1
|
Lei J, Chen J, Chen J, Fang J, Zhou Z, Xu A. Epigallocatechin-3-gallate induces immunogenic cell death and enhances cancer immunotherapy in colorectal cancer. Biochem Biophys Res Commun 2024; 736:150907. [PMID: 39471680 DOI: 10.1016/j.bbrc.2024.150907] [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: 08/15/2024] [Revised: 10/13/2024] [Accepted: 10/24/2024] [Indexed: 11/01/2024]
Abstract
The induction of immunogenic cell death (ICD) can activate antitumor immune response to potentiate cancer immunotherapy. In this study, we observed the antitumor activity following combinatorial therapy with anti-CTLA4 antibody and epigallocatechin-3-gallate (EGCG) in CT26 tumors.Indeed, EGCG triggered colon cancer cells ICD with the secretion of high-mobility group protein B1 (HMGB1) and the surface expression of calreticulin (CRT) and heat shock protein 70 (HSP70). Mice treated with EGCG promoted the maturation of dendritic cells and enhanced the effector function of CD8+ T cells within tumors to remodel the tumor immune microenvironment. Overall, these results indicate that EGCG, a novel ICD inducer, triggers ICD in CRC, and provides a new concept for cancer immunotherapy.
Collapse
Affiliation(s)
- Jun Lei
- Department of Laboratory Medicine, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China; State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.
| | - Jingli Chen
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jian Chen
- Chengdu Academy of Agriculture and Forestry Sciences, Chengdu, China
| | - Jialing Fang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zihao Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.
| | - Aifang Xu
- Department of Laboratory Medicine, Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
2
|
Chen Y, Wang Z, Zhang C, Su Y, Zhou T, Hu K. Revealing the mechanism of natural product-induced immunogenic cell death: opening a new chapter in tumor immunotherapy. Front Immunol 2024; 15:1470071. [PMID: 39445013 PMCID: PMC11496055 DOI: 10.3389/fimmu.2024.1470071] [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: 07/25/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
This review underscores the role of natural products in inducing immunogenic cell death (ICD) as a key strategy in tumor immunotherapy. It reveals that natural products can activate ICD through multiple pathways-apoptosis, autophagy, pyroptosis, and necroptosis-leading to the release of danger-associated molecular patterns (DAMPs), dendritic cell activation, and improved antigen presentation, which together stimulate a potent anti-tumor immune response. The study also demonstrates the enhanced therapeutic potential of combining natural products with immune checkpoint inhibitors. With a focus on translating preclinical findings into clinical practice, this review consolidates recent discoveries and suggests future research paths, offering both theoretical insights and practical guidance for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Yukun Chen
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenzhi Wang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Zhang
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yisa Su
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tian Zhou
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaiwen Hu
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
3
|
Cornillon P, Bouleftour W, Reynaud T, Pigne G, Maillet D, Hamizi S, Beguinot M. Immunogenicity of radiotherapy on bone metastases from prostate adenocarcinoma: What is the future for the combination with radiotherapy/immunotherapy? TUMORI JOURNAL 2024; 110:319-326. [PMID: 38745528 DOI: 10.1177/03008916241249366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Bone metastatic prostate cancers (PCa) are resistant to usual immunotherapies such as checkpoint inhibitors. The main hypothesis related to this immunoresistance is the lack of antigens to stimulate anti-tumor immunity. External radiation is a potential inducer antigens presentation and thus to immunotherapy proprieties. The aim of this review is to describe the tumor microenvironment specificities, especially in bone metastasis and the immune modifications after radiation therapy on a metastatic castration-resistant PCa population. PCa microenvironment is immunosuppressive because of many tumor factors. The complex interplay between PCa cells and bone microenvironment leads to a 'vicious circle' promoting bone metastasis. Furthermore, the immune and bone systems, are connected through an osteoclastogenic cytokine: the Receptor Activator Nuclear Factor Kappa B ligand. Adapted doses of ionizing radiation play a dual role on the tumor. Indeed, radiotherapy leads to immunogenicity by inducing damage associated with molecular patterns. However, it also induces an immunosuppressive effect by increasing the number of immunosuppressive cells. Interestingly, the abscopal effect could be used to optimize immunotherapy potential, especially on bone metastasis. Radiotherapy and immunotherapy combination is a promising strategy, however further studies are necessary to determine the more efficient types of radiation and to control the abscopal effect.
Collapse
Affiliation(s)
- Pierre Cornillon
- Department of Medical Oncology, North Hospital, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Wafa Bouleftour
- Department of Medical Oncology, North Hospital, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Thomas Reynaud
- Department of Radiotherapy, North Hospital, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Gregoire Pigne
- Department of Radiotherapy, North Hospital, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Denis Maillet
- Department of Medical Oncology, IMMUCARE, Centre Hospitalier Lyon Sud, Institut de Cancérologie des Hospices de Lyon, Pierre-Bénite, France
| | - Salima Hamizi
- Department of Medical Oncology, North Hospital, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Marie Beguinot
- Department of Medical Oncology, Medipole Lyon Villeurbanne Mutualist Clinic, Lyon, France
| |
Collapse
|
4
|
Brito TLD, Edson EA, Dias Florêncio KG, Machado-Neto JA, Garnique ADMB, Mesquita Luiz JP, Cunha FDQ, Alves-Filho JC, Haygood M, Wilke DV. Tartrolon D induces immunogenic cell death in melanoma. Chem Biol Interact 2024; 400:111177. [PMID: 39097071 DOI: 10.1016/j.cbi.2024.111177] [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: 02/26/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Tartrolon D (TRL) is produced by Teredinibacter turnerae, a symbiotic cellulose-degrading bacteria in shipworm gills. Immunogenic cell death (ICD) induction contributes to a better and longer-lasting response to anticancer treatment. Tumor cells undergoing ICD trigger activation of the immune system, as a vaccine. AIMS This study aimed to evaluate ICD induction by TRL. MAIN METHODS Cell viability was evaluated by SRB assay. Cell stress, cell death, ICD features and antigen-presenting molecules were evaluated by flow cytometry and immunoblot. KEY FINDINGS TRL showed antiproliferative activity on 7 tumor cell lines (L929, HCT 116, B16-F10, WM293A, SK-MEL-28, PC-3M, and MCF-7) and a non-tumor cell (HEK293A), with an inhibition concentration mean (IC50) ranging from 0.03 μM to 13 μM. Metastatic melanomas, SK-MEL-28, B16-F10, and WM293A, were more sensitive cell lines, with IC50 ranging from 0.07 to 1.2 μM. TRL induced apoptosis along with autophagy and endoplasmic reticulum stress and release of typical damage-associated molecular patterns (DAMPs) of ICD such calreticulin, ERp57, and HSP70 exposure, and HMGB1 release. Additionally, melanoma B16-F10 exposed to TRL increased expression of antigen-presenting molecules MHC II and CD1d and induced activation of splenocytes of C57BL/6 mice. SIGNIFICANCE In spite of recent advances provided by target therapy and immunotherapy, advanced metastatic melanoma is incurable for more than half of patients. ICD inducers yield better and long-lasting responses to anticancer treatment. Our findings shed light on an anticancer candidate of marine origin that induces ICD in melanoma.
Collapse
Affiliation(s)
- Thaís Lima de Brito
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil.
| | - Evelline Araújo Edson
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil.
| | - Katharine Gurgel Dias Florêncio
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil.
| | | | | | - João Paulo Mesquita Luiz
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil.
| | - Fernando de Queiroz Cunha
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil.
| | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil.
| | | | - Diego Veras Wilke
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil.
| |
Collapse
|
5
|
Zhang LL, Zhang DJ, Shi JX, Huang MY, Yu JM, Chen XJ, Wei X, Zou L, Lu JJ. Immunogenic cell death inducers for cancer therapy: An emerging focus on natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155828. [PMID: 38905847 DOI: 10.1016/j.phymed.2024.155828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Immunogenic cell death (ICD) is a specific form of regulated cell death induced by a variety of stressors. During ICD, the dying cancer cells release damage-associated molecular patterns (DAMPs), which promote dendritic cell maturation and tumor antigen presentation, subsequently triggering a T-cell-mediated anti-tumor immune response. In recent years, a growing number of studies have demonstrated the potential of natural products to induce ICD and enhance tumor cell immunogenicity. Moreover, there is an increasing interest in identifying new ICD inducers from natural products. PURPOSE This study aimed to emphasize the potential of natural products and their derivatives as ICD inducers to promote research on using natural products in cancer therapy and provide ideas for future novel immunotherapies based on ICD induction. METHOD This review included a thorough search of the PubMed, Web of Science, Scopus, and Google Scholar databases to identify natural products with ICD-inducing capabilities. A comprehensive search for clinical trials on natural ICD inducers was also conducted using ClinicalTrials.gov, as well as the approved patents using the Espacenet and CNKI Patent Database. RESULTS Natural compounds that induce ICD can be categorized into several groups, such as polyphenols, flavonoids, terpenoids, and alkaloids. Natural products can induce the release of DAMPs by triggering endoplasmic reticulum stress, activation of autophagy-related pathways, and reactive oxygen species generation, etc. Ultimately, they activate anti-tumor immune response and improve the efficacy of cancer treatments. CONCLUSION A growing number of ICD inducers from natural products with promising anti-cancer potential have been identified. The detailed information presented in this review will contribute to the further development of natural ICD inducers and cancer treatment strategies based on ICD-induced responses.
Collapse
Affiliation(s)
- Le-Le Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China
| | - Du-Juan Zhang
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jia-Xin Shi
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jia-Mei Yu
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xu-Jia Chen
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xiao Wei
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
6
|
Malla R, Kumari S, Ganji SP, Srilatha M, Nellipudi HR, Nagaraju GP. Reactive oxygen species of tumor microenvironment: Harnessing for immunogenic cell death. Biochim Biophys Acta Rev Cancer 2024; 1879:189154. [PMID: 39019409 DOI: 10.1016/j.bbcan.2024.189154] [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: 03/01/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
The tumor microenvironment (TME) is a dynamic and complex system that undergoes continuous changes in its network architecture, notably affecting redox homeostasis. These alterations collectively shape a diverse ecosystem actively supporting tumor progression by influencing the cellular and molecular components of the TME. Despite the remarkable clinical advancements in cancer immunotherapy, its spectrum of clinical utility is limited by the altered TME and inadequate tumor immunogenicity. Recent studies have revealed that some conventional and targeted therapy strategies can augment the efficacy of immunotherapy even in patients with less immunogenic solid tumors. These strategies provoke immunogenic cell death (ICD) through the ROS-dependent liberation of damage-associated molecular patterns (DAMPs). These DAMPs recognize and bind with Pattern Recognition Receptors (PRRs) on immune cells, activating and maturing defense cells, ultimately leading to a robust antitumor immune response. The present review underscores the pivotal role of redox homeostasis in orchestrating the transition of TME from a cold to a hot phenotype and the ROS-ICD axis in immune response induction. Additionally, it provides up-to-date insights into strategies that leverage ROS generation to induce ICD. The comprehensive analysis aims to develop ROS-based effective cancer immunotherapies for less immunogenic tumors.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Seema Kumari
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Swapna Priya Ganji
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | | | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
7
|
Han Y, Tian X, Zhai J, Zhang Z. Clinical application of immunogenic cell death inducers in cancer immunotherapy: turning cold tumors hot. Front Cell Dev Biol 2024; 12:1363121. [PMID: 38774648 PMCID: PMC11106383 DOI: 10.3389/fcell.2024.1363121] [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: 12/29/2023] [Accepted: 04/23/2024] [Indexed: 05/24/2024] Open
Abstract
Immunotherapy has emerged as a promising cancer treatment option in recent years. In immune "hot" tumors, characterized by abundant immune cell infiltration, immunotherapy can improve patients' prognosis by activating the function of immune cells. By contrast, immune "cold" tumors are often less sensitive to immunotherapy owing to low immunogenicity of tumor cells, an immune inhibitory tumor microenvironment, and a series of immune-escape mechanisms. Immunogenic cell death (ICD) is a promising cellular process to facilitate the transformation of immune "cold" tumors to immune "hot" tumors by eliciting innate and adaptive immune responses through the release of (or exposure to) damage-related molecular patterns. Accumulating evidence suggests that various traditional therapies can induce ICD, including chemotherapy, targeted therapy, radiotherapy, and photodynamic therapy. In this review, we summarize the biological mechanisms and hallmarks of ICD and introduce some newly discovered and technologically innovative inducers that activate the immune system at the molecular level. Furthermore, we also discuss the clinical applications of combing ICD inducers with cancer immunotherapy. This review will provide valuable insights into the future development of ICD-related combination therapeutics and potential management for "cold" tumors.
Collapse
Affiliation(s)
| | | | | | - Zhenyong Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
8
|
Alotaibi F, Alshammari K, Alotaibi BA, Alsaab H. Destabilizing the genome as a therapeutic strategy to enhance response to immune checkpoint blockade: a systematic review of clinical trials evidence from solid and hematological tumors. Front Pharmacol 2024; 14:1280591. [PMID: 38264532 PMCID: PMC10803447 DOI: 10.3389/fphar.2023.1280591] [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: 08/20/2023] [Accepted: 12/11/2023] [Indexed: 01/25/2024] Open
Abstract
Background: Genomic instability is increased alterations in the genome during cell division and is common among most cancer cells. Genome instability enhances the risk of initial carcinogenic transformation, generating new clones of tumor cells, and increases tumor heterogeneity. Although genome instability contributes to malignancy, it is also an "Achilles' heel" that constitutes a therapeutically-exploitable weakness-when sufficiently advanced, it can intrinsically reduce tumor cell survival by creating DNA damage and mutation events that overwhelm the capacity of cancer cells to repair those lesions. Furthermore, it can contribute to extrinsic survival-reducing events by generating mutations that encode new immunogenic antigens capable of being recognized by the immune system, particularly when anti-tumor immunity is boosted by immunotherapy drugs. Here, we describe how genome-destabilization can induce immune activation in cancer patients and systematically review the induction of genome instability exploited clinically, in combination with immune checkpoint blockade. Methods: We performed a systematic review of clinical trials that exploited the combination approach to successfully treat cancers patients. We systematically searched PubMed, Cochrane Central Register of Controlled Trials, Clinicaltrials.gov, and publication from the reference list of related articles. The most relevant inclusion criteria were peer-reviewed clinical trials published in English. Results: We identified 1,490 studies, among those 164 were clinical trials. A total of 37 clinical trials satisfied the inclusion criteria and were included in the study. The main outcome measurements were overall survival and progression-free survival. The majority of the clinical trials (30 out of 37) showed a significant improvement in patient outcome. Conclusion: The majority of the included clinical trials reported the efficacy of the concept of targeting DNA repair pathway, in combination with immune checkpoint inhibitors, to create a "ring of synergy" to treat cancer with rational combinations.
Collapse
Affiliation(s)
- Faizah Alotaibi
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Alahsa, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Kanaan Alshammari
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- Oncology Department, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Badi A. Alotaibi
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Hashem Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| |
Collapse
|
9
|
Li M, Che N, Liu X, Xuan Y, Jin Y. Dauricine regulates prostate cancer progression by inhibiting PI3K/AKT-dependent M2 polarization of macrophages. Biochem Pharmacol 2023; 217:115838. [PMID: 37778445 DOI: 10.1016/j.bcp.2023.115838] [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: 07/26/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
M2 type tumor-associated macrophages, an essential component of the tumor microenvironment (TME), have been proved to contribute to tumor metastasis. Dauricine (Dau) has recently received widespread attention due to its multiple targets and low price. However, the effect of Dau on macrophage polarization of TME remains unclear. In this study, we investigated the effect of Dau on prostate cancer (PCa) metastasis and specifically its correlation to macrophage polarization. Our results showed that Dau efficiently suppressed M2 polarization of macrophages induced by interleukin (IL) -4 and IL-13. Mechanistically, Dau inhibited the activity of PI3K/AKT signaling pathway, which subsequently suppressed macrophage differentiation to M2 type. Importantly, our study indicated that Dau decreased the release of chitinase 3-like protein 1 (CHI3L1) from M2 macrophages, which ultimately inhibited the M2 macrophage-mediated progression of PCa cells in vitro and in vivo. Taken together, our data demonstrated that Dau suppressed M2 polarization of macrophages via downregulation of the PI3K/AKT signaling pathway, in turn, preventing proliferation, epithelial-mesenchymal transition, migration, and invasion of PCa cells. Thus, this study reveals a previously unrecognized function of Dau in inhibition of PCa progression via intervention in M2 polarization of macrophages.
Collapse
Affiliation(s)
- Mengxuan Li
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China
| | - Nan Che
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China
| | - Xingzhe Liu
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China; Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Yanhua Xuan
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China; Department of Pathology, Yanbian University College of Medicine, Yanji, China.
| | - Yu Jin
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China; Department of Human Anatomy and Histoembryology, Yanbian University College of Medicine, Yanji, China.
| |
Collapse
|
10
|
Li L, Zou Y, Wang L, Yang L, Li Y, Liao A, Chen Z, Yu Z, Guo J, Han S. Nanodelivery of scutellarin induces immunogenic cell death for treating hepatocellular carcinoma. Int J Pharm 2023:123114. [PMID: 37301243 DOI: 10.1016/j.ijpharm.2023.123114] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/17/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023]
Abstract
Hepatocellular carcinoma (HCC) causes the immunosuppressive tumor microenvironment (TME) resistant to current immunotherapy. The immunogenic apoptosis (currently termed immunogenic cell death, ICD) of cancer cells may induce the adaptive immunity against tumors, thereby providing great potential for treating HCC. In this study, we have confirmed the potential of scutellarin (SCU, a flavonoid found in Erigeron breviscapus) for triggering ICD in HCC cells. To facilitate in vivo application of SCU for HCC immunotherapy, an aminoethyl anisamide-targeted polyethylene glycol-modified poly(lactide-co-glycolide) (PLGA-PEG-AEAA) was produced to facilitate SCU delivery in this study. The resultant nanoformulation (PLGA-PEG-AEAA.SCU) remarkably promoted blood circulation and tumor delivery in the orthotopic HCC mouse model. Consequently, PLGA-PEG-AEAA.SCU reversed the immune suppressive TME and achieved the immunotherapeutic efficacy, resulting in significantly longer survival of mice, without inducing toxicity. These findings uncover the ICD potential of SCU and provide a promising strategy for HCC immunotherapy.
Collapse
Affiliation(s)
- Linlin Li
- Center for Prenatal Diagnosis and Reproductive Medicine, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Lingzhi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Leilei Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yutong Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Anqi Liao
- Center for Prenatal Diagnosis and Reproductive Medicine, The First Hospital of Jilin University, Changchun 130021, China; School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zheng Chen
- Key Laboratory of High-Performance Plastics, Ministry of Education, National & Local Joint Engineering Laboratory for Synthesis Technology of High-Performance Polymer, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Shulan Han
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
11
|
Kalami A, Shahgolzari M, Khosroushahi AY, Fiering S. Combining in situ vaccination and immunogenic apoptosis to treat cancer. Immunotherapy 2023; 15:367-381. [PMID: 36852419 DOI: 10.2217/imt-2022-0137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Immunization approaches are designed to stimulate the immune system and eliminate the tumor. Studies indicate that cancer immunization combined with certain chemotherapeutics and immunostimulatory agents can improve outcomes. Chemotherapeutics-based immunogenic cell death makes the tumor more recognizable by the immune system. In situ vaccination (ISV) utilizes established tumors as antigen sources and directly applies an immune adjuvant to the tumor to reverse a cold tumor microenvironment to a hot one. Immunogenic cell death and ISV highlight for the immune system the tumor antigens that are recognizable by immune cells and support a T-cell attack of the tumor cells. This review presents the concept of immunogenic apoptosis and ISV as a powerful platform for cancer immunization.
Collapse
Affiliation(s)
- Arman Kalami
- Biotechnology Research Center, Student Research Committee, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Shahgolzari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Steven Fiering
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth & Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
12
|
Huang FY, Dai SZ, Xu WT, Xiong W, Sun Y, Huang YH, Wang JY, Lin YY, Chen H, Tan GH, Zheng WP. 3'-epi-12β-hydroxyfroside-mediated autophagy degradation of RIPK1/RIPK3 necrosomes leads to anergy of immunogenic cell death in triple-negative breast cancer cells. Pharmacol Res 2023; 187:106613. [PMID: 36535569 DOI: 10.1016/j.phrs.2022.106613] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Increasing studies have suggested that some cardiac glycosides, such as conventional digoxin (DIG) and digitoxin, can induce immunogenic cell death (ICD) in various tumors. We previously found that 3'-epi-12β-hydroxyfroside (HyFS), a novel cardenolide compound isolated by our group, could induce cytoprotective autophagy through inactivation of the Akt/mTOR pathway. However, whether HyFS can induce ICD remains unknown. In this study, we extend our work to further investigate whether HyFS could induce both autophagy and ICD, and we investigated the relationship between autophagy and ICD in three TNBC cell lines. Unexpectedly, compared to DIG, we found that HyFS could induce complete autophagy flux but not ICD in three human triple-negative breast cancer (TNBC) cell lines and one murine TNBC model. Inhibition of HyFS-induced autophagy resulted in the production of ICD in TNBC MDA-MB-231, MDA-MB-436, and HCC38 cells. A further mechanism study showed that formation of RIPK1/RIPK3 necrosomes was necessary for ICD induction in DIG-treated TNBC cells, while HyFS treatment led to receptor-interacting serine-threonine kinase (RIPK)1/3 necrosome degradation via an autophagy process. Additionally, inhibition of HyFS-induced autophagy by the autophagy inhibitor chloroquine resulted in the reoccurrence of ICD and reversion of the tumor microenvironment, leading to more significant antitumor effects in immunocompetent mice than in immunodeficient mice. These findings indicate that HyFS-mediated autophagic degradation of RIPK1/RIPK3 necrosomes leads to inactivation of ICD in TNBC cells. Moreover, combined treatment with HyFS and an autophagy inhibitor may enhance the antitumor activities, suggesting an alternative therapeutic for TNBC treatment.
Collapse
Affiliation(s)
- Feng-Ying Huang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Shu-Zhen Dai
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Wen-Tian Xu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Wei Xiong
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Yan Sun
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Yong-Hao Huang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Jin-Yan Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Ying-Ying Lin
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Hengyu Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China.
| | - Guang-Hong Tan
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China.
| | - Wu-Ping Zheng
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 570311, China.
| |
Collapse
|
13
|
Zhang R, Neighbors J, Schell T, Hohl R. Schweinfurthin induces ICD without ER stress and caspase activation. Oncoimmunology 2022; 11:2104551. [PMID: 35936984 PMCID: PMC9354771 DOI: 10.1080/2162402x.2022.2104551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Our previous study showed that one of the schweinfurthin compounds, 5’-methoxyschweinfurthin G (MeSG), not only enhances the anti-tumor effect of anti-PD1 antibody in the B16F10 murine melanoma model, but also provokes durable, protective anti-tumor immunity. Here we further investigated the mechanisms by which MeSG treatment induces immunogenic cell death (ICD). MeSG induced significant cell surface calreticulin (CRT) exposure in a time and concentration dependent manner as well as increased phagocytosis of tumor cells by dendritic cells in vitro. Interestingly, this CRT exposure differs from the canonical pathway in several aspects. MeSG does not cause ER stress and does not require PERK to induce CRT exposure. Caspase inhibitors partially rescue cells from MeSG-induced apoptosis, but fail to reduce CRT exposure. MeSG does not cause ERp57 exposure and the absence of ERp57 expression does not reduce CRT exposure. Finally, an intact ER to Golgi transport system is required for this phenomenon. These results lend support to the development of the schweinfurthin family as drugs to enhance clinical response to immunotherapy and highlight the need for additional research on the mechanisms of ICD induction.
Collapse
Affiliation(s)
| | - J.D. Neighbors
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Hershey, PA, USA
| | - T.D. Schell
- Penn State Cancer Institute, Hershey, PA, USA
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, USA
| | - R.J. Hohl
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Hershey, PA, USA
| |
Collapse
|
14
|
Florêncio KGD, Edson EA, Fernandes KSDS, Luiz JPM, Pinto FDCL, Pessoa ODL, Cunha FDQ, Machado-Neto JA, Wilke DV. Chromomycin A 5 induces bona fide immunogenic cell death in melanoma. Front Immunol 2022; 13:941757. [PMID: 36439184 PMCID: PMC9682167 DOI: 10.3389/fimmu.2022.941757] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/12/2022] [Indexed: 08/27/2023] Open
Abstract
PURPOSE Some first-line cytotoxic chemotherapics, e.g. doxorubicin, paclitaxel and oxaliplatin, induce activation of the immune system through immunogenic cell death (ICD). Tumor cells undergoing ICD function as a vaccine, releasing damage-associated molecular patterns (DAMPs), which act as adjuvants, and neoantigens of the tumor are recognized as antigens. ICD induction is rare, however it yields better and long-lasting antitumor responses to chemotherapy. Advanced metastatic melanoma (AMM) is incurable for more than half of patients. The discovery of ICD inducers against AMM is an interesting drug discovery strategy with high translational potential. Here we evaluated ICD induction of four highly cytotoxic chromomycins A (CA5-8). METHODS ICD features and DAMPs were evaluated using several in vitro techniques with metastatic melanoma cell line (B16-F10) exposed to chromomcins A5-8 such as flow cytometry, western blot, RT-PCR and luminescence. Additionally in vivo vaccination assays with CA5-treated cells in a syngeneic murine model (C57Bl/6) were performed to confirm ICD evaluating the immune cells activation and their antitumor activity. RESULTS B16-F10 treated with CA5-8 and doxorubicin exhibited ICD features such as autophagy and apoptosis, externalization of calreticulin, and releasing of HMGB1. However, CA5-treated cells had the best profile, also inducing ATP release, ERp57 externalization, phosphorylation of eIF2α and altering expression of transcription of genes related to autophagy, endoplasmic reticulum stress, and apoptosis. Bona fide ICD induction by CA5 was confirmed by vaccination of C57BL/6 mice with CA5-treated cells which activated antigen-presenting cells and T lymphocytes and stimulated antitumor activity. CONCLUSION CA5 induces bona fide immunogenic cell death on melanoma.
Collapse
Affiliation(s)
- Katharine Gurgel Dias Florêncio
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - Evelline Araújo Edson
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - Keilla Santana da Silva Fernandes
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - João Paulo Mesquita Luiz
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | - Fernando de Queiroz Cunha
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | - Diego Veras Wilke
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| |
Collapse
|
15
|
Rodrigues MC, Morais JAV, Ganassin R, Oliveira GRT, Costa FC, Morais AAC, Silveira AP, Silva VCM, Longo JPF, Muehlmann LA. An Overview on Immunogenic Cell Death in Cancer Biology and Therapy. Pharmaceutics 2022; 14:pharmaceutics14081564. [PMID: 36015189 PMCID: PMC9413301 DOI: 10.3390/pharmaceutics14081564] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
Immunogenic cell death (ICD) is a modality of regulated cell death that is sufficient to promote an adaptive immune response against antigens of the dying cell in an immunocompetent host. An important characteristic of ICD is the release and exposure of damage-associated molecular patterns, which are potent endogenous immune adjuvants. As the induction of ICD can be achieved with conventional cytotoxic agents, it represents a potential approach for the immunotherapy of cancer. Here, different aspects of ICD in cancer biology and treatment are reviewed.
Collapse
Affiliation(s)
- Mosar Corrêa Rodrigues
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - José Athayde Vasconcelos Morais
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Rayane Ganassin
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Giulia Rosa Tavares Oliveira
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Fabiana Chagas Costa
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Amanda Alencar Cabral Morais
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Ariane Pandolfo Silveira
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Victor Carlos Mello Silva
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - João Paulo Figueiró Longo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Luis Alexandre Muehlmann
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
- Correspondence:
| |
Collapse
|
16
|
Karp DD, Camidge DR, Infante JR, Ames TD, Price MR, Jimeno J, Bryce AH. Phase I study of PT-112, a novel pyrophosphate-platinum immunogenic cell death inducer, in advanced solid tumours. EClinicalMedicine 2022; 49:101430. [PMID: 35747193 PMCID: PMC9156977 DOI: 10.1016/j.eclinm.2022.101430] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND PT-112, the first pyrophosphate-platinum conjugate, causes immunogenic cell death in experimental models, leading to recruitment of tumour-infiltrating lymphocytes. PT-112 also associates with bone (osteotropism), likely driven by its pyrophosphate moiety. This is the first-in-human study of PT-112 monotherapy, exploring its safety and efficacy in a patient population where standard of care therapies were exhausted and novel treatment options are needed. METHODS Patients with progressing, advanced solid tumours received PT-112 intravenously (1 h) on days 1, 8, 15 of a 28-day cycle in an open-label, multi-centre 3 + 3 dose-escalation trial, conducted at four US research sites. The primary objective was to assess safety and pharmacokinetics, and to identify a recommended phase 2 dose (RP2D). Eligibility criteria included: age ≥18 years, Eastern Collaborative Oncology Group (ECOG) Performance Status of 0-1, and disease evaluable by Response Evaluation Criteria in Solid Tumours (RECIST) v1·1 or by informative tumour markers. Patients receiving ≥1 dose of PT-112 were included in the safety and pharmacokinetic analyses, with the exploratory efficacy analysis including patients receiving ≥1 dose at 125 mg/m2. This study is registered at ClinicalTrials.gov, number NCT02266745, with the dose-escalation portion of the study closed. FINDINGS Between July 7th, 2014 and September 18th, 2018, 66 heavily pre-treated patients (median 4 prior lines, IQR 2-6) were enrolled and treated across 11 doses (12-420 mg/m2). Treatment-related adverse events included fatigue (23 patients, 35%), nausea (16 patients, 24%), and peripheral neuropathy (14 patients, 21%). Grade 3 events were experienced by 18 patients (27%), with no grade 4-5 events observed. The recommended phase 2 dose was determined to be 360 mg/m2. Nine (17%) of the 54 efficacy evaluable patients achieved progression-free survival ≥6 months. Durable partial responses were induced in non-small cell lung cancer (NSCLC), small cell lung cancer (SCLC), and thymoma. Radiographic and serum marker reductions were observed among ten patients with metastatic castration resistant prostate cancer, four of whom survived two years or longer. INTERPRETATION PT-112 is safe and well-tolerated in a heavily pre-treated population. Prolonged responses were noted against thymoma and lung cancer, along with radiographic and serum marker improvement in prostate cancer. Given the heterogeneous patient population, subsequent studies will be needed to characterize the risk/benefit ratio in more homogenous settings. Further development of PT-112 is ongoing, as single-agent and in combination with immune checkpoint inhibition. FUNDING Funding was provided by Promontory Therapeutics Inc.
Collapse
Affiliation(s)
- Daniel D. Karp
- Investigational Cancer Therapeutics Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd #U421, Houston, TX 77030, USA
- Corresponding author.
| | - D. Ross Camidge
- Medical Oncology, University of Colorado Cancer Center Anschutz Cancer Pavilion, Aurora, CO, USA
| | - Jeffery R. Infante
- Department of Drug Development, Sarah Cannon Research Institute, Nashville, TN, USA
| | | | | | - José Jimeno
- Promontory Therapeutics Inc., New York, NY, USA
| | - Alan H. Bryce
- Department of Internal Medicine, Division of Hematology/Oncology, Mayo Clinic Cancer Center, Phoenix, AZ, USA
| |
Collapse
|
17
|
Matsuo Y, Tsuchihashi H, Takatori K, Fukaya H, Kuroda M, Mimaki Y. Cytotoxic triterpene and steroidal glycosides from the seeds of Digitalis purpurea and the synergistic cytotoxicity of steroidal glycosides and etoposide in SBC-3 cells. Bioorg Chem 2022; 122:105697. [DOI: 10.1016/j.bioorg.2022.105697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/03/2022] [Accepted: 02/18/2022] [Indexed: 11/02/2022]
|
18
|
Jiang W, Dong W, Li M, Guo Z, Wang Q, Liu Y, Bi Y, Zhou H, Wang Y. Nitric Oxide Induces Immunogenic Cell Death and Potentiates Cancer Immunotherapy. ACS NANO 2022; 16:3881-3894. [PMID: 35238549 DOI: 10.1021/acsnano.1c09048] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor cells undergoing immunogenic cell death (ICD) release immunogenic damage-associated molecular patterns (DAMPs) to trigger a long-term protective antitumor response. ICD can be induced by certain pathogens, chemotherapeutics, and physical modalities. In this work, we demonstrate that a gaseous molecule, specifically nitric oxide (NO), can induce a potent ICD effect. NO exerts cytotoxic effects that are accompanied by the emission of DAMPs based on the endoplasmic reticulum stress and mitochondrial dysfunction pathways. Released DAMPs elicit immunological protection against a subsequent rechallenge of syngeneic tumor cells in immunocompetent mice. We prepare polynitrosated polyesters with high NO storage capacity through a facile polycondensation reaction followed by a postsynthetic modification. The polynitrosated polyesters-based NO nanogenerator (NanoNO) that enables efficient NO delivery and controlled NO release in tumors induces a sufficient ICD effect. In different immune-intact models of tumors, the NanoNO exhibits significant tumor growth suppression and increases the local dose of immunogenic signals and T cell infiltrations, ultimately prolonging survival. In addition, the NanoNO synergizes with the PD-1 blockade to prevent metastasis. We conclude not only that NO is a potent ICD inducer for cancer immunotherapy but also that it expands the range of ICD inducers into the field of gaseous molecules.
Collapse
Affiliation(s)
- Wei Jiang
- Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Wang Dong
- Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Min Li
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zixuan Guo
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Qin Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yi Liu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yihui Bi
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Han Zhou
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yucai Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
19
|
Dong S, Guo X, Han F, He Z, Wang Y. Emerging role of natural products in cancer immunotherapy. Acta Pharm Sin B 2022; 12:1163-1185. [PMID: 35530162 PMCID: PMC9069318 DOI: 10.1016/j.apsb.2021.08.020] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/05/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy has become a new generation of anti-tumor treatment, but its indications still focus on several types of tumors that are sensitive to the immune system. Therefore, effective strategies that can expand its indications and enhance its efficiency become the key element for the further development of cancer immunotherapy. Natural products are reported to have this effect on cancer immunotherapy, including cancer vaccines, immune-check points inhibitors, and adoptive immune-cells therapy. And the mechanism of that is mainly attributed to the remodeling of the tumor-immunosuppressive microenvironment, which is the key factor that assists tumor to avoid the recognition and attack from immune system and cancer immunotherapy. Therefore, this review summarizes and concludes the natural products that reportedly improve cancer immunotherapy and investigates the mechanism. And we found that saponins, polysaccharides, and flavonoids are mainly three categories of natural products, which reflected significant effects combined with cancer immunotherapy through reversing the tumor-immunosuppressive microenvironment. Besides, this review also collected the studies about nano-technology used to improve the disadvantages of natural products. All of these studies showed the great potential of natural products in cancer immunotherapy.
Collapse
Key Words
- AKT, alpha-serine/threonine-specific protein kinase
- Adoptive immune-cells transfer immunotherapy
- B2M, beta-2-microglobulin
- BMDCs, bone marrow dendritic cells
- BPS, basil polysaccharide
- BTLA, B- and T-lymphocyte attenuator
- CAFs, cancer-associated fibroblasts
- CCL22, C–C motif chemokine 22
- CIKs, cytokine-induced killer cells
- COX-2, cyclooxygenase-2
- CRC, colorectal cancer
- CTL, cytotoxic T cell
- CTLA-4, cytotoxic T lymphocyte antigen-4
- Cancer immunotherapy
- Cancer vaccines
- DAMPs, damage-associated molecular patterns
- DCs, dendritic cells
- FDA, US Food and Drug Administration
- HCC, hepatocellular carcinoma
- HER-2, human epidermal growth factor receptor-2
- HIF-1α, hypoxia-inducible factor-1α
- HMGB1, high-mobility group box 1
- HSPs, heat shock proteins
- ICD, Immunogenic cell death
- ICTs, immunological checkpoints
- IFN-γ, interferon γ
- IL-10, interleukin-10
- Immuno-check points
- Immunosuppressive microenvironment
- LLC, Lewis lung cancer
- MDSCs, myeloid-derived suppressor cells
- MHC, major histocompatibility complex class
- MITF, melanogenesis associated transcription factor
- MMP-9, matrix metalloprotein-9
- Mcl-1, myeloid leukemia cell differentiation protein 1
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NKTs, natural killer T cells
- NSCLC, non-small cell lung cancer
- Natural products
- OVA, ovalbumin
- PD-1, programmed death-1
- PD-L1, programmed death receptor ligand 1
- PGE-2, prostaglandin E2
- PI3K, phosphoinositide 3-kinase
- ROS, reactive oxygen species
- STAT3, signal transducer and activator of transcription 3
- TAMs, tumor-associated macrophages
- TAP, transporters related with antigen processing
- TGF-β, transforming growth factor-β
- TILs, tumor infiltration lymphocytes
- TLR, Toll-like receptor
- TNF-α, tumor necrosis factor α
- TSA, tumor specific antigens
- Teffs, effective T cells
- Th1, T helper type 1
- Tregs, regulatory T cells
- VEGF, vascular endothelial growth factor
- bFGF, basic fibroblast growth factor
- mTOR, mechanistic target of rapamycin
Collapse
Affiliation(s)
- Songtao Dong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiangnan Guo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fei Han
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongjun Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
20
|
Wainwright CL, Teixeira MM, Adelson DL, Buenz EJ, David B, Glaser KB, Harata-Lee Y, Howes MJR, Izzo AA, Maffia P, Mayer AM, Mazars C, Newman DJ, Nic Lughadha E, Pimenta AM, Parra JA, Qu Z, Shen H, Spedding M, Wolfender JL. Future Directions for the Discovery of Natural Product-Derived Immunomodulating Drugs. Pharmacol Res 2022; 177:106076. [PMID: 35074524 DOI: 10.1016/j.phrs.2022.106076] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/07/2022] [Indexed: 02/06/2023]
Abstract
Drug discovery from natural sources is going through a renaissance, having spent many decades in the shadow of synthetic molecule drug discovery, despite the fact that natural product-derived compounds occupy a much greater chemical space than those created through synthetic chemistry methods. With this new era comes new possibilities, not least the novel targets that have emerged in recent times and the development of state-of-the-art technologies that can be applied to drug discovery from natural sources. Although progress has been made with some immunomodulating drugs, there remains a pressing need for new agents that can be used to treat the wide variety of conditions that arise from disruption, or over-activation, of the immune system; natural products may therefore be key in filling this gap. Recognising that, at present, there is no authoritative article that details the current state-of-the-art of the immunomodulatory activity of natural products, this in-depth review has arisen from a joint effort between the International Union of Basic and Clinical Pharmacology (IUPHAR) Natural Products and Immunopharmacology, with contributions from a Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporation number of world-leading researchers in the field of natural product drug discovery, to provide a "position statement" on what natural products has to offer in the search for new immunomodulatory argents. To this end, we provide a historical look at previous discoveries of naturally occurring immunomodulators, present a picture of the current status of the field and provide insight into the future opportunities and challenges for the discovery of new drugs to treat immune-related diseases.
Collapse
Affiliation(s)
- Cherry L Wainwright
- Centre for Natural Products in Health, Robert Gordon University, Aberdeen, UK.
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil.
| | - David L Adelson
- Molecular & Biomedical Science, University of Adelaide, Australia.
| | - Eric J Buenz
- Nelson Marlborough Institute of Technology, New Zealand.
| | - Bruno David
- Green Mission Pierre Fabre, Pierre Fabre Laboratories, Toulouse, France.
| | - Keith B Glaser
- AbbVie Inc., Integrated Discovery Operations, North Chicago, USA.
| | - Yuka Harata-Lee
- Molecular & Biomedical Science, University of Adelaide, Australia
| | - Melanie-Jayne R Howes
- Royal Botanic Gardens Kew, Richmond, Surrey, UK; Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, UK.
| | - Angelo A Izzo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Italy.
| | - Pasquale Maffia
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Italy; Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | - Alejandro Ms Mayer
- Department of Pharmacology, College of Graduate Studies, Midwestern University, IL, USA.
| | - Claire Mazars
- Green Mission Pierre Fabre, Pierre Fabre Laboratories, Toulouse, France.
| | | | | | - Adriano Mc Pimenta
- Laboratory of Animal Venoms and Toxins, Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - John Aa Parra
- Laboratory of Animal Venoms and Toxins, Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Zhipeng Qu
- Molecular & Biomedical Science, University of Adelaide, Australia
| | - Hanyuan Shen
- Molecular & Biomedical Science, University of Adelaide, Australia
| | | | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland.
| |
Collapse
|
21
|
Guo S, Burcus NI, Scott M, Jing Y, Semenov I. The role of reactive oxygen species in the immunity induced by nano-pulse stimulation. Sci Rep 2021; 11:23745. [PMID: 34887493 PMCID: PMC8660900 DOI: 10.1038/s41598-021-03342-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 11/25/2021] [Indexed: 11/29/2022] Open
Abstract
Reactive oxygen species (ROS) are byproducts of tumor cells treated with Nano-Pulse Stimulation (NPS). Recently, ROS have been suggested as a contributing factor in immunogenic cell death and T cell-mediated immunity. This research further investigated the role of NPS induced ROS in antitumor immunity. ROS production in 4T1-luc breast cancer cells was characterized using three detection reagents, namely, Amplex Red, MitoSox Red, and Dihydroethidium. The efficiency of ROS quenching was evaluated in the presence or absence of ROS scavengers and/or antioxidants. The immunogenicity of NPS treated tumor cells was assessed by ex vivo dendritic cell activation, in vivo vaccination assay and in situ vaccination with NPS tumor ablation. We found that NPS treatment enhanced the immunogenicity of 4T1-luc mouse mammary tumor, resulted in a potent in situ vaccination protection and induced long-term T cell immunity. ROS production derived from NPS treated breast cancer cells was an electric pulse dose-dependent phenomenon. Noticeably, the dynamic pattern of hydrogen peroxide production was different from that of superoxide production. Interestingly, regardless of NPS treatment, different ROS scavengers could either block or promote ROS production and stimulate or inhibit tumor cell growth. The activation of dendritic cells was not influenced by blocking ROS generation. The results from in vivo vaccination with NPS treated cancer cells suggests that ROS generation was not a prerequisite for immune protection.
Collapse
Affiliation(s)
- Siqi Guo
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA.
| | - Niculina I. Burcus
- grid.261368.80000 0001 2164 3177Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508 USA
| | - Megan Scott
- grid.261368.80000 0001 2164 3177Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508 USA
| | - Yu Jing
- grid.261368.80000 0001 2164 3177Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508 USA
| | - Iurii Semenov
- grid.261368.80000 0001 2164 3177Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508 USA
| |
Collapse
|
22
|
Role of Damage-Associated Molecular Pattern/Cell Death Pathways in Vaccine-Induced Immunity. Viruses 2021; 13:v13122340. [PMID: 34960608 PMCID: PMC8708515 DOI: 10.3390/v13122340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
Immune responses induced by natural infection and vaccination are known to be initiated by the recognition of microbial patterns by cognate receptors, since microbes and most vaccine components contain pathogen-associated molecular patterns. Recent discoveries on the roles of damage-associated molecular patterns (DAMPs) and cell death in immunogenicity have improved our understanding of the mechanism underlying vaccine-induced immunity. DAMPs are usually immunologically inert, but can transform into alarming signals to activate the resting immune system in response to pathogenic infection, cellular stress and death, or tissue damage. The activation of DAMPs and cell death pathways can trigger local inflammation, occasionally mediating adaptive immunity, including antibody- and cell-mediated immune responses. Emerging evidence indicates that the components of vaccines and adjuvants induce immunogenicity via the stimulation of DAMP/cell death pathways. Furthermore, strategies for targeting this pathway to enhance immunogenicity are being investigated actively. In this review, we describe various DAMPs and focus on the roles of DAMP/cell death pathways in the context of vaccines for infectious diseases and cancer.
Collapse
|
23
|
Antitumor Immune Response Triggered by Metal-Based Photosensitizers for Photodynamic Therapy: Where Are We? Pharmaceutics 2021; 13:pharmaceutics13111788. [PMID: 34834202 PMCID: PMC8620627 DOI: 10.3390/pharmaceutics13111788] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 11/17/2022] Open
Abstract
Metal complexes based on transition metals have rich photochemical and photophysical properties that are derived from a variety of excited state electronic configurations triggered by visible and near-infrared light. These properties can be exploited to produce powerful energy and electron transfer processes that can lead to oxygen-(in)dependent photobiological activity. These principles are the basis of photodynamic therapy (PDT), which is a clinically approved treatment that offers a promising, effective, and noninvasive complementary treatment or even an alternative to treat several types of cancers. PDT is based on a reaction involving a photosensitizer (PS), light, and oxygen, which ultimately generates cytotoxic reactive oxygen species (ROS). However, skin photosensitivity, due to the accumulation of PSs in skin cells, has hampered, among other elements, its clinical development and application. Therefore, these is an increasing interest in the use of (metal-based) PSs that are more specific to tumor cells. This may increase efficacy and corollary decrease side-effects. To this end, metal-containing nanoparticles with photosensitizing properties have recently been developed. In addition, several studies have reported that the use of immunogenic/immunomodulatory metal-based nanoparticles increases the antitumor efficacy of immune-checkpoint inhibitor-based immunotherapy mediated by anti-PD-(L)1 or CTLA-4 antibodies. In this review, we discuss the main metal complexes used as PDT PSs. Lastly, we review the preclinical studies associated with metal-based PDT PSs and immunotherapies. This therapeutic association could stimulate PDT.
Collapse
|
24
|
Fan F, Shen P, Ma Y, Ma W, Wu H, Liu H, An Q. Bullatacin triggers immunogenic cell death of colon cancer cells by activating endoplasmic reticulum chaperones. JOURNAL OF INFLAMMATION-LONDON 2021; 18:23. [PMID: 34112202 PMCID: PMC8194247 DOI: 10.1186/s12950-021-00289-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 05/27/2021] [Indexed: 11/27/2022]
Abstract
Background It is well accepted that the immune system efficiently contributes to positive outcomes of chemotherapeutic cancer treatment by activating immunogenic cell death (ICD). However, only a limited number of ICD-inducing compounds are well characterized at present; therefore, identification of novel ICD inducers is urgently needed for cancer drug discovery, and the need is becoming increasingly urgent. Methods Herein, we assessed the antitumour activity of bullatacin by MTS assay and apoptosis assay. ICD biomarkers, such as calreticulin (CRT), high-mobility group protein B1 (HMGB-1), heat shock protein (HSP)70, HSP90 and ATP, were assessed by Western blotting, ELISA and flow cytometry. Western blot and qPCR assays were performed to explore the underlying mechanisms of bullatacin-induced ICD. Flow cytometry was used to detect macrophage phagocytosis. Results First, bullatacin induced apoptosis in both SW480 cells and HT-29 cells in a time-dependent manner at 10 nM, as assessed by flow cytometry. Moreover, Western blot and flow cytometry assays showed that CRT and HSP90 (biomarkers of early ICD) significantly accumulated on the cell membrane surface after approximately 6 h of treatment with bullatacin. In addition, ELISAs and Western blot assays showed that the second set of hallmarks required for ICD (HMGB1, HSP70 and HSP90) were released in the conditioned media of both SW480 and HT-29 cells after 36 h of treatment. Furthermore, qPCR and Western blot assays indicated that bullatacin triggered ICD via activation of the endoplasmic reticulum stress (ERS) signalling pathway. Finally, bullatacin promoted macrophage phagocytosis. Conclusion This study documents that bullatacin, a novel ICD inducer, triggers immunogenic tumour cell death by activating ERS even at a relatively low concentration in vitro.
Collapse
Affiliation(s)
- Fangtian Fan
- Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, School of Pharmacy, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, 233003, Anhui, China.
| | - Peiliang Shen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yue Ma
- Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, School of Pharmacy, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, 233003, Anhui, China
| | - Wangbo Ma
- Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, School of Pharmacy, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, 233003, Anhui, China
| | - Hongyan Wu
- Institute of Biomedical Technology, Jiangsu Vocational College of Medicine, No.283 Jiefang South Road, Yancheng, 224005, China.
| | - Hao Liu
- Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, School of Pharmacy, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, 233003, Anhui, China
| | - Qing An
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| |
Collapse
|
25
|
Atanasov AG, Zotchev SB, Dirsch VM, Supuran CT. Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discov 2021; 20:200-216. [PMID: 33510482 PMCID: PMC7841765 DOI: 10.1038/s41573-020-00114-z] [Citation(s) in RCA: 1938] [Impact Index Per Article: 646.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 02/07/2023]
Abstract
Natural products and their structural analogues have historically made a major contribution to pharmacotherapy, especially for cancer and infectious diseases. Nevertheless, natural products also present challenges for drug discovery, such as technical barriers to screening, isolation, characterization and optimization, which contributed to a decline in their pursuit by the pharmaceutical industry from the 1990s onwards. In recent years, several technological and scientific developments - including improved analytical tools, genome mining and engineering strategies, and microbial culturing advances - are addressing such challenges and opening up new opportunities. Consequently, interest in natural products as drug leads is being revitalized, particularly for tackling antimicrobial resistance. Here, we summarize recent technological developments that are enabling natural product-based drug discovery, highlight selected applications and discuss key opportunities.
Collapse
Affiliation(s)
- Atanas G Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland.
- Department of Pharmacognosy, University of Vienna, Vienna, Austria.
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria.
| | - Sergey B Zotchev
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Claudiu T Supuran
- Università degli Studi di Firenze, NEUROFARBA Dept, Sezione di Scienze Farmaceutiche, Florence, Italy.
| |
Collapse
|
26
|
Cerella C, Dicato M, Diederich M. BH3 Mimetics in AML Therapy: Death and Beyond? Trends Pharmacol Sci 2020; 41:793-814. [PMID: 33032835 DOI: 10.1016/j.tips.2020.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/01/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022]
Abstract
B cell lymphoma 2 (BCL2) homology domain 3 (BH3) mimetics are targeted therapeutic agents that allow response prediction and patient stratification. BH3 mimetics are prototypical activators of the mitochondrial death program in cancer. They emerged as important modulators of cellular mechanisms contributing to poor therapeutic responses, including cancer cell stemness, cancer-specific metabolic routes, paracrine signaling to the tumor microenvironment, and immune modulation. We present an overview of the antagonism between BH3 mimetics and antiapoptotic BCL2 proteins. We focus on acute myeloid leukemia (AML), a cancer with reduced therapeutic options that have recently been improved by BH3 mimetics.
Collapse
Affiliation(s)
- Claudia Cerella
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 151-742, South Korea.
| |
Collapse
|
27
|
Kopecka J, Godel M, Dei S, Giampietro R, Belisario DC, Akman M, Contino M, Teodori E, Riganti C. Insights into P-Glycoprotein Inhibitors: New Inducers of Immunogenic Cell Death. Cells 2020; 9:cells9041033. [PMID: 32331368 PMCID: PMC7226521 DOI: 10.3390/cells9041033] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/18/2020] [Accepted: 04/19/2020] [Indexed: 12/11/2022] Open
Abstract
Doxorubicin is a strong inducer of immunogenic cell death (ICD), but it is ineffective in P-glycoprotein (Pgp)-expressing cells. Indeed, Pgp effluxes doxorubicin and impairs the immunesensitizing functions of calreticulin (CRT), an "eat-me" signal mediating ICD. It is unknown if classical Pgp inhibitors, designed to reverse chemoresistance, may restore ICD. We addressed this question by using Pgp-expressing cancer cells, treated with Tariquidar, a clinically approved Pgp inhibitor, and R-3 compound, a N,N-bis(alkanol)amine aryl ester derivative with the same potency of Tariquidar as Pgp inhibitor. In Pgp-expressing/doxorubicin-resistant cells, Tariquidar and R-3 increased doxorubicin accumulation and toxicity, reduced Pgp activity, and increased CRT translocation and ATP and HMGB1 release. Unexpectedly, only R-3 promoted phagocytosis by dendritic cells and activation of antitumor CD8+T-lymphocytes. Although Tariquidar did not alter the amount of Pgp present on cell surface, R-3 promoted Pgp internalization and ubiquitination, disrupting its interaction with CRT. Pgp knock-out restores doxorubicin-induced ICD in MDA-MB-231/DX cells that recapitulated the phenotype of R-3-treated cells. Our work demonstrates that plasma membrane-associated Pgp prevents a complete ICD notwithstanding the release of ATP and HMGB1, and the exposure of CRT. Pharmacological compounds reducing Pgp activity and amount may act as promising chemo- and immunesensitizing agents.
Collapse
Affiliation(s)
- Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy; (J.K.); (M.G.); (D.C.B.); (M.A.)
| | - Martina Godel
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy; (J.K.); (M.G.); (D.C.B.); (M.A.)
| | - Silvia Dei
- Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical and Nutriceutical Sciences, University of Firenze, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (S.D.); (E.T.)
| | - Roberta Giampietro
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, via Orabona 4, 70125 Bari, Italy; (R.G.); (M.C.)
| | - Dimas Carolina Belisario
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy; (J.K.); (M.G.); (D.C.B.); (M.A.)
| | - Muhlis Akman
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy; (J.K.); (M.G.); (D.C.B.); (M.A.)
| | - Marialessandra Contino
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, via Orabona 4, 70125 Bari, Italy; (R.G.); (M.C.)
| | - Elisabetta Teodori
- Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical and Nutriceutical Sciences, University of Firenze, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy; (S.D.); (E.T.)
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy; (J.K.); (M.G.); (D.C.B.); (M.A.)
- Correspondence: ; Tel.: +39-011-670-5857
| |
Collapse
|
28
|
Chen W, Wang S, Wu Y, Shen X, Guo Z, Li Q, Xing D. Immunogenic cell death: A link between gut microbiota and anticancer effects. Microb Pathog 2020; 141:103983. [DOI: 10.1016/j.micpath.2020.103983] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
|
29
|
Florean C, Dicato M, Diederich M. Immune-modulating and anti-inflammatory marine compounds against cancer. Semin Cancer Biol 2020; 80:58-72. [PMID: 32070764 DOI: 10.1016/j.semcancer.2020.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
Abstract
The recent advances in cancer immunotherapy confirm the crucial role of the immune system in cancer progression and treatment. Chronic inflammation and reduced immune surveillance are both features of the tumor microenvironment. Strategies aimed at reverting pro-tumor inflammation and stimulating the antitumor immune components are being actively searched, and the anticancer effects of many candidate drugs have been linked to their ability to modulate the immune system. Marine organisms constitute a rich reservoir of new bioactive molecules; some of them have already been exploited for pharmaceutical use, whereas many others are undergoing clinical or preclinical investigations for the treatment of different diseases, including cancer. In this review, we will discuss the immune-modulatory properties of marine compounds for their potential use in cancer prevention and treatment and as possible tools in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Cristina Florean
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
30
|
Song S, Lee JY, Ermolenko L, Mazumder A, Ji S, Ryu H, Kim H, Kim DW, Lee JW, Dicato M, Christov C, Schnekenburger M, Cerella C, Gérard D, Orlikova-Boyer B, Al-Mourabit A, Diederich M. Tetrahydrobenzimidazole TMQ0153 triggers apoptosis, autophagy and necroptosis crosstalk in chronic myeloid leukemia. Cell Death Dis 2020; 11:109. [PMID: 32034134 PMCID: PMC7007439 DOI: 10.1038/s41419-020-2304-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
By comparing imatinib-sensitive and -resistant chronic myeloid leukemia (CML) cell models, we investigated the molecular mechanisms by which tetrahydrobenzimidazole derivative TMQ0153 triggered caspase-dependent apoptosis at low concentrations accompanied by loss of mitochondrial membrane potential (MMP) and increase of cytosolic free Ca2+ levels. Interestingly, at higher concentrations, TMQ0153 induced necroptotic cell death with accumulation of ROS, both preventable by N-acetyl-L-cysteine (NAC) pretreatment. At necroptosis-inducing concentrations, we observed increased ROS and decreased ATP and GSH levels, concomitant with protective autophagy induction. Inhibitors such as bafilomycin A1 (baf-A1) and siRNA against beclin 1 abrogated autophagy, sensitized CML cells against TMQ0153 and enhanced necroptotic cell death. Importantly, TMQ153-induced necrosis led to cell surface exposure of calreticulin (CRT) and ERp57 as well as the release of extracellular ATP and high mobility group box (HMGB1) demonstrating the capacity of this compound to release immunogenic cell death (ICD) markers. We validated the anti-cancer potential of TMQ0153 by in vivo inhibition of K562 microtumor formation in zebrafish. Taken together, our findings provide evidence that cellular stress and redox modulation by TMQ0153 concentration-dependently leads to different cell death modalities including controlled necrosis in CML cell models.
Collapse
Affiliation(s)
- Sungmi Song
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea
| | - Jin-Young Lee
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea
| | - Ludmila Ermolenko
- Département SNCM (Substances Naturelles et Chimie Médicinale), ICSN-CNRS, LabEx LERMIT, Centre de Recherche de Gif-sur-Yvette, Avenue de la Terrasse (Bat. 27), 91190, Gif-sur-Yvette, France
| | - Aloran Mazumder
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea
| | - Seungwon Ji
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea
| | - Heeju Ryu
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea
| | - HyeJin Kim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea
| | - Dong-Wook Kim
- Catholic University, Seoul St. Mary's Hospital, Banpo dong 505, Seocho Gu, Seoul, Korea
| | - Jung Weon Lee
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Christo Christov
- Service d'Histologie, Faculté de Médicine, Université de Lorraine, and INSERM U1256 NGERE, 54000, Nancy, France
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Claudia Cerella
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea.,Laboratoire de Biologie Moléculaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Déborah Gérard
- Laboratoire de Biologie Moléculaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Barbora Orlikova-Boyer
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea.,Laboratoire de Biologie Moléculaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Ali Al-Mourabit
- Département SNCM (Substances Naturelles et Chimie Médicinale), ICSN-CNRS, LabEx LERMIT, Centre de Recherche de Gif-sur-Yvette, Avenue de la Terrasse (Bat. 27), 91190, Gif-sur-Yvette, France.
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08626, Korea.
| |
Collapse
|
31
|
Diederich M. Natural products target the hallmarks of chronic diseases. Biochem Pharmacol 2020; 173:113828. [PMID: 32001237 DOI: 10.1016/j.bcp.2020.113828] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
Natural compounds are known to display therapeutic potential against a variety of chronic conditions, including cancer and inflammation. The efficacy of these natural substances can be associated with numerous molecular scaffolds present in extracts of living organisms, both terrestrial and marine. Recently, investigators have identified the ability of natural compounds to trigger immunogenic cell death and subsequent activation of the adaptive immune system. Such findings indicate that the full therapeutic potential of natural products has yet to be defined, and further investigations on such agents will continue to yield novel drug candidates.
Collapse
Affiliation(s)
- Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
32
|
Menezes JC, Diederich MF. Natural dimers of coumarin, chalcones, and resveratrol and the link between structure and pharmacology. Eur J Med Chem 2019; 182:111637. [DOI: 10.1016/j.ejmech.2019.111637] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023]
|
33
|
Diederich M. Natural compound inducers of immunogenic cell death. Arch Pharm Res 2019; 42:629-645. [PMID: 30955159 DOI: 10.1007/s12272-019-01150-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/29/2019] [Indexed: 12/21/2022]
Abstract
Accumulating evidence shows that the anti-cancer potential of the immune response that can be activated by modulation of the immunogenicity of dying cancer cells. This regulated cell death process is called immunogenic cell death (ICD) and constitutes a new innovating anti-cancer strategy with immune-modulatory potential thanks to the release of damage-associated molecular patterns (DAMPs). Some conventional clinically-used chemotherapeutic drugs, as well as preclinically-investigated compounds of natural origins such as anthracyclines, microtubule-destabilizing agents, cardiac glycosides or hypericin derivatives, possess such an immune-stimulatory function by triggering ICD. Here, we discuss the effects of ICD inducers on the release of DAMPs and the activation of corresponding signaling pathways triggering immune recognition. We will discuss potential strategies allowing to overcome resistance mechanisms associated with this treatment approach as well as co-treatment strategies to overcome the immunosuppressive microenvironment. We will highlight the potential role of metronomic immune modulation as well as targeted delivery of ICD-inducing compounds with nanoparticles or liposomal formulations to improving the immunogenicity of ICD inducers aiming at long-term clinical benefits.
Collapse
Affiliation(s)
- Marc Diederich
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Building 29 Room 223, 1 Gwanak-ro, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|