101
|
Tang T, Huang X, Zhang G, Hong Z, Bai X, Liang T. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct Target Ther 2021; 6:72. [PMID: 33608497 PMCID: PMC7896069 DOI: 10.1038/s41392-020-00449-4] [Citation(s) in RCA: 263] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/31/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Despite great success in cancer immunotherapy, immune checkpoint-targeting drugs are not the most popular weapon in the armory of cancer therapy. Accumulating evidence suggests that the tumor immune microenvironment plays a critical role in anti-cancer immunity, which may result in immune checkpoint blockade therapy being ineffective, in addition to other novel immunotherapies in cancer patients. In the present review, we discuss the deficiencies of current cancer immunotherapies. More importantly, we highlight the critical role of tumor immune microenvironment regulators in tumor immune surveillance, immunological evasion, and the potential for their further translation into clinical practice. Based on their general targetability in clinical therapy, we believe that tumor immune microenvironment regulators are promising cancer immunotherapeutic targets. Targeting the tumor immune microenvironment, alone or in combination with immune checkpoint-targeting drugs, might benefit cancer patients in the future.
Collapse
Affiliation(s)
- Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China.
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China.
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Zhengtao Hong
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Xueli Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China.
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China.
| |
Collapse
|
102
|
The bovine dialysable leukocyte extract IMMUNEPOTENT CRP induces immunogenic cell death in breast cancer cells leading to long-term antitumour memory. Br J Cancer 2021; 124:1398-1410. [PMID: 33531687 PMCID: PMC8039030 DOI: 10.1038/s41416-020-01256-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 12/01/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
Background Cancer recurrence is a serious problem in breast cancer (BC) patients, and immunogenic cell death (ICD) has been proposed as a strategy to overcome this recurrence. IMMUNEPOTENT CRP (ICRP) acts as an immunomodulator and can be cytotoxic to cancer cells. Thus, we evaluated if ICRP induces ICD in BC cells. Methods Immunogenicity of ICRP-induced cell death was evaluated in vitro, analysing the principal biochemical characteristics of ICD in MCF-7, MDA-MB-231 and 4T1 cells. Ex vivo, we assessed the ability of killed cancer cells (KCC) obtained from ICRP-treated 4T1 cells (ICRP-KCC) to induce DC maturation, T-cell priming and T-cell-mediated cancer cytotoxicity. In vivo, we evaluated tumour establishment and antitumour immune memory after prophylactic ICRP-KCC vaccination in BALB/c mice. Results ICRP induced caspase-independent, ROS-dependent cell death, autophagosome formation, P-eIF2α, chaperone protein exposure, CD47 loss, ATP and HMBG1 release in BC cells. Additionally, ICRP-KCC promoted DC maturation, which triggered T-cell priming and cancer cytotoxicity. Prophylactic vaccination with ICRP-KCC prevented tumour establishment and induced long-term antitumour memory in BALB/c mice, involving DC maturation in lymph nodes, CD8+ T-cell augmentation in lymph nodes, peripheral blood and tumour site and ex vivo tumour-specific cytotoxicity by splenocytes. Conclusions ICRP induces ICD in BC cells, leading to long-term antitumour memory.
Collapse
|
103
|
Natural Compounds of Marine Origin as Inducers of Immunogenic Cell Death (ICD): Potential Role for Cancer Interception and Therapy. Cells 2021; 10:cells10020231. [PMID: 33504012 PMCID: PMC7912082 DOI: 10.3390/cells10020231] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/22/2022] Open
Abstract
Regulated cell death (RCD) has always been considered a tolerogenic event. Immunogenic cell death (ICD) occurs as a consequence of tumour cell death accompanied by the release of damage-associated molecular patterns (DAMPs), triggering an immune response. ICD plays a major role in stimulating the function of the immune system in cancer during chemotherapy and radiotherapy. ICD can therefore represent one of the routes to boost anticancer immune responses. According to the recommendations of the Nomenclature Committee on Cell Death (2018), apoptosis (type I cell death) and necrosis (type II cell death) represent are not the only types of RCD, which also includes necroptosis, pyroptosis, ferroptosis and others. Specific downstream signalling molecules and death-inducing stimuli can regulate distinct forms of ICD, which develop and promote the immune cell response. Dying cells deliver different potential immunogenic signals, such as DAMPs, which are able to stimulate the immune system. The acute exposure of DAMPs can prime antitumour immunity by inducing activation of antigen-presenting cells (APC), such as dendritic cells (DC), leading to the downstream response by cytotoxic T cells and natural killer cells (NK). As ICD represents an important target to direct and develop new pharmacological interventions, the identification of bioactive natural products, which are endowed with low side effects, higher tolerability and preferentially inducing immunogenic programmed cell death, represents a priority in biomedical research. The ability of ICD to drive the immune response depends on two major factors, neither of which is intrinsic to cell death: ‘Antigenicity and adjuvanticity’. Indeed, the use of natural ICD-triggering molecules, alone or in combination with different (immuno)therapies, can result in higher efficacy and tolerability. Here, we focused on natural (marine) compounds, particularly on marine microalgae derived molecules such as exopolysaccharides, sulphated polysaccharides, glycopeptides, glycolipids, phospholipids, that are endowed with ICD-inducing properties and sulfavants. Here, we discuss novel and repurposed small-molecule ICD triggers, as well as their ability to target important molecular pathways including the IL-6, TNF-α and interferons (IFNs), leading to immune stimulation, which could be used alone or in combinatorial immunotherapeutic strategies in cancer prevention and therapies.
Collapse
|
104
|
Zhang X, He T, Li Y, Chen L, Liu H, Wu Y, Guo H. Dendritic Cell Vaccines in Ovarian Cancer. Front Immunol 2021; 11:613773. [PMID: 33584699 PMCID: PMC7874064 DOI: 10.3389/fimmu.2020.613773] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/04/2020] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer (OC) is one of the most lethal malignant gynecologic tumors, characterized by an uncertain presentation and poor outcomes. With or without neoadjuvant chemotherapy, surgery followed by platinum-based chemotherapy and maintenance therapy are the basis for the treatment of ovarian cancer patients, but the outcome is still highly restricted by their advanced stage when diagnosed and high recurrence rate after chemotherapy. To enhance the anti-tumor effect and postpone recurrence, anti-VEGF agents and PARP inhibitors are suggested as maintenance therapy, but the population that can benefit from these treatments is small. Based on the interactions of immune cells in the tumor microenvironment, immunotherapies are being explored for ovarian cancer treatment. Disappointingly, the immune checkpoint inhibitors show relatively low responses in ovarian cancer. As shown in several studies that have uncovered a relationship between DC infiltration and outcome in ovarian cancer patients, dendritic cell (DC)-based treatments might have a potential effect on ovarian cancer. In this review, we summarize the functions of dendritic cells (DCs) in the tumor microenvironment, as well as the responses and drawbacks of existing clinical studies to draw a comprehensive picture of DC vaccine treatment in ovarian cancer and to discuss the promising future of immune biomarkers.
Collapse
Affiliation(s)
- Xi Zhang
- Department of OB/GYN, Peking University Third Hospital, Beijing, China
| | - Tianhui He
- Department of OB/GYN, Peking University Third Hospital, Beijing, China
| | - Yuan Li
- Department of OB/GYN, Peking University Third Hospital, Beijing, China
| | - Ling Chen
- Department of Neurosurgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongyu Liu
- Department of Neurosurgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Yu Wu
- Department of OB/GYN, Peking University Third Hospital, Beijing, China
| | - Hongyan Guo
- Department of OB/GYN, Peking University Third Hospital, Beijing, China
| |
Collapse
|
105
|
Huang Z, Hu H. Arginine Deiminase Induces Immunogenic Cell Death and Is Enhanced by N-acetylcysteine in Murine MC38 Colorectal Cancer Cells and MDA-MB-231 Human Breast Cancer Cells In Vitro. Molecules 2021; 26:511. [PMID: 33478072 PMCID: PMC7835909 DOI: 10.3390/molecules26020511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
The use of arginine deiminase (ADI) for arginine depletion therapy is an attractive anticancer approach. Combination strategies are needed to overcome the resistance of severe types of cancer cells to this monotherapy. In the current study, we report, for the first time, that the antioxidant N-acetylcysteine (NAC), which has been used in therapeutic practices for several decades, is a potent enhancer for targeted therapy that utilizes arginine deiminase. We demonstrated that pegylated arginine deiminase (ADI-PEG 20) induces apoptosis and G0/G1 phase arrest in murine MC38 colorectal cancer cells; ADI-PEG 20 induces Ca2+ overload and decreases the mitochondrial membrane potential in MC38 cells. ADI-PEG 20 induced the most important immunogenic cell death (ICD)-associated feature: cell surface exposure of calreticulin (CRT). The antioxidant NAC enhanced the antitumor activity of ADI-PEG 20 and strengthened its ICD-associated features including the secretion of high mobility group box 1 (HMGB1) and adenosine triphosphate (ATP). In addition, these regimens resulted in phagocytosis of treated MC38 cancer cells by bone marrow-derived dendritic cells (BMDCs). In conclusion, we describe, for the first time, that NAC in combination with ADI-PEG 20 not only possesses unique cytotoxic anticancer properties but also triggers the hallmarks of immunogenic cell death. Hence, ADI-PEG 20 in combination with NAC may represent a promising approach to treat ADI-sensitive tumors while preventing relapse and metastasis.
Collapse
Affiliation(s)
- Zhiying Huang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China;
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Haifeng Hu
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| |
Collapse
|
106
|
Wang L, Guan R, Xie L, Liao X, Xiong K, Rees TW, Chen Y, Ji L, Chao H. An ER‐Targeting Iridium(III) Complex That Induces Immunogenic Cell Death in Non‐Small‐Cell Lung Cancer. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013987] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Lili Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Ruilin Guan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Lina Xie
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Xinxing Liao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Thomas W. Rees
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| |
Collapse
|
107
|
Wang L, Guan R, Xie L, Liao X, Xiong K, Rees TW, Chen Y, Ji L, Chao H. An ER-Targeting Iridium(III) Complex That Induces Immunogenic Cell Death in Non-Small-Cell Lung Cancer. Angew Chem Int Ed Engl 2021; 60:4657-4665. [PMID: 33217194 DOI: 10.1002/anie.202013987] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Indexed: 01/01/2023]
Abstract
Immunogenic cell death (ICD) is a vital component of therapeutically induced anti-tumor immunity. An iridium(III) complex (Ir1), containing an N,N-bis(2-chloroethyl)-azane derivate, as an endoplasmic reticulum-localized ICD inducer for non-small cell lung cancer (NSCLC) is reported. The characteristic discharge of damage-associated molecular patterns (DAMPs), that is, cell surface exposure of calreticulin (CRT), extracellular exclusion of high mobility group box 1 (HMGB1), and ATP, were generated by Ir1 in A549 lung cancer cells, accompanied by an increase in endoplasmic reticulum stress and reactive oxygen species (ROS). The vaccination of immunocompetent mice with Ir1-treated dying cells elicited an antitumor CD8+ T cell response and Foxp3+ T cell depletion, which eventually resulted in long-acting anti-tumor immunity by the activation of ICD in lung cancer cells. Ir1 is the first Ir-based complex that is capable of developing an immunomodulatory response by immunogenic cell death.
Collapse
Affiliation(s)
- Lili Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Ruilin Guan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Lina Xie
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Xinxing Liao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Thomas W Rees
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| |
Collapse
|
108
|
Lenzo FL, Kato S, Pabla S, DePietro P, Nesline MK, Conroy JM, Burgher B, Glenn ST, Kuvshinoff B, Kurzrock R, Morrison C. Immune profiling and immunotherapeutic targets in pancreatic cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:119. [PMID: 33569421 PMCID: PMC7867882 DOI: 10.21037/atm-20-1076] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Immunotherapeutic approaches for pancreatic ductal adenocarcinoma (PDAC) are less successful as compared to many other tumor types. In this study, comprehensive immune profiling was performed in order to identify novel, potentially actionable targets for immunotherapy. Methods Formalin-fixed paraffin embedded (FFPE) specimens from 68 patients were evaluated for expression of 395 immune-related markers (RNA-seq), mutational burden by complete exon sequencing of 409 genes, PD-L1 expression by immunohistochemistry (IHC), pattern of tumor infiltrating lymphocytes (TILs) infiltration by CD8 IHC, and PD-L1/L2 copy number by fluorescent in situ hybridization (FISH). Results The seven classes of actionable genes capturing myeloid immunosuppression, metabolic immunosuppression, alternative checkpoint blockade, CTLA-4 immune checkpoint, immune infiltrate, and programmed cell death 1 (PD-1) axis immune checkpoint, discerned 5 unique clinically relevant immunosuppression expression profiles (from most to least common): (I) combined myeloid and metabolic immunosuppression [affecting 25 of 68 patients (36.8%)], (II) multiple immunosuppressive mechanisms (29.4%), (III) PD-L1 positive (20.6%), (IV) highly inflamed PD-L1 negative (10.3%); and (V) immune desert (2.9%). The Wilcoxon rank-sum test was used to compare the PDAC cohort with a comparison cohort (n=1,416 patients) for the mean expressions of the 409 genes evaluated. Multiple genes including TIM3, VISTA, CCL2, CCR2, TGFB1, CD73, and CD39 had significantly higher mean expression versus the comparison cohort, while three genes (LAG3, GITR, CD38) had significantly lower mean expression. Conclusions This study demonstrates that a clinically relevant unique profile of immune markers can be identified in PDAC and be used as a roadmap for personalized immunotherapeutic decision-making strategies.
Collapse
Affiliation(s)
| | - Shumei Kato
- Center for Personalized Cancer Therapy, Moores Cancer Center, La Jolla, CA, USA
| | | | | | | | - Jeffrey M Conroy
- OmniSeq, Inc., Buffalo, NY, USA.,Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Sean T Glenn
- OmniSeq, Inc., Buffalo, NY, USA.,Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Boris Kuvshinoff
- Department of Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Moores Cancer Center, La Jolla, CA, USA
| | - Carl Morrison
- OmniSeq, Inc., Buffalo, NY, USA.,Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
109
|
Hayashi K, Nikolos F, Lee YC, Jain A, Tsouko E, Gao H, Kasabyan A, Leung HE, Osipov A, Jung SY, Kurtova AV, Chan KS. Tipping the immunostimulatory and inhibitory DAMP balance to harness immunogenic cell death. Nat Commun 2020; 11:6299. [PMID: 33288764 PMCID: PMC7721802 DOI: 10.1038/s41467-020-19970-9] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Induction of tumor cell death is the therapeutic goal for most anticancer drugs. Yet, a mode of drug-induced cell death, known as immunogenic cell death (ICD), can propagate antitumoral immunity to augment therapeutic efficacy. Currently, the molecular hallmark of ICD features the release of damage-associated molecular patterns (DAMPs) by dying cancer cells. Here, we show that gemcitabine, a standard chemotherapy for various solid tumors, triggers hallmark immunostimualtory DAMP release (e.g., calreticulin, HSP70, and HMGB1); however, is unable to induce ICD. Mechanistic studies reveal gemcitabine concurrently triggers prostaglandin E2 release as an inhibitory DAMP to counterpoise the adjuvanticity of immunostimulatory DAMPs. Pharmacological blockade of prostaglandin E2 biosythesis favors CD103+ dendritic cell activation that primes a Tc1-polarized CD8+ T cell response to bolster tumor rejection. Herein, we postulate that an intricate balance between immunostimulatory and inhibitory DAMPs could determine the outcome of drug-induced ICD and pose COX-2/prostaglandin E2 blockade as a strategy to harness ICD.
Collapse
Affiliation(s)
- K Hayashi
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - F Nikolos
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Y C Lee
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei City, Taiwan
| | - A Jain
- Alkek Center for Molecular Discovery, Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - E Tsouko
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - H Gao
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - A Kasabyan
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - H E Leung
- Alkek Center for Molecular Discovery, Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - A Osipov
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - S Y Jung
- Alkek Center for Molecular Discovery, Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - A V Kurtova
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - K S Chan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
110
|
Huang X, Tang T, Zhang G, Hong Z, Xu J, Yadav DK, Bai X, Liang T. Genomic investigation of co-targeting tumor immune microenvironment and immune checkpoints in pan-cancer immunotherapy. NPJ Precis Oncol 2020; 4:29. [PMID: 33299118 PMCID: PMC7666137 DOI: 10.1038/s41698-020-00136-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Drugs that target immune checkpoints (ICPs) have become the most popular weapons in cancer immunotherapy; however, they are only beneficial for a small fraction of patients. Accumulating evidence suggests that the tumor immune microenvironment (TIME) plays a critical role in anti-cancer immunity. This study aimed to assess the potential merits and feasibility of combinational targeting ICPs and TIME in cancer immunotherapy. A total of 31 cancer type-specific datasets in TCGA were individually collected by the publicly available web servers for multiple bioinformatic analyses of ICPs and TIME factors. GEPIA was used to calculate the prognostic indexes, STRING was used to construct protein-protein interactions, cBioPortal was used for visualization and comparison of genetic alterations, and TISIDB was used to explore the correlation to tumor-infiltrating lymphocytes (TILs). Intriguingly, TIME factors were identified to have more global coverage and prognostic significance across multiple cancer types compared with ICPs, thus offering more general targetability in clinical therapy. Moreover, TIME factors showed interactive potential with ICPs, and genomic alteration of TIME factors coupled with that of ICPs, at least in pancreatic cancer. Furthermore, TIME factors were found to be significantly associated with TILs, including but not limited to pancreatic cancer. Finally, the clinical significance and translational potential of further combination therapies that incorporate both ICP inhibitors and TIME factor-targeted treatments were discussed. Together, TIME factors are promising immunotherapeutic targets, and a combination strategy of TIME factors-targeted therapies with ICP inhibitors may benefit more cancer patients in the future.
Collapse
Affiliation(s)
- Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China.
| | - Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Zhengtao Hong
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Jian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Dipesh Kumar Yadav
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Xueli Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China.
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
111
|
Wang Y, Xie W, Humeau J, Chen G, Liu P, Pol J, Zhang Z, Kepp O, Kroemer G. Autophagy induction by thiostrepton improves the efficacy of immunogenic chemotherapy. J Immunother Cancer 2020; 8:jitc-2019-000462. [PMID: 32221018 PMCID: PMC7206967 DOI: 10.1136/jitc-2019-000462] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) is a peculiar modality of cellular demise that elicits adaptive immune responses and triggers T cell-dependent immunity. METHODS Fluorescent biosensors were employed for an unbiased drug screen approach aiming at the identification of ICD enhancers. RESULTS Here, we discovered thiostrepton as an enhancer of ICD able to boost chemotherapy-induced ATP release, calreticulin exposure and high-mobility group box 1 exodus. Moreover, thiostrepton enhanced anticancer immune responses of oxaliplatin (OXA) in vivo in immunocompetent mice, yet failed to do so in immunodeficient animals. Consistently, thiostrepton combined with OXA altered the ratio of cytotoxic T lymphocytes to regulatory T cells, thus overcoming immunosuppression and reinstating anticancer immunosurveillance. CONCLUSION Altogether, these results indicate that thiostrepton can be advantageously combined with chemotherapy to enhance anticancer immunogenicity.
Collapse
Affiliation(s)
- Yan Wang
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Xie
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Juliette Humeau
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Guo Chen
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- College of Life Sciences, Nankai University, Tianjin, China
| | - Peng Liu
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Oliver Kepp
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Guido Kroemer
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Pôle de Biologie, Paris, France, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
112
|
Song X, Zhou Z, Li H, Xue Y, Lu X, Bahar I, Kepp O, Hung MC, Kroemer G, Wan Y. Pharmacologic Suppression of B7-H4 Glycosylation Restores Antitumor Immunity in Immune-Cold Breast Cancers. Cancer Discov 2020; 10:1872-1893. [PMID: 32938586 DOI: 10.1158/2159-8290.cd-20-0402] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/15/2020] [Accepted: 09/11/2020] [Indexed: 11/16/2022]
Abstract
Despite widespread utilization of immunotherapy, treating immune-cold tumors has proved to be a challenge. Here, we report that expression of the immune checkpoint molecule B7-H4 is prevalent among immune-cold triple-negative breast cancers (TNBC), where its expression inversely correlates with that of PD-L1. Glycosylation of B7-H4 interferes with its interaction/ubiquitination by AMFR, resulting in B7-H4 stabilization. B7-H4 expression inhibits doxorubicin-induced cell death through the suppression of eIF2α phosphorylation required for calreticulin exposure vis-à-vis the cancer cells. NGI-1, which inhibits B7-H4 glycosylation causing its ubiquitination and subsequent degradation, improves the immunogenic properties of cancer cells treated with doxorubicin, enhancing their phagocytosis by dendritic cells and their capacity to elicit CD8+ IFNγ-producing T-cell responses. In preclinical models of TNBC, a triple combination of NGI-1, camsirubicin (a noncardiotoxic doxorubicin analogue) and PD-L1 blockade was effective in reducing tumor growth. Collectively, our findings uncover a strategy for targeting the immunosuppressive molecule B7-H4. SIGNIFICANCE: This work unravels the regulation of B7-H4 stability by ubiquitination and glycosylation, which affects tumor immunogenicity, particularly regarding immune-cold breast cancers. The inhibition of B7-H4 glycosylation can be favorably combined with immunogenic chemotherapy and PD-L1 blockade to achieve superior immuno-infiltration of cold tumors, as well as improved tumor growth control.See related commentary by Pearce and Läubli, p. 1789.This article is highlighted in the In This Issue feature, p. 1775.
Collapse
Affiliation(s)
- Xinxin Song
- Department of Obstetrics and Gynecology, Department of Pharmacology, the Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Process Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Zhuan Zhou
- Department of Obstetrics and Gynecology, Department of Pharmacology, the Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Process Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Hongchun Li
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China.,Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yifan Xue
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Oliver Kepp
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Paris, France
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taiwan
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Beijing, China.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Yong Wan
- Department of Obstetrics and Gynecology, Department of Pharmacology, the Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Process Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
113
|
Zhou L, Zhang P, Wang H, Wang D, Li Y. Smart Nanosized Drug Delivery Systems Inducing Immunogenic Cell Death for Combination with Cancer Immunotherapy. Acc Chem Res 2020; 53:1761-1772. [PMID: 32819102 DOI: 10.1021/acs.accounts.0c00254] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cancer immunotherapy, which suppresses tumor relapse and metastasis by boosting host immunity and inducing long-term immune memory effects, is emerging as a vital approach to improve the prognosis of patients. Although remarkable efficacy has been observed in some patients, challenges including low response rate, drug resistance, and immune-related adverse effects still limit the clinical application of cancer immunotherapy in broad types of tumors. Immunotherapeutic agents are used to enhance tumor immunogenicity and reverse the effects of the immunosuppressive tumor microenvironment (ITM), but the benefits of monotherapy are mild and transient due to off-target distribution of drugs. To overcome these issues, smart nanosized drug delivery systems (sNDDS) have been developed to enhance tissue specificity, co-deliver multiple drugs, prime immune cells, and amplify immune responses in tumors. Moreover, accumulating knowledge in cancer biology, immunology, and material science has also greatly promoted the development of sNDDS for enhancing cancer immunotherapy.In this Account, we will discuss the approaches of our group in designing sNDDS to induce immunogenic cell death (ICD) for combination with cancer immunotherapy. We propose a brief overview on the design of nanocarriers, intelligent moieties and immunotherapeutic agents in sNDDS. Then, we discuss the strategies to remodel ITM by leveraging ICD as well as cooperating with programmed cell death protein 1 ligand blockade and indoleamine 2,3-dioxygenase 1 inhibition. We have synthesized a series of stimuli-responsive polymers and prodrugs to fabricate sNDDS and have integrated multiple immunotherapeutic drugs into one platform for combinational immunotherapy. Last, we present an outlook on future design of sNDDS and possible directions for enhancing cancer immunotherapy. Building on the concept of enhancing tumor immunogenicity and reversing ITM, we hope this Account will contribute to the rational design of sNDDS for co-delivery of multiple drugs with amplified immunotherapeutic efficacy.
Collapse
Affiliation(s)
- Lei Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China
| | - Hao Wang
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Dangge Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China
| |
Collapse
|
114
|
Casadei B, Argnani L, Morigi A, Lolli G, Broccoli A, Pellegrini C, Nanni L, Stefoni V, Coppola PE, Carella M, Cavo M, Zinzani PL. Effectiveness of chemotherapy after anti-PD-1 blockade failure for relapsed and refractory Hodgkin lymphoma. Cancer Med 2020; 9:7830-7836. [PMID: 32881376 PMCID: PMC7643640 DOI: 10.1002/cam4.3262] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/03/2020] [Accepted: 06/10/2020] [Indexed: 12/22/2022] Open
Abstract
Programmed death‐1 (PD1) blockade is an efficient and safe therapeutic option in patients with relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL). However, a substantial proportion of patients’ progresses or loses the response to anti‐PD1 treatment. We retrospectively investigated the effectiveness of salvage chemotherapies (CHT) for unsatisfactory response to anti‐PD1, in 25 R/R cHL patients. Twenty‐three patients (92%) were refractory to the last treatment before anti‐PD1. After a median of 14 cycles (range 3‐52), 68% (17/25) of patients had unsatisfactory responses to anti‐PD1 therapy, whereas 6 had a partial response (PR) and 2 patients achieved complete response (CR), with an overall response rate (ORR) of 32%. After a median time of 1.5 months, 15 patients received a single agent treatment and 10 had a multi‐agents regimen, due to the failure of PD1 blockade. The ORR was 60% (8 CR and 7 PR). Seven patients (3 in PR and 4 in CR) underwent a consolidation strategy with stem cell transplantation. Median progression‐free survival (PFS) with salvage treatment was reached at 19.1 months, while median PFS after anti‐PD1 has been reached at 8.2 months. After a median follow‐up of 32.4 months, 6 patients died while 13 are still in CR. The median overall estimated from the start of CHT was not reached. The efficacy of treatment following anti‐PD1 is not yet established, especially in lymphoma patients. To note, in our series, a subset of heavily pre‐treated and chemo‐refractory patients increased response rates to and survival with CHT given after exposure to immune‐checkpoint inhibitors.
Collapse
Affiliation(s)
- Beatrice Casadei
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Lisa Argnani
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Alice Morigi
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Ginevra Lolli
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Alessandro Broccoli
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Cinzia Pellegrini
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Laura Nanni
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Vittorio Stefoni
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Paolo E Coppola
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Matteo Carella
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Michele Cavo
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Pier Luigi Zinzani
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| |
Collapse
|
115
|
Lathwal A, Kumar R, Raghava GP. OvirusTdb: A database of oncolytic viruses for the advancement of therapeutics in cancer. Virology 2020; 548:109-116. [DOI: 10.1016/j.virol.2020.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 12/28/2022]
|
116
|
Zhu H, Shan Y, Ge K, Lu J, Kong W, Jia C. Oxaliplatin induces immunogenic cell death in hepatocellular carcinoma cells and synergizes with immune checkpoint blockade therapy. Cell Oncol (Dordr) 2020; 43:1203-1214. [PMID: 32797385 DOI: 10.1007/s13402-020-00552-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common and devastating malignancies. Oxaliplatin, a platinum-based chemotherapeutic agent, is approved for the treatment of several malignancies, including HCC. However, its role in HCC is not well established. This study was designed to investigate the potential of oxaliplatin as an immunogenic cell death (ICD) inducer and to explore its regulatory effects on the response of HCC to immune checkpoint blockade therapy. METHODS Murine and human HCC cells were treated with oxaliplatin, followed by evaluation of the expression of ICD-related biomarkers. Murine HCC cells (H22) were subcutaneously inoculated into mice to establish a syngeneic tumor graft model, after which tumor sizes and in vivo immune cell activation were evaluated. To assess putative synergistic effects of oxaliplatin with anti-PD-1 antibodies on H22 tumors, tumor parameters and secreted cytokines were quantified. RESULTS ICD-related biomarkers were found to be enhanced after treatment of human and murine HCC cells with oxaliplatin. Additionally, we found that the number of mature dendritic cells (DCs) was increased after immature DCs were cocultured with oxaliplatin-treated H22 cells. The numbers of CD8+ T cells and mature DCs were found to be increased in vivo whereas, in contrast, the number of Treg cells was decreased. The tumor sizes were smaller in the oxaliplatin group than in the control group. In the syngeneic tumor graft model, we found that combination therapy with oxaliplatin and anti-PD-1 antibodies could achieve better outcomes than monotherapy, as indicated by (i) inhibition of tumor growth and TGF-β secretion and (ii) augmentation of inflammatory cytokine secretion. CONCLUSIONS Our data indicate that oxaliplatin can be used as an inducer of ICD and as a modulator of the tumor immune microenvironment. Combination therapies composed of oxaliplatin and immune checkpoint inhibitors may open up novel avenues for the treatment of HCC.
Collapse
Affiliation(s)
- Hanzhang Zhu
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, the Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Yuqiang Shan
- Department of Gastrointestinal Surgery, Hangzhou First People's Hospital, the Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Ke Ge
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, the Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Jun Lu
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, the Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Wencheng Kong
- Department of Gastrointestinal Surgery, Hangzhou First People's Hospital, the Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Changku Jia
- Department of Hepatopancreatobiliary Surgery, Hangzhou First People's Hospital, the Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
117
|
Qin X, Denton WD, Huiting LN, Smith KS, Feng H. Unraveling the regulatory role of endoplasmic-reticulum-associated degradation in tumor immunity. Crit Rev Biochem Mol Biol 2020; 55:322-353. [PMID: 32633575 DOI: 10.1080/10409238.2020.1784085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During malignant transformation and cancer progression, tumor cells face both intrinsic and extrinsic stress, endoplasmic reticulum (ER) stress in particular. To survive and proliferate, tumor cells use multiple stress response pathways to mitigate ER stress, promoting disease aggression and treatment resistance. Among the stress response pathways is ER-associated degradation (ERAD), which consists of multiple components and steps working together to ensure protein quality and quantity. In addition to its established role in stress responses and tumor cell survival, ERAD has recently been shown to regulate tumor immunity. Here we summarize current knowledge on how ERAD promotes protein degradation, regulates immune cell development and function, participates in antigen presentation, exerts paradoxical roles on tumorigenesis and immunity, and thus impacts current cancer therapy. Collectively, ERAD is a critical protein homeostasis pathway intertwined with cancer development and tumor immunity. Of particular importance is the need to further unveil ERAD's enigmatic roles in tumor immunity to develop effective targeted and combination therapy for successful treatment of cancer.
Collapse
Affiliation(s)
- Xiaodan Qin
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - William D Denton
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - Leah N Huiting
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - Kaylee S Smith
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - Hui Feng
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
118
|
Repurposing Food and Drug Administration-Approved Drugs to Promote Antitumor Immunity. ACTA ACUST UNITED AC 2020; 25:88-99. [PMID: 30896530 DOI: 10.1097/ppo.0000000000000368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
There has been a major resurgence of interest in immune-based approaches to treat cancer, based largely on the success of checkpoint inhibitors (anti-cytotoxic T-lymphocyte-associated antigen 4, anti-programmed cell death 1, and anti-programmed cell death ligand 1 antibodies) in several malignancies. However, not all tumors respond to checkpoint therapy, and there is clearly a need for additional approaches for enhancing tumor immunity. We summarize the critical elements necessary for mounting an efficacious T-cell response to a tumor. We cite drugs approved by the Food and Drug Administration for no-cancer indications that could be repurposed and used as part of an antitumor immune cocktail. We also list cancer drugs not initially intended to impact tumor immunity (soft repurposing) but that have been found to modulate the immune system. We highlight those drugs that might be used in combination with checkpoint inhibitors to increase response rates and survival of cancer patients. Our focus will be on drugs for which there are limited but existing human data. We cite supporting mechanistic mouse data as well. Repurposing drugs to modulate antitumor immunity is an opportunity to rapidly bring new, effective, and affordable treatments to cancer patients.
Collapse
|
119
|
Sun Y, Feng X, Wan C, Lovell JF, Jin H, Ding J. Role of nanoparticle-mediated immunogenic cell death in cancer immunotherapy. Asian J Pharm Sci 2020; 16:129-132. [PMID: 33995609 PMCID: PMC8105413 DOI: 10.1016/j.ajps.2020.05.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/15/2020] [Accepted: 05/31/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy, which suppresses cancer progression by activating the anti-cancer immunity of patients, shows utility in treating multiple types of cancers. Immunogenic cell death (ICD) induced by most clinical treatment modalities plays a critical role in promoting cancer immunotherapy by releasing tumor-associated antigens and neoantigens and exposing “danger signals” to stimulate immune cells. This comment article presents the different roles of nanoparticles in various treatment modalities of cancers, including chemotherapy, radiotherapy, photodynamic and photothermal therapies, and therapy with radiated tumor cell-released nanoparticles, which often activate anti-cancer immunological effects by inducing ICD of cancer cells, and highlights the challenges and opportunities of ICD-related cancer immunotherapy in the clinic.
Collapse
Affiliation(s)
- Yajie Sun
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangru Feng
- China Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Chao Wan
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York. Buffalo, New York 14260, USA
| | - Honglin Jin
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jianxun Ding
- China Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
120
|
Oliveira MMS, Westerberg LS. Cytoskeletal regulation of dendritic cells: An intricate balance between migration and presentation for tumor therapy. J Leukoc Biol 2020; 108:1051-1065. [PMID: 32557835 DOI: 10.1002/jlb.1mr0520-014rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/28/2022] Open
Abstract
Dendritic cells (DCs) are the main players in many approaches for cancer therapy. The idea with DC tumor therapy is to promote activation of tumor infiltrating cytotoxic T cells that kill tumor cells. This requires that DCs take up tumor Ag and present peptides on MHC class I molecules in a process called cross-presentation. For this process to be efficient, DCs have to migrate to the tumor draining lymph node and there activate the machinery for cross-presentation. In this review, we will discuss recent progress in understanding the role of actin regulators for control of DC migration and Ag presentation. The potential to target actin regulators for better DC-based tumor therapy will also be discussed.
Collapse
Affiliation(s)
- Mariana M S Oliveira
- Department of Microbiology Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
121
|
Elhassanny AEM, Soliman E, Marie M, McGuire P, Gul W, ElSohly M, Van Dross R. Heme-Dependent ER Stress Apoptosis: A Mechanism for the Selective Toxicity of the Dihydroartemisinin, NSC735847, in Colorectal Cancer Cells. Front Oncol 2020; 10:965. [PMID: 32626657 PMCID: PMC7313430 DOI: 10.3389/fonc.2020.00965] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/15/2020] [Indexed: 01/05/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death in the United States. Artemisinin derivatives, including the dihydroartemisinin (DHA) monomers, are widely used as clinical agents for the treatment of malaria. Numerous studies demonstrate that these molecules also display antineoplastic activity with minimal toxicity. Of interest, dimeric DHA molecules are more active than their monomeric counterparts. Our previous data showed that the DHA dimer, NSC735847, was a potent inducer of death in different cancer cell types. However, the mechanism of action and activity of NSC735847 in colon cancer cells was not explored. The present study investigated the anticancer activity of NSC735847 and four structurally similar analog in human tumorigenic (HT-29 and HCT-116) and non-tumorigenic (FHC) colon cell lines. NSC735847 was more cytotoxic toward tumorigenic than non-tumorigenic colonocytes. In addition, NSC735847 exhibited greater cytotoxicity and tumor selectivity than the NSC735847 derivatives. To gain insight into mechanisms of NSC735847 activity, the requirement for endoplasmic reticulum (ER) stress and oxidative stress was tested. The data show that ER stress played a key role in the cytotoxicity of NSC735847 while oxidative stress had little impact on cell fate. In addition, it was observed that the cytotoxic activity of NSC735847 required the presence of heme, but not iron. The activity of NSC735847 was then compared to clinically utilized CRC therapeutics. NSC735847 was cytotoxic toward colon tumor cells at lower concentrations than oxaliplatin (OX). In addition, cell death was achieved at lower concentrations in colon cancer cells that were co-treated with folinic acid (Fol), 5-FU (F), and NSC735847 (FolFNSC), than Fol, F, and OX (FolFOX). The selective activity of NSC735847 and its ability to induce cytotoxicity at low concentrations suggest that NSC735847 may be an alternative for oxaliplatin in the FolFOX regimen for patients who are unable to tolerate its adverse effects.
Collapse
Affiliation(s)
- Ahmed E M Elhassanny
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Eman Soliman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mona Marie
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Paul McGuire
- Medical Doctor Program, Brody School of Medicine, Greenville, NC, United States
| | - Waseem Gul
- ElSohly Laboratories Inc., Oxford, MS, United States.,National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS, United States
| | - Mahmoud ElSohly
- ElSohly Laboratories Inc., Oxford, MS, United States.,National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS, United States
| | - Rukiyah Van Dross
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,Center for Health Disparities, East Carolina University, Greenville, NC, United States
| |
Collapse
|
122
|
Jian M, Ren L, He G, Lin Q, Tang W, Chen Y, Chen J, Liu T, Ji M, Wei Y, Chang W, Xu J. A novel patient-derived organoids-based xenografts model for preclinical drug response testing in patients with colorectal liver metastases. J Transl Med 2020; 18:234. [PMID: 32532289 PMCID: PMC7291745 DOI: 10.1186/s12967-020-02407-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022] Open
Abstract
Backgrounds Cancer-related mortality in patients with colorectal cancer (CRC) is predominantly caused by development of colorectal liver metastases (CLMs). How to screen the sensitive chemotherapy and targeted therapy is the key element to improve the prognosis of CLMs patients. The study aims to develop patient-derived organoids-based xenografted liver metastases (PDOX-LM) model of CRC, to recapitulate the clinical drug response. Methods We transplanted human CRC primary tumor derived organoids in murine spleen to obtain xenografted liver metastases in murine liver. Immunohistochemistry (IHC) staining, whole-exome and RNA sequencing, and drug response testing were utilized to identify the homogeneity in biological and genetic characteristics, and drug response between the PDOX-LM models and donor liver metastases. Results We successfully established PDOX-LM models from patients with CLMs. IHC staining showed that positive expression of CEA, Ki67, VEGF, FGFR2 in donor liver metastases were also well preserved in matched xenografted liver metastases. Whole-exon sequencing and transcriptome analysis showed that both xenografted and donor liver metastases were highly concordant in somatic variants (≥ 0.90 frequency of concordance) and co-expression of driver genes (Pearson’s correlation coefficient reach up to 0.99, P = 0.001). Furthermore, drug response testing showed that the PDOX-LM models can closely recapitulated the clinical response to mFOLFOX6 regiments. Conclusions This PDOX-LM model provides a more convenient and informative platform for preclinical testing of individual tumors by retaining the histologic and genetic features of donor liver metastases. This technology holds great promise to predict treatment sensitivity for patients with CLMs undergoing chemotherapy.
Collapse
Affiliation(s)
- Mi Jian
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China
| | - Li Ren
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China
| | - Guodong He
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China
| | - Qi Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China
| | - Wentao Tang
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China
| | - Yijiao Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China
| | - Jingwen Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China
| | - Tianyu Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China
| | - Meiling Ji
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China
| | - Ye Wei
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China.,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China
| | - Wenju Chang
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China. .,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China.
| | - Jianmin Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200030, China. .,Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, 200030, China.
| |
Collapse
|
123
|
Humeau J, Sauvat A, Cerrato G, Xie W, Loos F, Iannantuoni F, Bezu L, Lévesque S, Paillet J, Pol J, Leduc M, Zitvogel L, de Thé H, Kepp O, Kroemer G. Inhibition of transcription by dactinomycin reveals a new characteristic of immunogenic cell stress. EMBO Mol Med 2020; 12:e11622. [PMID: 32323922 PMCID: PMC7207166 DOI: 10.15252/emmm.201911622] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy still constitutes the standard of care for the treatment of most neoplastic diseases. Certain chemotherapeutics from the oncological armamentarium are able to trigger pre‐mortem stress signals that lead to immunogenic cell death (ICD), thus inducing an antitumor immune response and mediating long‐term tumor growth reduction. Here, we used an established model, built on artificial intelligence to identify, among a library of 50,000 compounds, anticancer agents that, based on their molecular descriptors, were predicted to induce ICD. This algorithm led us to the identification of dactinomycin (DACT, best known as actinomycin D), a highly potent cytotoxicant and ICD inducer that mediates immune‐dependent anticancer effects in vivo. Since DACT is commonly used as an inhibitor of DNA to RNA transcription, we investigated whether other experimentally established or algorithm‐selected, clinically employed ICD inducers would share this characteristic. As a common leitmotif, a panel of pharmacological ICD stimulators inhibited transcription and secondarily translation. These results establish the inhibition of RNA synthesis as an initial event for ICD induction.
Collapse
Affiliation(s)
- Juliette Humeau
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Faculty of Medicine Kremlin Bicêtre, Université Paris Sud, Paris Saclay, Paris, France
| | - Allan Sauvat
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Giulia Cerrato
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Faculty of Medicine Kremlin Bicêtre, Université Paris Sud, Paris Saclay, Paris, France
| | - Wei Xie
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Faculty of Medicine Kremlin Bicêtre, Université Paris Sud, Paris Saclay, Paris, France
| | - Friedemann Loos
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Francesca Iannantuoni
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Hospital Doctor Peset - FISABIO, Valencia, Spain
| | - Lucillia Bezu
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Hospital Doctor Peset - FISABIO, Valencia, Spain
| | - Sarah Lévesque
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Faculty of Medicine Kremlin Bicêtre, Université Paris Sud, Paris Saclay, Paris, France
| | - Juliette Paillet
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Faculty of Medicine Kremlin Bicêtre, Université Paris Sud, Paris Saclay, Paris, France
| | - Jonathan Pol
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Marion Leduc
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Laurence Zitvogel
- Faculty of Medicine Kremlin Bicêtre, Université Paris Sud, Paris Saclay, Paris, France.,Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,INSERM U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Hugues de Thé
- College de France, INSERM UMR 1050, CNRS UMR 7241, PSL University, Paris, France.,INSERM UMR 944, CNRS UMR 7212, Equipe labellisée par la Ligue contre le Cancer, IRSL, Hopital St. Louis, Université de Paris, Paris, France
| | - Oliver Kepp
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
124
|
Pagès F, André T, Taieb J, Vernerey D, Henriques J, Borg C, Marliot F, Ben Jannet R, Louvet C, Mineur L, Bennouna J, Desrame J, Faroux R, Kirilovsky A, Duval A, Laurent-Puig P, Svrcek M, Hermitte F, Catteau A, Galon J, Emile JF. Prognostic and predictive value of the Immunoscore in stage III colon cancer patients treated with oxaliplatin in the prospective IDEA France PRODIGE-GERCOR cohort study. Ann Oncol 2020; 31:921-929. [PMID: 32294529 DOI: 10.1016/j.annonc.2020.03.310] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The Immunoscore (IS), which prognostically classifies stage I-III colon cancer (CC) patients, was evaluated in the International Duration Evaluation of Adjuvant Therapy (IDEA) France cohort study investigating 3 versus 6 months of oxaliplatin-based adjuvant chemotherapy in stage III CC patients. PATIENTS AND METHODS Densities of CD3+ and CD8+ T cells in the tumor and invasive margin were determined by immunohistochemistry, quantified by digital pathology, and converted to IS. Mismatch repair status was determined by immunohistochemistry or by pentaplex PCR. Prediction of disease-free survival (DFS) by IS was analyzed by a multivariable Cox regression model in each study arm. Harrell's C-statistics were used to investigate the IS performance. RESULTS Samples of 1322 patients were available. IS Low, Intermediate (Int), and High were observed in 43.6%, 47.0%, and 9.4% of patients, respectively. IS Low identified patients at higher risk of relapse or death compared with Int + High [hazard ratio (HR) = 1.54; 95% confidence interval (CI) 1.24-1.93, P = 0.0001]. The 3-year DFS was 66.80% (95% CI 62.23-70.94) for IS Low and 77.14% (95% CI 73.50-80.35) for IS Int + High. In multivariable analysis, IS remained significantly independently associated with DFS (P = 0.003) when adjusted for sex, histological grade, T/N stage, and microsatellite instability. For mFOLFOX6-treated patients (91.6% of the cohort), a statistical significant interaction was observed for the predictive value of IS for treatment duration (3 versus 6 months) in terms of DFS (P = 0.057). IS Int + High significantly predicted benefit of 6 months of treatment (HR = 0.53; 95% CI 0.37-0.75; P = 0.0004), including clinically low- and high-risk stage III CC (all P < 0.001). Conversely, patients with IS Low (46.4%) did not significantly benefit from the 6-month mFOLFOX6 versus the 3-month mFOLFOX6. CONCLUSIONS The prognostic value of IS for DFS was confirmed in patients with stage III CC treated with oxaliplatin-based chemotherapy. Its predictive value for DFS benefit of longer duration of mFOLFOX6 adjuvant treatment was found in IS Int + High. These results will be validated in an external independent cohort. CLINICALTRIALS. GOV REGISTRATION NCT03422601; EudraCT Number: 2009-010384-16.
Collapse
Affiliation(s)
- F Pagès
- Department of Immunology, Européen Georges Pompidou Hospital, AP-HP, Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| | - T André
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne University and AP-HP, Paris, France
| | - J Taieb
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Department of Gastroenterology and Digestive Oncology, Européen Georges Pompidou Hospital, AP-HP, Paris, France
| | - D Vernerey
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France; University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - J Henriques
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France; University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - C Borg
- University Hospital (CHRU) Besançon, UMR1098 UBFC/EFS/INSERM, Immuno-Oncologie et Biotechnologies en Cancérologie, Besançon, France
| | - F Marliot
- Department of Immunology, Européen Georges Pompidou Hospital, AP-HP, Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - R Ben Jannet
- EA4340-Biomarqueurs et essais cliniques en Cancérologie et Onco-Hématologie (BECCOH), Versailles University, Boulogne, France; Ambroise Paré Hospital, AP-HP, Boulogne, France
| | - C Louvet
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - L Mineur
- Department of Radiation Therapy, Institut Sainte Catherine, Avignon, France
| | - J Bennouna
- Department of Medical Oncology, University Hospital of Nantes, Nantes, France
| | - J Desrame
- Department of Medical Oncology, Private Hospital Jean Mermoz, Lyon, France
| | - R Faroux
- Department of Gastroenterology, CHD Vendée, La Roche sur Yon, France
| | - A Kirilovsky
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - A Duval
- Sorbonne University and INSERM, UMRS 938, Équipe Instabilité des microsatellites et cancer, Saint Antoine Research Center, Equipe Labellisée Ligue Contre le Cancer and SIRIC CURAMUS, AP-HP, Paris, France
| | - P Laurent-Puig
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Department of Biology, Européen Georges Pompidou Hospital, AP-HP, Paris, France
| | - M Svrcek
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne University and AP-HP, Paris, France; Sorbonne University and INSERM, UMRS 938, Équipe Instabilité des microsatellites et cancer, Saint Antoine Research Center, Equipe Labellisée Ligue Contre le Cancer and SIRIC CURAMUS, AP-HP, Paris, France
| | | | | | - J Galon
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - J-F Emile
- University Hospital (CHRU) Besançon, UMR1098 UBFC/EFS/INSERM, Immuno-Oncologie et Biotechnologies en Cancérologie, Besançon, France; EA4340-Biomarqueurs et essais cliniques en Cancérologie et Onco-Hématologie (BECCOH), Versailles University, Boulogne, France; Ambroise Paré Hospital, AP-HP, Boulogne, France
| |
Collapse
|
125
|
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: 2.2] [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]
|
126
|
Serrano-Del Valle A, Naval J, Anel A, Marzo I. Novel Forms of Immunomodulation for Cancer Therapy. Trends Cancer 2020; 6:518-532. [PMID: 32460005 DOI: 10.1016/j.trecan.2020.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
In recent years immunotherapy has provided new hope for cancer patients. However, some patients eventually relapse. Immunological responses are thought to underlie the long-term effects of conventional or targeted therapies. Whether this influence emerges from direct effects on cancer cells through immunogenic cell death (ICD) or by modulating the immune environment requires further clarification. ICD-related molecular mechanisms are also shared by cell-intrinsic defense responses that combat foreign intrusions. Indeed, we could potentially mimic and harness these processes to improve cancer immunogenicity. In addition, the microbiome is materializing as a missing factor in the cancer-immune therapy axis. The emerging idea of manipulating the gut microbiota to improve responses to anticancer therapy is becoming increasingly popular, but further clinical authentication is needed.
Collapse
Affiliation(s)
- Alfonso Serrano-Del Valle
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain.
| | - Javier Naval
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| | - Alberto Anel
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| | - Isabel Marzo
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| |
Collapse
|
127
|
Deo KM, Sakoff J, Gilbert J, Zhang Y, Aldrich Wright JR. Synthesis, characterisation and influence of lipophilicity on cellular accumulation and cytotoxicity of unconventional platinum(iv) prodrugs as potent anticancer agents. Dalton Trans 2020; 48:17228-17240. [PMID: 31728483 DOI: 10.1039/c9dt04049h] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Lipophilic platinum(iv) complexes were synthesised of the type [Pt(HL)(AL)(OH)(R)]2+ and [Pt(HL)(AL)(R)2]2+ (HL = 5,6-dimethyl-1,10-phenanthroline or 1,10-phenanthroline; AL = 1S,2S-diaminocyclohexane and R = increasingly lipophilic carboxylate axial ligands (C10-18)) from hydrophilic platinum(ii) precursors that exhibit exceptional anticancer activity. The increased overall lipophilicity of the complexes suggested the formation of spontaneously self-assembled structures in an aqueous environment. The anti-proliferative properties were assessed against one non-cancerous and a panel of cancerous cell lines. Nanomolar levels of activity were observed against several cell lines, with the lowest GI50 of 3.4 nm against the Du145 prostate cancer cell line and over 1100-fold greater activity than cisplatin against HT29 colon carcinoma. RP-HPLC was utilised to establish the relative lipophilicities of each complex. While there seemed to be an increase in cellular accumulation for the lipophilic derivatives in some instances, ICP-MS studies showed no clear correlation between increasing lipophilicity, cellular accumulation and cytotoxicity.
Collapse
Affiliation(s)
- Krishant M Deo
- Nanoscale Organisation and Dynamics Group, Western Sydney University, Campbelltown, NSW 2560, Australia.
| | | | | | | | | |
Collapse
|
128
|
Papadia P, Micoli K, Barbanente A, Ditaranto N, Hoeschele JD, Natile G, Marzano C, Gandin V, Margiotta N. Platinum(IV) Complexes of trans-1,2-diamino-4-cyclohexene: Prodrugs Affording an Oxaliplatin Analogue that Overcomes Cancer Resistance. Int J Mol Sci 2020; 21:E2325. [PMID: 32230896 PMCID: PMC7177638 DOI: 10.3390/ijms21072325] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 11/23/2022] Open
Abstract
Six platinum(IV) compounds derived from an oxaliplatin analogue containing the unsaturated cyclic diamine trans-1,2-diamino-4-cyclohexene (DACHEX), in place of the 1,2-diaminocyclohexane, and a range of axial ligands, were synthesized and characterized. The derivatives with at least one axial chlorido ligand demonstrated solvent-assisted photoreduction. The electrochemical redox behavior was investigated by cyclic voltammetry; all compounds showed reduction potentials suitable for activation in vivo. X-ray photoelectron spectroscopy (XPS) data indicated an X-ray-induced surface reduction of the Pt(IV) substrates, which correlates with the reduction potentials measured by cyclic voltammetry. The cytotoxic activity was assessed in vitro on a panel of human cancer cell lines, also including oxaliplatin-resistant cancer cells, and compared with that of the reference compounds cisplatin and oxaliplatin; all IC50 values were remarkably lower than those elicited by cisplatin and somewhat lower than those of oxaliplatin. Compared to the other Pt(IV) compounds of the series, the bis-benzoate derivative was by far (5-8 times) the most cytotoxic showing that low reduction potential and high lipophilicity are essential for good cytotoxicity. Interestingly, all the complexes proved to be more active than cisplatin and oxaliplatin even in three-dimensional spheroids of A431 human cervical cancer cells.
Collapse
Affiliation(s)
- Paride Papadia
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy
| | - Katia Micoli
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Alessandra Barbanente
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Nicoletta Ditaranto
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - James D. Hoeschele
- Department of Chemistry, Eastern Michigan University, Ypsilanti, MI 48197, USA
| | - Giovanni Natile
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Cristina Marzano
- Dipartimento di Scienze del Farmaco, Università di Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Valentina Gandin
- Dipartimento di Scienze del Farmaco, Università di Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Nicola Margiotta
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
129
|
Khoury A, Deo KM, Aldrich-Wright JR. Recent advances in platinum-based chemotherapeutics that exhibit inhibitory and targeted mechanisms of action. J Inorg Biochem 2020; 207:111070. [PMID: 32299045 DOI: 10.1016/j.jinorgbio.2020.111070] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/22/2022]
Abstract
Current platinum-based drugs used in chemotherapy, like cisplatin and its derivatives, are greatly limited due to side-effects and drug resistance. This has inspired the search for novel platinum-based drugs that deviate from the conventional mechanism of action seen with current chemotherapeutics. This review highlights recent advances in platinum(II) and platinum(IV)-based complexes that have been developed within the past six years. The platinum compounds explored within this review are those that display a more targeted approach by incorporating ligands that act on selected cellular targets within cancer cells. This includes mitochondria, overexpressed receptors or proteins and enzymes that contribute to cancer cell proliferation. These types of platinum compounds have shown significant improvements in anticancer activity and as such, this review highlights the importance of pursuing these new designed platinum drugs for cancer therapy, with the potential of undergoing clinical trials.
Collapse
Affiliation(s)
- Aleen Khoury
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Krishant M Deo
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | | |
Collapse
|
130
|
Matha K, Lollo G, Taurino G, Respaud R, Marigo I, Shariati M, Bussolati O, Vermeulen A, Remaut K, Benoit JP. Bioinspired hyaluronic acid and polyarginine nanoparticles for DACHPt delivery. Eur J Pharm Biopharm 2020; 150:1-13. [PMID: 32113915 DOI: 10.1016/j.ejpb.2020.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/30/2020] [Accepted: 02/21/2020] [Indexed: 12/15/2022]
Abstract
This work here presented provides insights over a novel biodegradable polymeric nanosystem made of hyaluronic acid and polyarginine for diaminocyclohexane-platinum (DACHPt) encapsulation. Using mild conditions based on ionic gelation technique, monodispersed blank and DACHPt-loaded nanoparticles (NP) with a size of around 200 nm and negative ζ potential (-35 mV) were obtained. The freeze-drying process was optimized to improve the stability and shelf-life of the developed nanoparticles. After reconstitution, nanoparticles maintained their size showing an association efficiency of around 70% and a high drug loading (8%). In vitro cytotoxicity studies revealed that DACHPt-loaded nanoparticles had a superior anticancer activity compared with oxaliplatin solution. The IC50 was reduced by a factor of two in HT-29 cells (IC50 39 µM vs 74 µM, respectively), and resulted almost 1.3 fold lower in B6KPC3 cells (18 µM vs 23 µM respectively). Whereas toxic effects of both drug and DACHPt-loaded nanoparticles were comparable in the A549 cell line (IC50 11 µM vs 12 µM). DACHPt-loaded nanoparticles were also able to modulate immunogenic cell death (ICD) in vitro. After incubation with B6KPC3 cells, an increase in HMGB1 (high-mobility group box 1) production associated with ATP release occurred. Then, in vivo pharmacokinetic studies were performed after intravenous injection (IV) of DACHPt-loaded nanoparticles and oxaliplatin solution in healthy mice (35.9 µg of platinum equivalent/mouse). An AUC six times higher (24 h * mg/L) than the value obtained following the administration of oxaliplatin solution (3.76 h * mg/L) was found. Cmax was almost five times higher than the control (11.4 mg/L for NP vs 2.48 mg/L). Moreover, the reduction in volume of distribution and clearance clearly indicated a more limited tissue distribution. A simulated repeated IV regimen was performed in silico and showed no accumulation of platinum from the nanoparticles. Overall, the proposed approach discloses a novel nano-oncological treatment based on platinum derivative with improved antitumor activity in vitro and in vivo stability as compared to the free drug.
Collapse
Affiliation(s)
- Kevin Matha
- Micro et Nanomédecines Translationnelles, MINT, UNIV Angers, UMR INSERM 1066, UMR CNRS 6021, Angers, France; CHU Angers, Département Pharmacie, 4 rue Larrey, 49933 Angers cedex 9, France
| | - Giovanna Lollo
- University of Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Bd 11 Novembre 1918, 69622 Villeurbanne, France
| | - Giuseppe Taurino
- Department of Biomedical, Biotechnological, and Translational Sciences, University of Parma, 43100 Parma, Italy
| | - Renaud Respaud
- Centre d'Étude des Pathologies Respiratoires-CEPR, Institut National de la Santé et de la Recherche Médicale-INSERM, Unité Mixte de Recherche UMR 1100, Labex Mabimprove, 37000 Tours, France; Centre Hospitalier Régional Universitaire-CHRU de Tours, Hôpital Trousseau, Service de Pharmacie, 37170 Chambray-les-Tours, France
| | - Ilaria Marigo
- Istituto Oncologico Veneto, IOV-IRCCS, 35128 Padova, Italy
| | - Molood Shariati
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Ovidio Bussolati
- Department of Biomedical, Biotechnological, and Translational Sciences, University of Parma, 43100 Parma, Italy
| | - An Vermeulen
- Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Jean-Pierre Benoit
- Micro et Nanomédecines Translationnelles, MINT, UNIV Angers, UMR INSERM 1066, UMR CNRS 6021, Angers, France; CHU Angers, Département Pharmacie, 4 rue Larrey, 49933 Angers cedex 9, France.
| |
Collapse
|
131
|
Yamazaki T, Buqué A, Ames TD, Galluzzi L. PT-112 induces immunogenic cell death and synergizes with immune checkpoint blockers in mouse tumor models. Oncoimmunology 2020; 9:1721810. [PMID: 32117585 PMCID: PMC7028345 DOI: 10.1080/2162402x.2020.1721810] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/17/2022] Open
Abstract
PT-112 is a novel platinum-pyrophosphate conjugate under clinical development for cancer therapy. PT-112 mediates cytostatic and cytotoxic effects against a variety of human and mouse cancer cell lines in vitro. The cytotoxic response to PT-112 is associated with the emission of danger signals underpinning the initiation of anticancer immunity, including calreticulin exposure on the surface of dying cells, as well as ATP and HMGB1 secretion. Consistently, mouse cancer cells succumbing to PT-112 in vitro can be used to provide syngeneic, immunocompetent mice with immunological protection against a subsequent challenge with living tumor cells of the same type. Moreover, PT-112 administration synergizes with PD-1 or PD-L1 blockade in the control of mouse cancers in immunologically competent settings, as it simultaneously recruits immune effector cells and depletes immunosuppressive cells in the tumor microenvironment. Finally, PT-112 employed intratumorally in the context of immune checkpoint inhibition initiates a robust immune response that has systemic outreach and limits the growth of untreated, distant lesions. Thus, PT-112 induces the immunogenic demise of cancer cells, and hence stands out as a promising combinatorial partner of immune checkpoint blockers, especially for the treatment of otherwise immunologically cold tumors.
Collapse
Affiliation(s)
- Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | | | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.,Université de Paris, Paris, France
| |
Collapse
|
132
|
Therapeutic Cancer Vaccination with Ex Vivo RNA-Transfected Dendritic Cells-An Update. Pharmaceutics 2020; 12:pharmaceutics12020092. [PMID: 31979205 PMCID: PMC7076681 DOI: 10.3390/pharmaceutics12020092] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/18/2020] [Indexed: 12/19/2022] Open
Abstract
Over the last two decades, dendritic cell (DC) vaccination has been studied extensively as active immunotherapy in cancer treatment and has been proven safe in all clinical trials both with respect to short and long-term side effects. For antigen-loading of dendritic cells (DCs) one method is to introduce mRNA coding for the desired antigens. To target the whole antigenic repertoire of a tumor, even the total tumor mRNA of a macrodissected biopsy sample can be used. To date, reports have been published on a total of 781 patients suffering from different tumor entities and HIV-infection, who have been treated with DCs loaded with mRNA. The majority of those were melanoma patients, followed by HIV-infected patients, but leukemias, brain tumors, prostate cancer, renal cell carcinomas, pancreatic cancers and several others have also been treated. Next to antigen-loading, mRNA-electroporation allows a purposeful manipulation of the DCs’ phenotype and function to enhance their immunogenicity. In this review, we intend to give a comprehensive summary of what has been published regarding clinical testing of ex vivo generated mRNA-transfected DCs, with respect to safety and risk/benefit evaluations, choice of tumor antigens and RNA-source, and the design of better DCs for vaccination by transfection of mRNA-encoded functional proteins.
Collapse
|
133
|
Vanmeerbeek I, Sprooten J, De Ruysscher D, Tejpar S, Vandenberghe P, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: chemotherapy-induced immunogenic cell death in immuno-oncology. Oncoimmunology 2020; 9:1703449. [PMID: 32002302 PMCID: PMC6959434 DOI: 10.1080/2162402x.2019.1703449] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
The term ‘immunogenic cell death’ (ICD) denotes an immunologically unique type of regulated cell death that enables, rather than suppresses, T cell-driven immune responses that are specific for antigens derived from the dying cells. The ability of ICD to elicit adaptive immunity heavily relies on the immunogenicity of dying cells, implying that such cells must encode and present antigens not covered by central tolerance (antigenicity), and deliver immunostimulatory molecules such as damage-associated molecular patterns and cytokines (adjuvanticity). Moreover, the host immune system must be equipped to detect the antigenicity and adjuvanticity of dying cells. As cancer (but not normal) cells express several antigens not covered by central tolerance, they can be driven into ICD by some therapeutic agents, including (but not limited to) chemotherapeutics of the anthracycline family, oxaliplatin and bortezomib, as well as radiation therapy. In this Trial Watch, we describe current trends in the preclinical and clinical development of ICD-eliciting chemotherapy as partner for immunotherapy, with a focus on trials assessing efficacy in the context of immunomonitoring.
Collapse
Affiliation(s)
- Isaure Vanmeerbeek
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Dirk De Ruysscher
- Maastricht University Medical Center, Department of Radiation Oncology (MAASTRO Clinic), GROW-School for Oncology and Developmental Biology, Maastricht, Netherlands
| | - Sabine Tejpar
- Department of Oncology, KU Leuven, Leuven, Belgium.,UZ Leuven, Leuven, Belgium
| | - Peter Vandenberghe
- Department of Haematology, UZ Leuven, and Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.,Université de Paris, Paris, France
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
134
|
Aznar MA, Molina C, Teijeira A, Rodriguez I, Azpilikueta A, Garasa S, Sanchez‐Paulete AR, Cordeiro L, Etxeberria I, Alvarez M, Rius‐Rocabert S, Nistal‐Villan E, Berraondo P, Melero I. Repurposing the yellow fever vaccine for intratumoral immunotherapy. EMBO Mol Med 2020; 12:e10375. [PMID: 31746149 PMCID: PMC6949490 DOI: 10.15252/emmm.201910375] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 11/27/2022] Open
Abstract
Live 17D is widely used as a prophylactic vaccine strain for yellow fever virus that induces potent neutralizing humoral and cellular immunity against the wild-type pathogen. 17D replicates and kills mouse and human tumor cell lines but not non-transformed human cells. Intratumoral injections with viable 17D markedly delay transplanted tumor progression in a CD8 T-cell-dependent manner. In mice bearing bilateral tumors in which only one is intratumorally injected, contralateral therapeutic effects are observed consistent with more prominent CD8 T-cell infiltrates and a treatment-related reduction of Tregs. Additive efficacy effects were observed upon co-treatment with intratumoral 17D and systemic anti-CD137 and anti-PD-1 immunostimulatory monoclonal antibodies. Importantly, when mice were preimmunized with 17D, intratumoral 17D treatment achieved better local and distant antitumor immunity. Such beneficial effects of prevaccination are in part explained by the potentiation of CD4 and CD8 T-cell infiltration in the treated tumor. The repurposed use of a GMP-grade vaccine to be given via the intratumoral route in prevaccinated patients constitutes a clinically feasible and safe immunotherapy approach.
Collapse
Affiliation(s)
- Maria Angela Aznar
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- Present address:
Center for Cellular ImmunotherapiesPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Carmen Molina
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
| | - Alvaro Teijeira
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- CIBERONCMadridSpain
- Instituto de investigación de Navarra (IDISNA)PamplonaSpain
| | - Inmaculada Rodriguez
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- CIBERONCMadridSpain
- Instituto de investigación de Navarra (IDISNA)PamplonaSpain
| | - Arantza Azpilikueta
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- Instituto de investigación de Navarra (IDISNA)PamplonaSpain
| | - Saray Garasa
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- Instituto de investigación de Navarra (IDISNA)PamplonaSpain
| | - Alfonso R Sanchez‐Paulete
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- Present address:
Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Luna Cordeiro
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- Instituto de investigación de Navarra (IDISNA)PamplonaSpain
| | - Iñaki Etxeberria
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
| | - Maite Alvarez
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
| | - Sergio Rius‐Rocabert
- Microbiology SectionDpto. CC, Farmaceuticas y de la SaludFacultad de FarmaciaUniversidad CEU San PabloCEU UniversityBoadilla del Monte, MadridSpain
- Instituto de Medicina Molecular Aplicada (IMMA)Universidad CEU San Pablo, Pablo‐CEU, CEU UniversitiesBoadilla del Monte, MadridSpain
| | - Estanislao Nistal‐Villan
- Microbiology SectionDpto. CC, Farmaceuticas y de la SaludFacultad de FarmaciaUniversidad CEU San PabloCEU UniversityBoadilla del Monte, MadridSpain
- Instituto de Medicina Molecular Aplicada (IMMA)Universidad CEU San Pablo, Pablo‐CEU, CEU UniversitiesBoadilla del Monte, MadridSpain
| | - Pedro Berraondo
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- CIBERONCMadridSpain
- Instituto de investigación de Navarra (IDISNA)PamplonaSpain
| | - Ignacio Melero
- Center for Applied Medical Research (CIMA)University of NavarraPamplonaSpain
- CIBERONCMadridSpain
- Instituto de investigación de Navarra (IDISNA)PamplonaSpain
| |
Collapse
|
135
|
Ye J, Mills BN, Zhao T, Han BJ, Murphy JD, Patel AP, Johnston CJ, Lord EM, Belt BA, Linehan DC, Gerber SA. Assessing the Magnitude of Immunogenic Cell Death Following Chemotherapy and Irradiation Reveals a New Strategy to Treat Pancreatic Cancer. Cancer Immunol Res 2020; 8:94-107. [PMID: 31719057 PMCID: PMC6946873 DOI: 10.1158/2326-6066.cir-19-0373] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/18/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to have a dismal prognosis, in part, due to ineffective treatment strategies. The efficacy of some chemotherapies and especially radiotherapy is mediated partially by the immune system. Therefore, we hypothesized that profiling the immune response following chemotherapy and/or irradiation can be used as a readout for treatment efficacy but also to help identify optimal therapeutic schedules for PDAC. Using murine models of PDAC, we demonstrated that concurrent administration of stereotactic body radiotherapy (SBRT) and a modified dose of FOLFIRINOX (mFX) resulted in superior tumor control when compared with single or sequential treatment groups. Importantly, this combined treatment schedule enhanced the magnitude of immunogenic cell death, which in turn amplified tumor antigen presentation by dendritic cells and intratumoral CD8+ T-cell infiltration. Concurrent therapy also resulted in systemic immunity contributing to the control of established metastases. These findings provide a rationale for pursuing concurrent treatment schedules of SBRT with mFX in PDAC.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Tony Zhao
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Booyeon J Han
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Joseph D Murphy
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Ankit P Patel
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Carl J Johnston
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Edith M Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - David C Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York.
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
136
|
Yamazaki T, Buqué A, Rybstein M, Chen J, Sato A, Galluzzi L. Methods to Detect Immunogenic Cell Death In Vivo. Methods Mol Biol 2020; 2055:433-452. [PMID: 31502164 DOI: 10.1007/978-1-4939-9773-2_20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In response to selected stressors, cancer cells can undergo a form of regulated cell death that-in immunocompetent syngeneic hosts-is capable of eliciting an adaptive immune response specific for dead cell-associated antigens. Thus, such variant of regulated cell death manifests with robust antigenicity and adjuvanticity. As compared to their normal counterparts, malignant cells are highly antigenic per se, implying that they express a variety of antigens that are not covered by central tolerance. However, the precise modality through which cancer cells die in response to stress has a major influence on adjuvanticity. Moreover, the adjuvanticity threshold to productively drive anticancer immune responses is considerably lower in tumor-naïve hosts as compared to their tumor-bearing counterparts, largely reflecting the establishment of peripheral tolerance to malignant lesions in the latter (but not in the former). So far, no cellular biomarker or combination thereof has been found to reliably predict the ability of cancer cell death to initiate antitumor immunity. Thus, although some surrogate biomarkers of adjuvanticity can be used for screening purposes, the occurrence of bona fide immunogenic cell death (ICD) can only be ascertained in vivo. Here, we describe two methods that can be harnessed to straightforwardly determine the immunogenicity of mouse cancer cells succumbing to stress in both tumor-naïve and tumor-bearing hosts.
Collapse
Affiliation(s)
- Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Marissa Rybstein
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Jonathan Chen
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA. .,Université Paris Descartes/Paris V, Paris, France.
| |
Collapse
|
137
|
Sveen A, Kopetz S, Lothe RA. Biomarker-guided therapy for colorectal cancer: strength in complexity. Nat Rev Clin Oncol 2020; 17:11-32. [PMID: 31289352 PMCID: PMC7577509 DOI: 10.1038/s41571-019-0241-1] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2019] [Indexed: 12/16/2022]
Abstract
The number of molecularly stratified treatment options available to patients with colorectal cancer (CRC) is increasing, with a parallel rise in the use of biomarkers to guide prognostication and treatment decision-making. The increase in both the number of biomarkers and their use has resulted in a progressively complex situation, evident both from the extensive interactions between biomarkers and from their sometimes complex associations with patient prognosis and treatment benefit. Current and emerging biomarkers also reflect the genomic complexity of CRC, and include a wide range of aberrations such as point mutations, amplifications, fusions and hypermutator phenotypes, in addition to global gene expression subtypes. In this Review, we provide an overview of current and emerging clinically relevant biomarkers and their role in the management of patients with CRC, illustrating the intricacies of biomarker interactions and the growing treatment opportunities created by the availability of comprehensive molecular profiling.
Collapse
Affiliation(s)
- Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research & K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research & K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
138
|
Baxevanis CN, Fortis SP, Perez SA. The balance between breast cancer and the immune system: Challenges for prognosis and clinical benefit from immunotherapies. Semin Cancer Biol 2019; 72:76-89. [PMID: 31881337 DOI: 10.1016/j.semcancer.2019.12.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Cancer evolution is a complex process influenced by genetic factors and extracellular stimuli that trigger signaling pathways to coordinate the continuous and dynamic interaction between tumor cells and the elements of the immune system. For over 20 years now, the immune mechanisms controlling cancer progression have been the focus of intensive research. It is well established that the immune system conveys protective antitumor immunity by destroying immunogenic tumor variants, but also facilitates tumor progression by shaping tumor immunogenicity in a process called "immunoediting". It is also clear that immune-guided tumor editing is associated with tumor evasion from immune surveillance and therefore reinforcing the endogenous antitumor immunity is a desired goal in the context of cancer therapies. The tumor microenvironment (TME) is a complex network which consists of various cell types and factors having important roles regarding tumor development and progression. Tumor infiltrating lymphocytes (TILs) and other tumor infiltrating immune cells (TIICs) are key to our understanding of tumor immune surveillance based on tumor immunogenicity, whereby the densities and location of TILs and TIICs in the tumor regions, as well as their functional programs (comprising the "immunoscore") have a prominent role for prognosis and prediction for several cancers. The presence of tertiary lymphoid structures (TLS) in the TME or in peritumoral areas has an influence on the locally produced antitumor immune response, and therefore also has a significant prognostic impact. The cross-talk between elements of the immune system with tumor cells in the TME is greatly influenced by hypoxia, the gut and/or the local microbiota, and several metabolic elements, which, in a dynamic interplay, have a crucial role for tumor cell heterogeneity and reprogramming of immune cells along their activation and differentiation pathways. Taking into consideration the recent clinical success with the application immunotherapies for the treatment of several cancer types, increasing endeavors have been made to gain better insights into the mechanisms underlying phenotypic and metabolic profiles in the context of tumor progression and immunotherapy. In this review we will address (i) the role of TILs, TIICs and TLS in breast cancer (BCa); (ii) the different metabolic-based pathways used by immune and breast cancer cells; and (iii) implications for immunotherapy-based strategies in BCa.
Collapse
Affiliation(s)
- Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 11522, Athens, Greece.
| | - Sotirios P Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 11522, Athens, Greece
| | - Sonia A Perez
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 11522, Athens, Greece
| |
Collapse
|
139
|
Ajina R, Zahavi DJ, Zhang YW, Weiner LM. Overcoming malignant cell-based mechanisms of resistance to immune checkpoint blockade antibodies. Semin Cancer Biol 2019; 65:28-37. [PMID: 31866479 DOI: 10.1016/j.semcancer.2019.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/09/2019] [Accepted: 12/14/2019] [Indexed: 12/12/2022]
Abstract
Traditional cancer treatment approaches have focused on surgery, radiation therapy, and cytotoxic chemotherapy. However, with rare exceptions, metastatic cancers were considered to be incurable by traditional therapy. Over the past 20 years a fourth modality - immunotherapy - has emerged as a potentially curative approach for patients with advanced metastatic cancer. However, in many patients cancer "finds a way" to evade the anti-tumor effects of immunotherapy. Immunotherapy resistance mechanisms can be employed by both cancer cells and the non-cancer elements of tumor microenvironment. This review focuses on the resistance mechanisms that are specifically mediated by cancer cells. In order to extend the impact of immunotherapy to more patients and across all cancer types, and to inhibit the development of acquired resistance, the underlying biology driving immune escape needs to be better understood. Elucidating mechanisms of immune escape may shed light on new therapeutic targets, and lead to successful combination therapeutic strategies.
Collapse
Affiliation(s)
- Reham Ajina
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, United States
| | - David J Zahavi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, United States
| | - Yong-Wei Zhang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, United States
| | - Louis M Weiner
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, United States.
| |
Collapse
|
140
|
Du B, Waxman DJ. Medium dose intermittent cyclophosphamide induces immunogenic cell death and cancer cell autonomous type I interferon production in glioma models. Cancer Lett 2019; 470:170-180. [PMID: 31765733 DOI: 10.1016/j.canlet.2019.11.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/05/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023]
Abstract
Cyclophosphamide treatment on a medium-dose, intermittent chemotherapy (MEDIC) schedule activates both innate and adaptive immunity leading to major regression of implanted gliomas. Here, we show that this MEDIC treatment regimen induces tumor cell autonomous type-I interferon signaling, followed by release of soluble factors that activate interferon-stimulated genes in both tumor cells and tumor-infiltrating immune cells. In cultured GL261 and CT-2A glioma cells, activated cyclophosphamide stimulated production and release of type-I interferons, leading to robust activation of downstream gene targets. Antibody against the type-I interferon receptor IFNAR1 blocked the cyclophosphamide-stimulated induction of these genes in both cultured glioma cells and implanted gliomas. Furthermore, IFNAR1 antibody strongly inhibited the MEDIC cyclophosphamide-stimulated increases in tumor cell infiltration of macrophages, dendritic cells, B-cells, as well as natural killer cells and cytotoxic T-cells and their cytotoxic effectors. Finally, cyclophosphamide-treated dying glioma cells producing type-I interferons were an effective vaccine against drug-naïve glioma cells implanted in vivo. Thus, cyclophosphamide induces local, tumor cell-centric increases in type-I interferon signaling, which activates immunogenic cell death and is essential for the striking antitumor immune responses that MEDIC cyclophosphamide treatment elicits in these glioma models.
Collapse
Affiliation(s)
- Bin Du
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, 02215, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
141
|
Sprooten J, Garg AD. Type I interferons and endoplasmic reticulum stress in health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:63-118. [PMID: 32138904 PMCID: PMC7104985 DOI: 10.1016/bs.ircmb.2019.10.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFNs) comprise of pro-inflammatory cytokines created, as well as sensed, by all nucleated cells with the main objective of blocking pathogens-driven infections. Owing to this broad range of influence, type I IFNs also exhibit critical functions in many sterile inflammatory diseases and immunopathologies, especially those associated with endoplasmic reticulum (ER) stress-driven signaling pathways. Indeed, over the years accumulating evidence has indicated that the presence of ER stress can influence the production, or sensing of, type I IFNs induced by perturbations like pattern recognition receptor (PRR) agonists, infections (bacterial, viral or parasitic) or autoimmunity. In this article we discuss the link between type I IFNs and ER stress in various diseased contexts. We describe how ER stress regulates type I IFNs production or sensing, or how type I IFNs may induce ER stress, in various circumstances like microbial infections, autoimmunity, diabetes, cancer and other ER stress-related contexts.
Collapse
Affiliation(s)
- Jenny Sprooten
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
142
|
Han P, Hanlon D, Sobolev O, Chaudhury R, Edelson RL. Ex vivo dendritic cell generation-A critical comparison of current approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:251-307. [PMID: 31759433 DOI: 10.1016/bs.ircmb.2019.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells, required for the initiation of naïve and memory T cell responses and regulation of adaptive immunity. The discovery of DCs in 1973, which culminated in the Nobel Prize in Physiology or Medicine in 2011 for Ralph Steinman and colleagues, initially focused on the identification of adherent mononuclear cell fractions with uniquely stellate dendritic morphology, followed by key discoveries of their critical immunologic role in initiating and maintaining antigen-specific immunity and tolerance. The medical promise of marshaling these key capabilities of DCs for therapeutic modulation of antigen-specific immune responses has guided decades of research in hopes to achieve genuine physiologic partnership with the immune system. The potential uses of DCs in immunotherapeutic applications include cancer, infectious diseases, and autoimmune disorders; thus, methods for rapid and reliable large-scale production of DCs have been of great academic and clinical interest. However, difficulties in obtaining DCs from lymphoid and peripheral tissues, low numbers and poor survival in culture, have led to advancements in ex vivo production of DCs, both for probing molecular details of DC function as well as for experimenting with their clinical utility. Here, we review the development of a diverse array of DC production methodologies, ranging from cytokine-based strategies to genetic engineering tools devised for enhancing DC-specific immunologic functions. Further, we explore the current state of DC therapies in clinic, as well as emerging insights into physiologic production of DCs inspired by existing therapies.
Collapse
Affiliation(s)
- Patrick Han
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Douglas Hanlon
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Olga Sobolev
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Rabib Chaudhury
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Richard L Edelson
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States.
| |
Collapse
|
143
|
Nath S, Obaid G, Hasan T. The Course of Immune Stimulation by Photodynamic Therapy: Bridging Fundamentals of Photochemically Induced Immunogenic Cell Death to the Enrichment of T-Cell Repertoire. Photochem Photobiol 2019; 95:1288-1305. [PMID: 31602649 PMCID: PMC6878142 DOI: 10.1111/php.13173] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
Photodynamic therapy (PDT) is a potentially immunogenic and FDA-approved antitumor treatment modality that utilizes the spatiotemporal combination of a photosensitizer, light and oftentimes oxygen, to generate therapeutic cytotoxic molecules. Certain photosensitizers under specific conditions, including ones in clinical practice, have been shown to elicit an immune response following photoillumination. When localized within tumor tissue, photogenerated cytotoxic molecules can lead to immunogenic cell death (ICD) of tumor cells, which release damage-associated molecular patterns and tumor-specific antigens. Subsequently, the T-lymphocyte (T cell)-mediated adaptive immune system can become activated. Activated T cells then disseminate into systemic circulation and can eliminate primary and metastatic tumors. In this review, we will detail the multistage cascade of events following PDT of solid tumors that ultimately lead to the activation of an antitumor immune response. More specifically, we connect the fundamentals of photochemically induced ICD with a proposition on potential mechanisms for PDT enhancement of the adaptive antitumor response. We postulate a hypothesis that during the course of the immune stimulation process, PDT also enriches the T-cell repertoire with tumor-reactive activated T cells, diversifying their tumor-specific targets and eliciting a more expansive and rigorous antitumor response. The implications of such a process are likely to impact the outcomes of rational combinations with immune checkpoint blockade, warranting investigations into T-cell diversity as a previously understudied and potentially transformative paradigm in antitumor photodynamic immunotherapy.
Collapse
Affiliation(s)
- Shubhankar Nath
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Girgis Obaid
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
144
|
Zhang Y, Yang S, Yang Y, Liu T. Resveratrol induces immunogenic cell death of human and murine ovarian carcinoma cells. Infect Agent Cancer 2019; 14:27. [PMID: 31636696 PMCID: PMC6798484 DOI: 10.1186/s13027-019-0247-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose This study aimed to clarify whether immunogenic cell death (ICD) contributed to the anti-tumor action of resveratrol against ovarian carcinoma. Methods Resveratrol suppressed cell proliferation and induced apoptosis in ovarian carcinoma cells. In addition, resveratrol treatment stimulated cell surface exposure of calreticulin, HMGB1 secretion and ATP release. Results Vaccination with resveratrol-pretreated ID8 cells significantly inhibited growth of subsequent inoculated xenograft tumor. Direct administration with resveratrol suppressed tumor progression accompanied with compromised cell proliferation and enhanced cell apoptosis. We further characterized increases of both mature dendritic cells and cytotoxic T cells in xenograft tumor in response to resveratrol treatment, which also inhibited TGF-β production and stimulated both IL12p7 and IFN-γ secretion. Most importantly, we demonstrated that combination with PD-1 antibody greatly inhibited tumor growth, while depletion of CD8+ T cells by neutralizing antibody restored xenograft progression. Conclusion Our data suggested resveratrol exerted anti-tumor action against ovarian cancer via both apoptosis and ICD pathways.
Collapse
Affiliation(s)
- Yanke Zhang
- The Affiliated Hospital of Medical School of Ningbo University, No.247 Renmin Road, Jiangbei District, Ningbo, 315020 Zhejiang China
| | - Sufen Yang
- The Affiliated Hospital of Medical School of Ningbo University, No.247 Renmin Road, Jiangbei District, Ningbo, 315020 Zhejiang China
| | - Yang Yang
- The Affiliated Hospital of Medical School of Ningbo University, No.247 Renmin Road, Jiangbei District, Ningbo, 315020 Zhejiang China
| | - Teng Liu
- The Affiliated Hospital of Medical School of Ningbo University, No.247 Renmin Road, Jiangbei District, Ningbo, 315020 Zhejiang China
| |
Collapse
|
145
|
Lapenta C, Donati S, Spadaro F, Lattanzi L, Urbani F, Macchia I, Sestili P, Spada M, Cox MC, Belardelli F, Santini SM. Lenalidomide improves the therapeutic effect of an interferon-α-dendritic cell-based lymphoma vaccine. Cancer Immunol Immunother 2019; 68:1791-1804. [PMID: 31620858 DOI: 10.1007/s00262-019-02411-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/05/2019] [Indexed: 12/25/2022]
Abstract
The perspective of combining cancer vaccines with immunomodulatory drugs is currently regarded as a highly promising approach for boosting tumor-specific T cell immunity and eradicating residual malignant cells. The efficacy of dendritic cell (DC) vaccination in combination with lenalidomide, an anticancer drug effective in several hematologic malignancies, was investigated in a follicular lymphoma (FL) model. First, we evaluated the in vitro activity of lenalidomide in modulating the immune responses of lymphocytes co-cultured with a new DC subset differentiated with IFN-α (IFN-DC) and loaded with apoptotic lymphoma cells. We next evaluated the efficacy of lenalidomide and IFN-DC-based vaccination, either alone or in combination, in hu-PBL-NOD/SCID mice bearing established human lymphoma. We found that lenalidomide reduced Treg frequency and IL-10 production in vitro, improved the formation of immune synapses of CD8 + lymphocytes with lymphoma cells and enhanced anti-lymphoma cytotoxicity. Treatment of lymphoma-bearing mice with either IFN-DC vaccination or lenalidomide led to a significant decrease in tumor growth and lymphoma cell spread. Lenalidomide treatment was shown to substantially inhibit tumor-induced neo-angiogenesis rather than to exert a direct cytotoxic effect on lymphoma cells. Notably, the combined treatment with the vaccine plus lenalidomide was more effective than either single treatment, resulting in the significant regression of established tumors and delayed tumor regrowth upon treatment discontinuation. In conclusion, our data demonstrate that IFN-DC-based vaccination plus lenalidomide exert an additive therapeutic effect in xenochimeric mice bearing established lymphoma. These results may pave the way to evaluate this combination in the clinical ground.
Collapse
Affiliation(s)
- Caterina Lapenta
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Simona Donati
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Francesca Spadaro
- Servizio Grandi Strumentazioni e Core Facilities, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Laura Lattanzi
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Francesca Urbani
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.,Scuola di Dottorato in Biotecnologie Mediche e Medicina Traslazionale, Tor Vergata University, 00133, Rome, Italy
| | - Iole Macchia
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paola Sestili
- Servizio Grandi Strumentazioni e Core Facilities, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Massimo Spada
- Centro nazionale sperimentazione e benessere animale, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Maria Christina Cox
- Unità di Ematologia, Azienda Ospedaliera Sant'Andrea, Università La Sapienza, 00189, Rome, Italy
| | - Filippo Belardelli
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), 00133, Rome, Italy
| | - Stefano M Santini
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
146
|
Abstract
New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.
Collapse
|
147
|
Louttit C, Park KS, Moon JJ. Bioinspired nucleic acid structures for immune modulation. Biomaterials 2019; 217:119287. [PMID: 31247511 PMCID: PMC6635102 DOI: 10.1016/j.biomaterials.2019.119287] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 12/27/2022]
Abstract
Nucleic acids have both extensive physiological function and structural potential, rendering them quintessential engineering biomaterials. As carriers of precisely-tunable genetic information, both DNA and RNA can be synthetically generated to form a myriad of structures and to transmit specific genetic code. Importantly, recent studies have shown that DNA and RNA, both in their native and engineered forms, can function as potent regulators of innate immunity, capable of initiating and modulating immune responses. In this review, we highlight recent advances in biomaterials inspired by the various interactions of nucleic acids and the immune system. We discuss key advances in self-assembled structures based on exogenous nucleic acids and engineering approaches to apply endogenous nucleic acids as found in immunogenic cell death and extracellular traps. In addition, we discuss new strategies to control dinucleotide signaling and provide recent examples of biomaterials designed for cancer immunotherapy with STING agonists.
Collapse
Affiliation(s)
- Cameron Louttit
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kyung Soo Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - James J Moon
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
148
|
Anderson R, Theron AJ, Rapoport BL. Immunopathogenesis of Immune Checkpoint Inhibitor-Related Adverse Events: Roles of the Intestinal Microbiome and Th17 Cells. Front Immunol 2019; 10:2254. [PMID: 31616428 PMCID: PMC6775220 DOI: 10.3389/fimmu.2019.02254] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/06/2019] [Indexed: 12/18/2022] Open
Abstract
The advent of novel, innovative, and effective anti-cancer immunotherapies has engendered an era of renewed optimism among cancer specialists and their patients. Foremost among these successful immunotherapies are monoclonal antibodies (MAbs) which target immune checkpoint inhibitor (ICI) molecules, most prominently cytotoxic T-lymphocyte-associated protein (CTLA-4) and programmed cell death protein-1 (PD-1) and its major ligand, PD-L1. These immunotherapeutic agents are, however, often associated with the occurrence of immune-mediated toxicities known as immune-related adverse events (IRAEs). The incidence of severe toxicities increases substantially when these agents are used together, particularly with CTLA-4 in combination with PD-1 or PD-L1 antagonists. Accordingly, dissociating the beneficial anti-tumor therapeutic activity of these agents from the emergence of IRAEs represents a significant challenge to attaining the optimum efficacy of ICI-targeted immunotherapy of cancer. This situation is compounded by an increasing awareness, possibly unsurprising, that both the beneficial and harmful effects of ICI-targeted therapies appear to result from an over-reactive immune system. Nevertheless, this challenge may not be insurmountable. This contention is based on acquisition of recent insights into the role of the gut microbiome and its products as determinants of the efficacy of ICI-targeted immunotherapy, as well as an increasing realization of the enigmatic involvement of Th17 cells in both anti-tumor activity and the pathogenesis of some types of IRAEs. Evidence linking the beneficial and harmful activities of ICI-targeted immunotherapy, recent mechanistic insights focusing on the gut microbiome and Th17 cells, as well as strategies to attenuate IRAEs in the setting of retention of therapeutic activity, therefore represent the major thrusts of this review.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Annette J Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Bernardo L Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
149
|
García-Foncillas J, Sunakawa Y, Aderka D, Wainberg Z, Ronga P, Witzler P, Stintzing S. Distinguishing Features of Cetuximab and Panitumumab in Colorectal Cancer and Other Solid Tumors. Front Oncol 2019; 9:849. [PMID: 31616627 PMCID: PMC6763619 DOI: 10.3389/fonc.2019.00849] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/19/2019] [Indexed: 12/27/2022] Open
Abstract
Cetuximab and panitumumab are two distinct monoclonal antibodies (mAbs) targeting the epidermal growth factor receptor (EGFR), and both are widely used in combination with chemotherapy or as monotherapy to treat patients with RAS wild-type metastatic colorectal cancer. Although often considered interchangeable, the two antibodies have different molecular structures and can behave differently in clinically relevant ways. More specifically, as an immunoglobulin (Ig) G1 isotype mAb, cetuximab can elicit immune functions such as antibody-dependent cell-mediated cytotoxicity involving natural killer cells, T-cell recruitment to the tumor, and T-cell priming via dendritic cell maturation. Panitumumab, an IgG2 isotype mAb, does not possess these immune functions. Furthermore, the two antibodies have different binding sites on the EGFR, as evidenced by mutations on the extracellular domain that can confer resistance to one of the two therapeutics or to both. We consider a comparison of the properties of these two antibodies to represent a gap in the literature. We therefore compiled a detailed, evidence-based educational review of the known molecular, clinical, and functional differences between the two antibodies and concluded that they are distinct therapeutic agents that should be considered individually during treatment planning. Available data for one agent can only partly be extrapolated to the other. Looking to the future, the known immune activity of cetuximab may provide a rationale for this antibody as a combination partner with investigational chemotherapy plus immunotherapy regimens for colorectal cancer.
Collapse
Affiliation(s)
- Jesús García-Foncillas
- Cancer Institute, University Hospital Fundacion Jimenez Diaz, Autonomous University of Madrid, Madrid, Spain
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Dan Aderka
- Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Zev Wainberg
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | | | | | - Sebastian Stintzing
- Department of Hematology, Oncology, and Tumor Immunology (CCM) Charité Universitaetsmedizin, Berlin, Germany
| |
Collapse
|
150
|
Lévesque S, Le Naour J, Pietrocola F, Paillet J, Kremer M, Castoldi F, Baracco EE, Wang Y, Vacchelli E, Stoll G, Jolly A, De La Grange P, Zitvogel L, Kroemer G, Pol JG. A synergistic triad of chemotherapy, immune checkpoint inhibitors, and caloric restriction mimetics eradicates tumors in mice. Oncoimmunology 2019; 8:e1657375. [PMID: 31646107 PMCID: PMC6791453 DOI: 10.1080/2162402x.2019.1657375] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 01/16/2023] Open
Abstract
We have recently shown that chemotherapy with immunogenic cell death (ICD)-inducing agents can be advantageously combined with fasting regimens or caloric restriction mimetics (CRMs) to achieve superior tumor growth control via a T cell-dependent mechanism. Here, we show that the blockade of the CD11b-dependent extravasation of myeloid cells blocks such a combination effect as well. Based on the characterization of the myeloid and lymphoid immune infiltrates, including the expression pattern of immune checkpoint proteins (and noting a chemotherapy-induced overexpression of programmed death-ligand 1, PD-L1, on both cancer cells and leukocytes, as well as a reduced frequency of exhausted CD8+ T cells positive for programmed cell death 1 protein, PD-1), we then evaluated the possibility to combine ICD inducers, CRMs and targeting of the PD-1/PD-L1 interaction. While fasting or CRMs failed to improve tumor growth control by PD-1 blockade, ICD inducers alone achieved a partial sensitization to treatment with a PD-1-specific antibody. However, definitive cure of most of the tumor-bearing mice was only achieved by a tritherapy combining (i) ICD inducers exemplified by mitoxantrone and oxaliplatin, (ii) CRMs exemplified by hydroxycitrate and spermidine and substitutable for by fasting, and (iii) immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 interaction. Altogether, these results point to the possibility of synergistic interactions among distinct classes of anticancer agents.
Collapse
Affiliation(s)
- Sarah Lévesque
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
- Université Paris-Sud/Paris XI, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Julie Le Naour
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
- Université Paris-Sud/Paris XI, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Federico Pietrocola
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
| | - Juliette Paillet
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
- Université Paris-Sud/Paris XI, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Margerie Kremer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
| | - Francesca Castoldi
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
- Université Paris-Sud/Paris XI, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Elisa E. Baracco
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
- Université Paris-Sud/Paris XI, Faculté de Médecine, Kremlin-Bicêtre, France
| | - Yan Wang
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
| | - Erika Vacchelli
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
| | - Gautier Stoll
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
| | | | | | - Laurence Zitvogel
- Université Paris-Sud/Paris XI, Faculté de Médecine, Kremlin-Bicêtre, France
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Jonathan G. Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Sorbonne Université, Paris, France
| |
Collapse
|