - Xiaoxia Gu
- Department of Oncology, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226000 China
| | - Yuhua Tao
- Department of Oncology, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226000 China
| | - Hongjuan Ji
- Department of Oncology, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226000 China
| | - Xian Miao
- Department of Oncology, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226000 China
| | - Shuijie Shen
- Department of Oncology, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226000 China
| | - Haiyang Zang
- Department of Spleen and Stomach, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226000 China
| |
Collapse
|
32
|
Cui J, Xu H, Shi J, Fang K, Liu J, Liu F, Chen Y, Liang H, Zhang Y, Piao H. Carbonic anhydrase IX inhibitor S4 triggers release of DAMPs related to immunogenic cell death in glioma cells via endoplasmic reticulum stress pathway. Cell Commun Signal 2023; 21:167. [PMID: 37386564 PMCID: PMC10311836 DOI: 10.1186/s12964-023-01180-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Immunogenic cell death (ICD), which releases danger-associated molecular patterns (DAMP) that induce potent anticancer immune response, has emerged as a key component of therapy-induced anti-tumor immunity. The aim of this work was to analyze whether the carbonic anhydrase IX inhibitor S4 can elicit ICD in glioma cells. METHODS The effects of S4 on glioma cell growth were evaluated using the CCK-8, clonogenic and sphere assays. Glioma cell apoptosis was determined by flow cytometry. Surface-exposed calreticulin (CRT) was inspected by confocal imaging. The supernatants of S4-treated cells were concentrated for the determination of HMGB1and HSP70/90 expression by immunoblotting. RNA-seq was performed to compare gene expression profiles between S4-treated and control cells. Pharmacological inhibition of apoptosis, autophagy, necroptosis and endoplasmic reticulum (ER) stress was achieved by inhibitors. In vivo effects of S4 were evaluated in glioma xenografts. Immunohistochemistry (IHC) was performed to stain Ki67 and CRT. RESULTS S4 significantly decreased the viability of glioma cells and induced apoptosis and autophagy. Moreover, S4 triggered CRT exposure and the release of HMGB1 and HSP70/90. Inhibition of either apoptosis or autophagy significantly reversed S4-induced release of DAMP molecules. RNA-seq analysis indicated that the ER stress pathway was deregulated upon exposure to S4. Both PERK-eIF2α and IRE1α- XBP1 axes were activated in S4-treated cells. Furthermore, pharmacological inhibition of PERK significantly suppressed S4-triggered ICD markers and autophagy. In glioma xenografts, S4 significantly reduced tumor growth. CONCLUSIONS Altogether, these findings suggest S4 as a novel ICD inducer in glioma and might have implications for S4-based immunotherapy. Video Abstract.
Collapse
Affiliation(s)
- Jing Cui
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Huizhe Xu
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Ji Shi
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Kun Fang
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Jia Liu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Feng Liu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
- Institute of Cancer Stem Cell, Dalian Medical University, No.9 Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Yi Chen
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Haiyang Liang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Ye Zhang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| |
Collapse
|
33
|
Sprooten J, Laureano RS, Vanmeerbeek I, Govaerts J, Naulaerts S, Borras DM, Kinget L, Fucíková J, Špíšek R, Jelínková LP, Kepp O, Kroemer G, Krysko DV, Coosemans A, Vaes RD, De Ruysscher D, De Vleeschouwer S, Wauters E, Smits E, Tejpar S, Beuselinck B, Hatse S, Wildiers H, Clement PM, Vandenabeele P, Zitvogel L, Garg AD. Trial watch: chemotherapy-induced immunogenic cell death in oncology. Oncoimmunology 2023; 12:2219591. [PMID: 37284695 PMCID: PMC10240992 DOI: 10.1080/2162402x.2023.2219591] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/25/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
Immunogenic cell death (ICD) refers to an immunologically distinct process of regulated cell death that activates, rather than suppresses, innate and adaptive immune responses. Such responses culminate into T cell-driven immunity against antigens derived from dying cancer cells. The potency of ICD is dependent on the immunogenicity of dying cells as defined by the antigenicity of these cells and their ability to expose immunostimulatory molecules like damage-associated molecular patterns (DAMPs) and cytokines like type I interferons (IFNs). Moreover, it is crucial that the host's immune system can adequately detect the antigenicity and adjuvanticity of these dying cells. Over the years, several well-known chemotherapies have been validated as potent ICD inducers, including (but not limited to) anthracyclines, paclitaxels, and oxaliplatin. Such ICD-inducing chemotherapeutic drugs can serve as important combinatorial partners for anti-cancer immunotherapies against highly immuno-resistant tumors. In this Trial Watch, we describe current trends in the preclinical and clinical integration of ICD-inducing chemotherapy in the existing immuno-oncological paradigms.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Raquel S. Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Daniel M. Borras
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lisa Kinget
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Jitka Fucíková
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Radek Špíšek
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Lenka Palová Jelínková
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Liguecontre le Cancer, Université de Paris, sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Liguecontre le Cancer, Université de Paris, sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Institut du Cancer Paris CARPEM, Paris, France
| | - Dmitri V. Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Insitute Ghent, Ghent University, Ghent, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Rianne D.W. Vaes
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Steven De Vleeschouwer
- Department Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Department Neuroscience, Laboratory for Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Els Wauters
- Laboratory of Respiratory Diseases and Thoracic Surgery (Breathe), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Sabine Tejpar
- Molecular Digestive Oncology, Department of Oncology, Katholiek Universiteit Leuven, Leuven, Belgium
- Cell Death and Inflammation Unit, VIB-Ugent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Benoit Beuselinck
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Paul M. Clement
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Peter Vandenabeele
- Cell Death and Inflammation Unit, VIB-Ugent Center for Inflammation Research (IRC), Ghent, Belgium
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Laurence Zitvogel
- Tumour Immunology and Immunotherapy of Cancer, European Academy of Tumor Immunology, Gustave Roussy Cancer Center, Inserm, Villejuif, France
| | - Abhishek D. Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Wen Y, You D, Bai Y, Cao J, Zhang L, Xia X. Development and validation of immunogenic cell death-applied prediction model for esophageal carcinoma. Am J Cancer Res 2023; 13:2104-2115. [PMID: 37293167 PMCID: PMC10244091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/13/2023] [Indexed: 06/10/2023] Open
Abstract
Evidence suggests that immunogenic cell death (ICD) releases cancer antigens that promote cytotoxic T-cell responses, potentially improving immunotherapy. However, the relationship between ICDs and esophageal cancer (EC) remains unclear. This study aimed to determine the role of ICDs in EC and to construct an ICD-based prognostic panel. RNA-seq data of EC and the corresponding clinical information were downloaded from the UCSC-Xena platform to explore the association between ICD gene expression and EC prognosis. The GSE53625 dataset was used to validate the proposed model. Differentially expressed genes (DEGs) between different molecular subtypes were identified to construct a new ICD-related prognosis panel and generate molecular subtypes using ConsensusClusterPlus. We created a prognostic profile based on the ICD and a nomogram based on the risk score. Compared with normal samples, ICD gene expression of malignant samples were significantly increased. 161 patients with EC were successfully divided into three subtypes (SubA, SubB, and SubC). Patients with EC in the SubC group had the best survival and lowest ICD score, whereas patients in the SubB group had the worst prognosis. DEGs between subtypes were evaluated, and risk panels were established using LASSO-Cox regression analysis. The prognosis of low-risk patients was significantly better than that of high-risk patients in both cohorts. The area under the curve of the receiver operating characteristic curve indicated that the risk group had a good prognostic value. Our study identified the molecular subtypes of EC and ICD-based prognostic signatures. Our three-gene risk panel could serve as a biomarker for effectively assessing the prognostic risk of patients with EC.
Collapse
Affiliation(s)
- Yazhou Wen
- Department of Anesthesiology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care HospitalNanjing, Jiangsu, China
| | - Dongshan You
- Department of Medical Oncology, The Affiliated Chuzhou Hospital of Anhui Medical University/The First People’s Hospital of ChuzhouChuzhou, Anhui, China
| | - Yongkang Bai
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjing, Jiangsu, China
| | - Jing Cao
- Department of Medical Oncology, The Affiliated Chuzhou Hospital of Anhui Medical University/The First People’s Hospital of ChuzhouChuzhou, Anhui, China
| | - Louqian Zhang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjing, Jiangsu, China
| | - Xiaoyang Xia
- Department of Medical Oncology, The Affiliated Chuzhou Hospital of Anhui Medical University/The First People’s Hospital of ChuzhouChuzhou, Anhui, China
| |
Collapse
|
35
|
Singh R, Srivastava P, Manna PP. Chemokine-targeted nanoparticles: stimulation of the immune system in cancer immunotherapy. EXPLORATION OF IMMUNOLOGY 2023:123-147. [DOI: 10.37349/ei.2023.00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/24/2023] [Indexed: 01/06/2025]
Abstract
Surgery, chemotherapy, radiation therapy, and immunotherapy are potential therapeutic choices for many malignant and metastatic cancers. Despite adverse side effects and pain, surgery and chemotherapy continue to be the most common cancer treatments. However, patients treated with immunotherapy had better cancer control than those who got other treatments. There are two methods to activate immunological pathways: systemically and locally. To modify the tumor microenvironment (TME), the former uses systemic cytokine/chemokine (CK) delivery, whilst the latter uses immunological checkpoints or small molecule inhibitors. Organic and inorganic nanomaterials (NMs) enhanced the efficacy of cancer immunotherapy. NMs can transmit drugs, peptides, antigens, antibodies, whole cell membranes, etc. Surface-modified NMs precisely target and enter the tissues. The inner core of surface-modified NMs is composed of chemicals with limited bioavailability and biocompatibility, resulting in prolonged blood retention and decreased renal clearance. These platforms hinder or prevent many immune cell activities and modify the TME, enhancing the efficiency of cancer immunotherapy. By inhibiting CK/CK receptor signaling, cell migration and other immune responses could be controlled. Developing CK-targeted nanoparticles (NPs) that inhibit CK signaling or take advantage of the ligand-receptor connection is possible. Surface chemical modification of NMs with CKs or specific peptides has several medicinal applications, including tissue-specific drug delivery and limited cell migration in cancer-afflicted conditions. This review covers current developments in the role of different groups of CK-loaded NP in tumor therapy targeting immune cells and cancer. It also covers the role of NP targeting CK signaling which aids in immunogenic cell death (ICD) and induction of antitumor immunity. In addition, CK gene silencing and its capacity to prevent cancer metastasis as well as inhibition of immune cell migration to modulate the TME are discussed.
Collapse
Affiliation(s)
- Ranjeet Singh
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Prateek Srivastava
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India;Current address: Postdoctoral Fellow, Ben-Gurion University of the Negev, Beersheba 8410501, Israel
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
36
|
Nikoo M, Hassan ZF, Mardasi M, Rostamnezhad E, Roozbahani F, Rahimi S, Mohammadi J. Hepatocellular carcinoma (HCC) immunotherapy by anti-PD-1 monoclonal antibodies: A rapidly evolving strategy. Pathol Res Pract 2023; 247:154473. [PMID: 37207558 DOI: 10.1016/j.prp.2023.154473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest cancers in the world, with a high relapse rate. Delayed symptom onset observed in 70-80% of patients leads to diagnosis in advanced stages commonly associated with chronic liver disease. Programmed cell death protein 1 (PD-1) blockade therapy has recently emerged as a promising therapeutic option in the clinical management of several advanced malignancies, including HCC, due to the activation of exhausted tumor-infiltrating lymphocytes and improved outcomes of T-cell function. However, many people with HCC do not respond to PD-1 blockade therapy, and the diversity of immune-related adverse events (irAEs) restricts their clinical utility. Therefore, numerous effective combinatory strategies, including combinations with anti-PD-1 antibodies and other therapeutic methods ranging from chemotherapy to targeted therapies, are evolving to improve therapeutic outcomes and evoke synergistic anti-tumor impressions in patients with advanced HCC. Unfortunately, combined therapy may have more side effects than single-agent treatment. Nonetheless, identifying appropriate predictive biomarkers can aid in managing potential immune-related adverse events by distinguishing patients who respond best to PD-1 inhibitors as single agents or in combination strategies. In the present review, we summarize the therapeutic potential of PD-1 blockade therapy for advanced HCC patients. Besides, a glimpse of the pivotal predictive biomarkers influencing a patient's response to anti-PD-1 antibodies will be provided.
Collapse
Affiliation(s)
- Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Mahsa Mardasi
- Biotechnology Department, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University G. C., Evin, Tehran, Iran
| | - Elmira Rostamnezhad
- Department of Molecular Genetics, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Fatemeh Roozbahani
- Department of Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sahel Rahimi
- Industrial and Environmental Biotechnology Department, National Institute of Genetic Engineering and Biotechnology(NIGEB), Tehran, Iran
| | - Javad Mohammadi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.
| |
Collapse
|
37
|
Kothari N, Postwala H, Pandya A, Shah A, Shah Y, Chorawala MR. Establishing the applicability of cancer vaccines in combination with chemotherapeutic entities: current aspect and achievable prospects. Med Oncol 2023; 40:135. [PMID: 37014489 DOI: 10.1007/s12032-023-02003-y] [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: 02/21/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023]
Abstract
Cancer immunotherapy is one of the recently developed cancer treatment modalities. When compared with conventional anticancer drug regimens, immunotherapy has shown significantly better outcomes in terms of quality of life and overall survival. It incorporates a wide range of immunomodulatory modalities that channel the effects of the immune system either by broadly modulating the host immune system or by accurately targeting distinct tumor antigens. One such treatment modality that has gained interest is cancer vaccine therapy which acts by developing antibodies against tumor cells. Cancer vaccines target individual peptides or groups of antigens that are released by tumor cells and presented by the APCs. This also initiates an effective process to activate the host immune responses. Studies on various types of cancer vaccines are conducted, out of which only few are approved by FDA for clinical uses. Despite of documented safety and efficacy of conventional chemotherapy and cancer vaccines, individually they did not produce substantial results in eradication of the cancer as a monotherapy. Hence, the combination approach holds the extensive potential to provide significant improvement in disease outcomes. Certain chemotherapy has immunomodulatory effects and is proven to synergize with cancer vaccines thereby enhancing their anti-tumor activities. Chemotherapeutic agents are known to have immunostimulatory mechanisms apart from its cytotoxic effect and intensify the anti-tumor activities of vaccines by various mechanisms. This review highlights various cancer vaccines, their mechanism, and how their activity gets affected by chemotherapeutic agents. It also aims at summarizing the evidence-based outcome of the combination approach of a cancer vaccine with chemotherapy and a brief on future aspects.
Collapse
Affiliation(s)
- Nirjari Kothari
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, India
| | - Humzah Postwala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, India
| | - Aanshi Pandya
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, India
| | - Aayushi Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, India
| | - Yesha Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, India.
| |
Collapse
|
38
|
Galluzzi L, Kepp O, Hett E, Kroemer G, Marincola FM. Immunogenic cell death in cancer: concept and therapeutic implications. J Transl Med 2023; 21:162. [PMID: 36864446 PMCID: PMC9979428 DOI: 10.1186/s12967-023-04017-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
Mammalian cells responding to specific perturbations of homeostasis can undergo a regulated variant of cell death that elicits adaptive immune responses. As immunogenic cell death (ICD) can only occur in a precise cellular and organismal context, it should be conceptually differentiated from instances of immunostimulation or inflammatory responses that do not mechanistically depend on cellular demise. Here, we critically discuss key conceptual and mechanistic aspects of ICD and its implications for cancer (immuno)therapy.
Collapse
Affiliation(s)
- 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.
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Erik Hett
- Sonata Therapeutics, Boston, MA, USA
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | | |
Collapse
|
39
|
Lu Y, You J. Strategy and application of manipulating DCs chemotaxis in disease treatment and vaccine design. Biomed Pharmacother 2023; 161:114457. [PMID: 36868016 DOI: 10.1016/j.biopha.2023.114457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
As the most versatile antigen-presenting cells (APCs), dendritic cells (DCs) function as the cardinal commanders in orchestrating innate and adaptive immunity for either eliciting protective immune responses against canceration and microbial invasion or maintaining immune homeostasis/tolerance. In fact, in physiological or pathological conditions, the diversified migratory patterns and exquisite chemotaxis of DCs, prominently manipulate their biological activities in both secondary lymphoid organs (SLOs) as well as homeostatic/inflammatory peripheral tissues in vivo. Thus, the inherent mechanisms or regulation strategies to modulate the directional migration of DCs even could be regarded as the crucial cartographers of the immune system. Herein, we systemically reviewed the existing mechanistic understandings and regulation measures of trafficking both endogenous DC subtypes and reinfused DCs vaccines towards either SLOs or inflammatory foci (including neoplastic lesions, infections, acute/chronic tissue inflammations, autoimmune diseases and graft sites). Furthermore, we briefly introduced the DCs-participated prophylactic and therapeutic clinical application against disparate diseases, and also provided insights into the future clinical immunotherapies development as well as the vaccines design associated with modulating DCs mobilization modes.
Collapse
Affiliation(s)
- Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, 291 Fucheng Road, Zhejiang 310018, PR China; Zhejiang-California International NanoSystems Institute, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
40
|
Miholjcic TBS, Halse H, Bonvalet M, Bigorgne A, Rouanne M, Dercle L, Shankar V, Marabelle A. Rationale for LDH-targeted cancer immunotherapy. Eur J Cancer 2023; 181:166-178. [PMID: 36657325 DOI: 10.1016/j.ejca.2022.11.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022]
Abstract
Immunotherapies have significantly improved the survival of patients in many cancers over the last decade. However, primary and secondary resistances are encountered in most patients. Unravelling resistance mechanisms to cancer immunotherapies is an area of active investigation. Elevated levels of circulating enzyme lactate dehydrogenase (LDH) have been historically considered in oncology as a marker of bad prognosis, usually attributed to elevated tumour burden and cancer metabolism. Recent evidence suggests that elevated LDH levels could be independent from tumour burden and contain a negative predictive value, which could help in guiding treatment strategies in immuno-oncology. In this review, we decipher the rationale supporting the potential of LDH-targeted therapeutic strategies to tackle the direct immunosuppressive effects of LDH on a wide range of immune cells, and enhance the survival of patients treated with cancer immunotherapies.
Collapse
Affiliation(s)
- Tina B S Miholjcic
- Faculté de Médecine, Université de Genève, Genève, Switzerland; Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Heloise Halse
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France; INSERM UMR 1163, Imagine Institute, Université de Paris, F-75015 Paris, France
| | - Mélodie Bonvalet
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France
| | - Amélie Bigorgne
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France; INSERM UMR 1163, Imagine Institute, Université de Paris, F-75015 Paris, France
| | - Mathieu Rouanne
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Gustave Roussy, Villejuif, France; Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Département d'Urologie, Hôpital Foch, UVSQ, Université Paris-Saclay, 92150 Suresnes, France
| | - Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY, USA
| | - Vishnu Shankar
- Immunology Program, School of Medicine, Stanford University, CA, USA
| | - 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, 94805 Villejuif, France; Centre d'Investigation Clinique BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France; Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
41
|
Zhang L, Montesdeoca N, Karges J, Xiao H. Immunogenic Cell Death Inducing Metal Complexes for Cancer Therapy. Angew Chem Int Ed Engl 2023; 62:e202300662. [PMID: 36807420 DOI: 10.1002/anie.202300662] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 02/23/2023]
Abstract
Cancer is one of the deadliest diseases worldwide. Recent statistics have shown that metastases and tumor relapse are the leading causes of cancer-associated deaths. While traditional treatments are able to efficiently remove the primary tumor, secondary tumors remain poorly accessible. Capitalizing on this there is an urgent need for novel treatment modalities. Among the most promising approaches, increasing research interest has been devoted to immunogenic cell death inducing agents that are able to trigger localized cell death of the cancer cells as well as induce an immune response inside the whole organism. Preliminary studies have shown that immunogenic cell death inducing compounds could be able to overcome metastatic and relapsing tumors. Herein, the application of metal complexes as immunogenic cell death inducing compounds is systematically reviewed.
Collapse
Affiliation(s)
- Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
| | - Nicolás Montesdeoca
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
| |
Collapse
|
42
|
Biological Use of Nanostructured Silica-Based Materials Functionalized with Metallodrugs: The Spanish Perspective. Int J Mol Sci 2023; 24:ijms24032332. [PMID: 36768659 PMCID: PMC9917151 DOI: 10.3390/ijms24032332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Since the pioneering work of Vallet-Regí's group on the design and synthesis of mesoporous silica-based materials with therapeutic applications, during the last 15 years, the potential use of mesoporous silica nanostructured materials as drug delivery vehicles has been extensively explored. The versatility of these materials allows the design of a wide variety of platforms that can incorporate numerous agents of interest (fluorophores, proteins, drugs, etc.) in a single scaffold. However, the use of these systems loaded with metallodrugs as cytotoxic agents against different diseases and with distinct therapeutic targets has been studied to a much lesser extent. This review will focus on the work carried out in this field, highlighting both the pioneering and recent contributions of Spanish groups that have synthesized a wide variety of systems based on titanium, tin, ruthenium, copper and silver complexes supported onto nanostructured silica. In addition, this article will also discuss the importance of the structural features of the systems for evaluating and modulating their therapeutic properties. Finally, the most interesting results obtained in the study of the potential therapeutic application of these metallodrug-functionalized silica-based materials against cancer and bacteria will be described, paying special attention to preclinical trials in vivo.
Collapse
|
43
|
Tatarova Z, Blumberg DC, Bensen A, Mills GB, Jonas O. Panobinostat Induced Spatial In Situ Biomarkers Predictive of Anti-PD-1 Efficacy in Mouse Mammary Carcinoma. Cells 2023; 12:308. [PMID: 36672243 PMCID: PMC9856407 DOI: 10.3390/cells12020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
Immunotherapies, including anti-PD-1 immune checkpoint blocking (ICB) antibodies, have revolutionized the treatment of many solid malignancies. However, their efficacy in breast cancer has been limited to a subset of patients with triple-negative breast cancer, where ICBs are routinely combined with a range of cytotoxic and targeted agents. Reliable biomarkers predictive of the therapeutic response to ICB in breast cancer are critically missing, though a combination response has been associated with immunogenic cell death (ICD). Here, we utilized a recently developed integrated analytical platform, the multiplex implantable microdevice assay (MIMA), to evaluate the presence and spatial cell relations of literature-based candidate markers predictive of ICB efficacy in luminal mouse mammary carcinoma. MIMA integrates (i) an implantable microdevice for the localized delivery of small amounts of drugs inside the tumor bed with (ii) sequential multiplex immunohistochemistry (mIHC) and spatial cell analysis pipelines to rapidly (within days) describe drug mechanisms of action and find predictive biomarkers in complex tumor tissue. We show that the expression of cleaved caspase-3, ICAM-1, neuropilin-1, myeloperoxidase, calreticulin, galectin-3, and PD-L1 were spatially associated with the efficacy of panobinostat, a pan-HDAC inhibitor that was previously shown to induce immunogenic cell death and synergize with anti-PD-1 in breast cancer. PD-L1 by itself, however, was not a reliable predictor. Instead, ICB efficacy was robustly identified through the in situ hotspot detection of galectin-3-positive non-proliferating tumor zones enriched in cell death and infiltrated by anti-tumor cytotoxic neutrophils positive for ICAM-1 and neuropilin-1. Such hotspots can be specifically detected using distance-based cluster analyses. Single-cell measurements of the functional states in the tumor microenvironment suggest that both qualitative and quantitative effects might drive effective therapy responses. Overall, the presented study provides (i) complementary biological knowledge about the earliest cell events of induced anti-tumor immunity in breast cancer, including the emergence of resistant cancer stem cells, and (ii) newly identified biomarkers in form of specific spatial cell associations. The approach used standard cell-type-, IHC-, and FFPE-based techniques, and therefore the identified spatial clustering of in situ biomarkers can be readily integrated into existing clinical or research workflows, including in luminal breast cancer. Since early drug responses were detected, the biomarkers could be especially applicable to window-of-opportunity clinical trials to rapidly discriminate between responding and resistant patients, thus limiting unnecessary treatment-associated toxicities.
Collapse
Affiliation(s)
- Zuzana Tatarova
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dylan C. Blumberg
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - AeSoon Bensen
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Gordon B. Mills
- Division of Oncologic Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| | - Oliver Jonas
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
44
|
Tan X, Fang P, Li K, You M, Cao Y, Xu H, Zhu X, Wang L, Wei X, Wen H, Li W, Shi L, Sun X, Yu D, Zhu H, Wang Z, Liu D, Shen H, Zhou W, An M. A HER2-targeted antibody-novel DNA topoisomerase I inhibitor conjugate induces durable adaptive antitumor immunity by activating dendritic cells. MAbs 2023; 15:2220466. [PMID: 37314961 DOI: 10.1080/19420862.2023.2220466] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/16/2023] Open
Abstract
We designed and developed a novel DNA topoisomerase I inhibitor MF-6, which was a more potent cytotoxin and a more potent inducer of immunogenic cell death compared with DXd. To utilize MF-6's ability to induce antitumor immunity, a human epidermal growth factor receptor 2 (HER2)-targeted antibody-drug conjugate (ADC) trastuzumab-L6 that included a cleavable linker and MF-6 was developed. Different from traditional cytotoxic ADC, the antitumor activity of trastuzumab-L6 was assessed by inducing tumor cell immunogenic cell death, activating dendritic cells and cytotoxic CD8+ T cells to acquire durable adaptive immune memory. Tumor cells treated with trastuzumab-L6 were committed to immunogenic cell death, with upregulation of damage-associated molecular patterns and antigen presentation molecules. In a syngeneic tumor model with a mouse cell line that expressed human HER2, immunocompetent mice showed greater antitumor efficacy compared with nude mice. The trastuzumab-L6-cured immunocompetent mice acquired adaptive antitumor memory and rejected subsequent tumor cell challenge. The trastuzumab-L6 efficacy was abrogated when cytotoxic CD8+ T cells were depleted and enhanced when regulatory CD4+ T cells were depleted. The combination of trastuzumab-L6 with immune checkpoint inhibitors significantly increased antitumor efficacy. Enhanced T cell infiltration, dendritic cell activation, and decreased type M2 macrophages in tumor post trastuzumab-L6 administration confirmed the immune-activating responses. In conclusion, trastuzumab-L6 was considered to be an immunostimulatory agent, rather than a traditional cytotoxic ADC, and its antitumor efficacy was enhanced when combined with an anti-PD-L1 and anti-CTLA-4 antibody, which suggested a potential therapeutic strategy.
Collapse
Affiliation(s)
- Xiaoding Tan
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, The People's Republic of China
| | - Peng Fang
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Kaiying Li
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Meng You
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Yuxia Cao
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Hui Xu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Xiaohong Zhu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Lu Wang
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Xin Wei
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Haiying Wen
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Wendi Li
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Lei Shi
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Xiaowei Sun
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Dongan Yu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Huikai Zhu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Zhenzhen Wang
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Datao Liu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Hui Shen
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, The People's Republic of China
| | - Wei Zhou
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd, Taizhou, Jiangsu Province, The People's Republic of China
| | - Maomao An
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, The People's Republic of China
| |
Collapse
|
45
|
Zhai J, Gu X, Liu Y, Hu Y, Jiang Y, Zhang Z. Chemotherapeutic and targeted drugs-induced immunogenic cell death in cancer models and antitumor therapy: An update review. Front Pharmacol 2023; 14:1152934. [PMID: 37153795 PMCID: PMC10160433 DOI: 10.3389/fphar.2023.1152934] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/04/2023] [Indexed: 05/10/2023] Open
Abstract
As traditional strategies for cancer treatment, some chemotherapy agents, such as doxorubicin, oxaliplatin, cyclophosphamide, bortezomib, and paclitaxel exert their anti-tumor effects by inducing immunogenic cell death (ICD) of tumor cells. ICD induces anti-tumor immunity through release of, or exposure to, damage-related molecular patterns (DAMPs), including high mobility group box 1 (HMGB1), calreticulin, adenosine triphosphate, and heat shock proteins. This leads to activation of tumor-specific immune responses, which can act in combination with the direct killing functions of chemotherapy drugs on cancer cells to further improve their curative effects. In this review, we highlight the molecular mechanisms underlying ICD, including those of several chemotherapeutic drugs in inducing DAMPs exposed during ICD to activate the immune system, as well as discussing the prospects for application and potential role of ICD in cancer immunotherapy, with the aim of providing valuable inspiration for future development of chemoimmunotherapy.
Collapse
|
46
|
Han P, Mo S, Wang Z, Xu J, Fu X, Tian Y. UXT at the crossroads of cell death, immunity and neurodegenerative diseases. Front Oncol 2023; 13:1179947. [PMID: 37152054 PMCID: PMC10154696 DOI: 10.3389/fonc.2023.1179947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
The ubiquitous expressed transcript (UXT), a member of the prefoldin-like protein family, modulates regulated cell death (RCD) such as apoptosis and autophagy-mediated cell death through nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), P53, P62, and methylation, and is involved in the regulation of cell metabolism, thereby affecting tumor progression. UXT also maintains immune homeostasis and reduces proteotoxicity in neuro-degenerative diseases through selective autophagy and molecular chaperones. Herein, we review and further elucidate the mechanisms by which UXT affects the regulation of cell death, maintenance of immune homeostasis, and neurodegenerative diseases and discuss the possible UXT involvement in the regulation of ferroptosis and immunogenic cell death, and targeting it to improve cancer treatment outcomes by regulating cell death and immune surveillance.
Collapse
Affiliation(s)
- Pengzhe Han
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Shaojian Mo
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
- Department of Biliary and Pancreatic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Zhengwang Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Jiale Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Xifeng Fu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
- Department of Biliary and Pancreatic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yanzhang Tian
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
- Department of Biliary and Pancreatic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- *Correspondence: Yanzhang Tian,
| |
Collapse
|
47
|
Zhu Y, Zhang S, Lai Y, Pan J, Chen F, Wang T, Wang F, Xu Z, Yang W, Yu H. Self-Cooperative Prodrug Nanovesicles Migrate Immune Evasion to Potentiate Chemoradiotherapy in Head and Neck Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203263. [PMID: 36344430 PMCID: PMC9798966 DOI: 10.1002/advs.202203263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Chemoradiotherapy is the standard of care for the clinical treatment of locally advanced head and neck cancers. However, the combination of ion radiation with free chemotherapeutics yields unsatisfactory therapeutic output and severe side effects due to the nonspecific biodistribution of the anticancer drugs. Herein, a self-cooperative prodrug nanovesicle is reported for highly tumor-specific chemoradiotherapy. The nanovesicles integrating a prodrug of oxaliplatin (OXA) can passively accumulate at the tumor site and penetrate deep into the tumor mass via matrix metalloproteinase 2-mediated cleavage of the polyethylene glycol corona. The OXA prodrug can be restored inside the tumor cells with endogenous glutathione to trigger immunogenic cell death (ICD) of the tumor cells and sensitize the tumor to ion radiation. The nanovesicles can be further loaded with the JAK inhibitor ruxolitinib to abolish chemoradiotherapy-induced programmed death ligand 1 (PD-L1) upregulation on the surface of the tumor cells, thereby prompting chemoradiotherapy-induced immunotherapy by blocking the interferon gamma-Janus kinase-signal transducer and activator of transcription axis. The prodrug nanoplatform reported herein might present a novel strategy to cooperatively enhance chemoradiotherapy of head and cancer and overcome PD-L1-dependent immune evasion.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
- Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Shunan Zhang
- Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Chemistry and Molecular EngineeringEast China Normal UniversityShanghai200241China
| | - Yi Lai
- Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of GastroenterologyHuadong HospitalShanghai Medical CollegeFudan UniversityShanghai200040China
| | - Jiaxing Pan
- Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Fangmin Chen
- Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Tingting Wang
- Department of Medical UltrasoundShanghai Tenth People's Hospital; Tongji UniversityShanghai200072China
| | - Fengyang Wang
- Department of Medical UltrasoundShanghai Tenth People's Hospital; Tongji UniversityShanghai200072China
| | - Zhiai Xu
- School of Chemistry and Molecular EngineeringEast China Normal UniversityShanghai200241China
| | - Wenjun Yang
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Haijun Yu
- Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
48
|
Serrano-Quintero A, Sequeda-Juárez A, Pérez-Hernández CA, Sosa-Delgado SM, Mendez-Tenorio A, Ramón-Gallegos E. Immunogenic analysis of epitope-based vaccine candidate induced by photodynamic therapy in MDA-MB-231 triple-negative breast cancer cells. Photodiagnosis Photodyn Ther 2022; 40:103174. [PMID: 36602069 DOI: 10.1016/j.pdpdt.2022.103174] [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: 08/05/2022] [Revised: 09/20/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Photodynamic therapy (PDT) is used to treat tumors through selective cytotoxic effects. PDT induces damage-associated molecular patterns (DAMPs) expression, which can cause an immunogenic death cell (IDC). In this study we identified potential immunogenic epitopes generated by PDT on triple-negative breast cancer cell line (MDA-MB-231). METHODS MDA-MB-231 cells were exposed to PDT using ALA (160 µg/mL)/630 nm at 8 J/cm2. Membrane proteins were extracted and separated by 2D PAGE. Proteins overexpressed were identified by LC-MS/MS and analyzed in silico through a peptide-HLA docking in order to identify the epitopes with more immunogenicity and antigenicity properties, as well as lower allergenicity and toxicity activity. The selected peptides were evaluated in response to macrophage activation and cytokine release by flow cytometry. RESULTS Differential proteins were overexpressed in the cells treated with PDT. A group of 16 peptides were identified from them, established in a rigorous selection by measuring antigenicity, immunogenicity, allergenicity, and toxicity in silico. The final selection was based on molecular dynamics, where 2 peptides showed the highest stability regarding to the RMSD value. These peptides were obtained from the proteins calreticulin and HSP90. The cytokine analysis evidenced macrophage activation by the releasing of TNF. CONCLUSION Two peptides were identified from calreticulin and HSP90; proteins induced by PDT in MDA-MB-231 cells. Both epitopes showed immunogenic potential as a peptide-based vaccine for triple-negative breast cancer.
Collapse
Affiliation(s)
- Alina Serrano-Quintero
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - Alfonso Sequeda-Juárez
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - C Angélica Pérez-Hernández
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - Sara M Sosa-Delgado
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - Alfonso Mendez-Tenorio
- Laboratorio de Bioinformática y Biotecnología Genómica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Eva Ramón-Gallegos
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico.
| |
Collapse
|
49
|
Fabian KP, Kowalczyk JT, Reynolds ST, Hodge JW. Dying of Stress: Chemotherapy, Radiotherapy, and Small-Molecule Inhibitors in Immunogenic Cell Death and Immunogenic Modulation. Cells 2022; 11:cells11233826. [PMID: 36497086 PMCID: PMC9737874 DOI: 10.3390/cells11233826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/11/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Innovative strategies to re-establish the immune-mediated destruction of malignant cells is paramount to the success of anti-cancer therapy. Accumulating evidence suggests that radiotherapy and select chemotherapeutic drugs and small molecule inhibitors induce immunogenic cell stress on tumors that results in improved immune recognition and targeting of the malignant cells. Through immunogenic cell death, which entails the release of antigens and danger signals, and immunogenic modulation, wherein the phenotype of stressed cells is altered to become more susceptible to immune attack, radiotherapies, chemotherapies, and small-molecule inhibitors exert immune-mediated anti-tumor responses. In this review, we discuss the mechanisms of immunogenic cell death and immunogenic modulation and their relevance in the anti-tumor activity of radiotherapies, chemotherapies, and small-molecule inhibitors. Our aim is to feature the immunological aspects of conventional and targeted cancer therapies and highlight how these therapies may be compatible with emerging immunotherapy approaches.
Collapse
|
50
|
Recent Advances in Cancer Vaccines: Challenges, Achievements, and Futuristic Prospects. Vaccines (Basel) 2022; 10:vaccines10122011. [PMID: 36560420 PMCID: PMC9788126 DOI: 10.3390/vaccines10122011] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a chronic disease, and it can be lethal due to limited therapeutic options. The conventional treatment options for cancer have numerous challenges, such as a low blood circulation time as well as poor solubility of anticancer drugs. Therapeutic cancer vaccines emerged to try to improve anticancer drugs' efficiency and to deliver them to the target site. Cancer vaccines are considered a viable therapeutic technique for most solid tumors. Vaccines boost antitumor immunity by delivering tumor antigens, nucleic acids, entire cells, and peptides. Cancer vaccines are designed to induce long-term antitumor memory, causing tumor regression, eradicate minimal residual illness, and prevent non-specific or unpleasant effects. These vaccines can assist in the elimination of cancer cells from various organs or organ systems in the body, with minimal risk of tumor recurrence or metastasis. Vaccines and antigens for anticancer therapy are discussed in this review, including current vaccine adjuvants and mechanisms of action for various types of vaccines, such as DNA- or mRNA-based cancer vaccines. Potential applications of these vaccines focusing on their clinical use for better therapeutic efficacy are also discussed along with the latest research available in this field.
Collapse
|