1
|
Li C, Liu Y, Mao H, Yang W, Liu S, Shan Y. Oncosis is the predominant type of cell death in rhabdomyolysis following exertional heat stroke. PLoS One 2025; 20:e0308586. [PMID: 40131898 PMCID: PMC11936275 DOI: 10.1371/journal.pone.0308586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/28/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Rhabdomyolysis (RM), particularly heat exhaustion-associated rhabdomyolysis (ehsRM), is a significant clinical issue associated with high mortality and healthcare costs. However, the cellular death mechanisms remain incompletely understood. Oncosis, a form of passive cell death distinct from apoptosis, is characterized by cell swelling and triggered by ATP depletion. Additionally, porimin, a specific biomarker, can uniquely identify oncosis. This study aims to investigate the role and mechanisms of oncosis in both in vitro and in vivo models of ehsRM. OBJECTIVE This study aims to investigate the role and mechanisms of oncosis in both in vitro and in vivo models of ehsRM. METHODS In the in vitro study, 6-8-week-old male rats were subjected to treadmill exercise at an ambient temperature of (39.5 ± 0.5)°C and relative humidity of 50%-60%, at a speed of 15 meters per minute until their core body temperature (Tc) reached 43.0°C to establish a heatstroke animal model. Skeletal muscle and blood samples from the gastrocnemius were collected for cytokine, biochemical, and histopathological analyses. Pathological findings revealed decreased muscle fiber density, structural disarray, swelling, degeneration, and hemorrhage. Ultrastructural analysis showed cell swelling, structural disarray, cytoplasmic vacuolation, mitochondrial swelling and degeneration, loss of cristae, and nuclear degeneration, indicating myocyte swelling and necrosis. Porimin, CytC, Bax, and caspase-1 expression increased, while Bcl-2 expression decreased. JC-1 staining indicated a decline in mitochondrial membrane potential and dysfunction. ATP levels decreased, and reactive oxygen species (ROS) production increased. In the in vivo study, HSKMC cells were subjected to 4 hours of heat shock at 43°C to establish a heatstroke-induced rhabdomyolysis cell model. Electron microscopy revealed cell swelling, cytoplasmic vacuolation, mitochondrial swelling and degeneration, and nuclear swelling; late-stage (necrotic-like death) was characterized by nucleolar dissolution, nuclear fragmentation, chromatin condensation, and collapse of cytoplasmic structures. After 24 hours post-modeling, the proportion of double-positive cells (porimin + /PI+) and ROS levels significantly increased, as did porimin expression, while mitochondrial membrane potential and ATP levels significantly decreased. The proportion of Annexin V + /PI + double-positive cells and caspase-3 levels showed no significant changes. RESULTS In both in vitro and in vivo studies, oncosis played a crucial role in ehsRM. Pathological and ultrastructural analyses demonstrated cell swelling, structural disarray, mitochondrial damage, and nuclear degeneration. Porimin, CytC, Bax, and caspase-1 expression increased, while Bcl-2 expression decreased. ATP levels decreased, and ROS production increased. In the in vivo study, the proportion of porimin + /PI + double-positive cells and ROS levels significantly increased, while mitochondrial membrane potential and ATP levels significantly decreased. The proportion of Annexin V + /PI + double-positive cells and caspase-3 levels showed no significant changes. CONCLUSION Oncosis is predominant in ehsRM, involving mitochondrial dysfunction, ATP depletion, and oxidative stress.
Collapse
Affiliation(s)
- Chengcheng Li
- Department of Emergency Medicine, The Sixth Medical Center of PLA General Hospital of Beijing,
- Department of Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University,
| | - Yang Liu
- Department of Emergency Medicine, The Sixth Medical Center of PLA General Hospital of Beijing,
- School of Medicine, South China University of Technology
| | - Handing Mao
- Department of Emergency Medicine, The Sixth Medical Center of PLA General Hospital of Beijing,
| | - Wenjun Yang
- Department of Emergency Medicine, The Sixth Medical Center of PLA General Hospital of Beijing,
| | - Shuyuan Liu
- Department of Emergency Medicine, The Sixth Medical Center of PLA General Hospital of Beijing,
| | - Yi Shan
- Department of Emergency Medicine, The Sixth Medical Center of PLA General Hospital of Beijing,
| |
Collapse
|
2
|
刘 洋, 贾 亿, 李 程, 毛 汉, 刘 树, 单 毅. [Dexmedetomidine attenuates heat stress-induced oncosis in human skeletal muscle cells by activating the Nrf2/Ho-1 pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:603-613. [PMID: 40159975 PMCID: PMC11955895 DOI: 10.12122/j.issn.1673-4254.2025.03.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Indexed: 04/02/2025]
Abstract
OBJECTIVES To investigate the protective effects of dexmedetomidine (DEX) against heat stress (HS)-induced oncosis in human skeletal muscle cells (HSKMCs) and its underlying mechanisms. METHODS A HSKMC model of HS-induced oncosis were established by 43 ℃ water bath for 4 h, and the effects of treatments with 30 μmol/L DEX, ML385 (a Nrf2 inhibitor) +DEX, si-Nrf2+HS, and si-Nrf2+DEX prior to modeling on cell viability was assessed using CCK-8 assay. Oncosis characteristics were evaluated using transmission electron microscopy and Annexin V-FITC/PI flow cytometry. The oxidative stress markers (GSH, GSH-Px, MDA, SOD and ROS), mitochondrial membrane potential, energy metabolism, and inflammatory cytokines (TNF-α, IL-6 and IL-1β) in the cells were quantified using standard kits, and the expressions of porimin, caspase-3 and Nrf2 pathway proteins were analyzed using Western blotting and qRT-PCR. RESULTS HS induced typical oncotic features in HSKMCs including organelle swelling and cytoplasmic vacuolization. DEX pretreatment significantly attenuated these changes, reduced Annexin V+/PI+ cell ratio and cellular porimin expression, and lowered the levels of ROS and MDA while restoring GSH and SOD levels. DEX pretreatment also significantly increased the mitochondrial membrane potential and ATP level, upregulated the expressions of Nrf2, p-Nrf2, HO-1 and NQO1, and suppressed the expressions of TNF-α, IL-6 and IL-1β. The protective effects of DEX were obviously attenuated by interventions with ML385 or si-Nrf2. CONCLUSIONS DEX mitigates HS-induced HSKMC oncosis by activating the Nrf2/HO-1 pathway to relieve oxidative stress, mitochondrial dysfunction, and inflammatory responses.
Collapse
Affiliation(s)
| | | | | | | | | | - 毅 单
- 单 毅,教授,副主任医师,硕士生导师,E-mail:
| |
Collapse
|
3
|
Guo J, Yang WT, Mai FY, Liang JR, Luo J, Zhou MC, Yu DD, Wang YL, Li CG. Unravelling oncosis: morphological and molecular insights into a unique cell death pathway. Front Immunol 2024; 15:1450998. [PMID: 39281670 PMCID: PMC11393741 DOI: 10.3389/fimmu.2024.1450998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 09/18/2024] Open
Abstract
Programmed cell death (PCD) is a fundamental biological process for maintaining cellular equilibrium and regulating development, health, and disease across all living organisms. Among the various types of PCD, apoptosis plays a pivotal role in numerous diseases, notably cancer. Cancer cells frequently develop mechanisms to evade apoptosis, increasing resistance to standard chemotherapy treatments. This resistance has prompted extensive research into alternative mechanisms of programmed cell death. One such pathway is oncosis, characterized by significant energy consumption, cell swelling, dilation of the endoplasmic reticulum, mitochondrial swelling, and nuclear chromatin aggregation. Recent research suggests that oncosis can impact conditions such as chemotherapeutic cardiotoxicity, myocardial ischemic injury, stroke, and cancer, mediated by specific oncosis-related proteins. In this review, we provide a detailed examination of the morphological and molecular features of oncosis and discuss various natural or small molecule compounds that can induce this type of cell death. Additionally, we summarize the current understanding of the molecular mechanisms underlying oncosis and its role in both normal physiology and pathological conditions. These insights aim to illuminate future research directions and propose innovative strategies for leveraging oncosis as a therapeutic tool against human diseases and cancer resistance.
Collapse
Affiliation(s)
- Jie Guo
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Wen-Tao Yang
- Pain Department of Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Feng-Yi Mai
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, China
| | - Jing-Rong Liang
- Pain Department of Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Jiao Luo
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, China
| | - Ming-Chao Zhou
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, China
| | - Dong-Dong Yu
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yu-Long Wang
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Chen-Guang Li
- Pain Department of Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| |
Collapse
|
4
|
Li F, Jia Y, Fang J, Gong L, Zhang Y, Wei S, Wu L, Jiang P. Neuroprotective Mechanism of MOTS-c in TBI Mice: Insights from Integrated Transcriptomic and Metabolomic Analyses. Drug Des Devel Ther 2024; 18:2971-2987. [PMID: 39050800 PMCID: PMC11268520 DOI: 10.2147/dddt.s460265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Background Traumatic brain injury (TBI) is a condition characterized by structural and physiological disruptions in brain function caused by external forces. However, as the highly complex and heterogenous nature of TBI, effective treatments are currently lacking. Mitochondrial open reading frame of the 12S rRNA-c (MOTS-c) has shown notable antinociceptive and anti-inflammatory effects, yet its detailed neuroprotective effects and mode of action remain incompletely understood. This study investigated the neuroprotective effects and the underlying mechanisms of MOTS-c. Methods Adult male C57BL/6 mice were randomly divided into three groups: control (CON) group, MOTS-c group and TBI group. Enzyme-linked immunosorbent assay (ELISA) kit method was used to measure the expression levels of MOTS-c in different groups. Behavioral tests were conducted to assess the effects of MOTS-c. Then, transcriptomics and metabolomics were performed to search Differentially Expressed Genes (DEGs) and Differentially Expressed Metabolites (DEMs), respectively. Moreover, the integrated transcriptomics and metabolomics analysis were employed using R packages and online Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Results ELISA kit method showed that TBI resulted in a decrease in the expression of MOTS-c. and peripheral administration of MOTS-c could enter the brain tissue after TBI. Behavioral tests revealed that MOTS-c improved memory, learning, and motor function impairments in TBI mice. Additionally, transcriptomic analysis screened 159 differentially expressed genes. Metabolomic analysis identified 491 metabolites with significant differences. Integrated analysis found 14 KEGG pathways, primarily related to metabolic pathways. Besides, several signaling pathways were enriched, including neuroactive ligand-receptor interaction and retrograde endocannabinoid signaling. Conclusion TBI reduced the expression of MOTS-c. MOTS-c reduced inflammatory responses, molecular damage, and cell death by down-regulating macrophage migration inhibitory factor (MIF) expression and activating the retrograde endocannabinoid signaling pathway. In addition, MOTS-c alleviated the response to hypoxic stress and enhanced lipid β-oxidation to provide energy for the body following TBI. Overall, our study offered new insights into the neuroprotective mechanisms of MOTS-c in TBI mice.
Collapse
Affiliation(s)
- Fengfeng Li
- Neurosurgery Department, Tengzhou Central People’s Hospital Affiliated to Xuzhou Medical University, Tengzhou, Shandong, 277500, People’s Republic of China
| | - Yang Jia
- Neurosurgery Department, Tengzhou Central People’s Hospital Affiliated to Xuzhou Medical University, Tengzhou, Shandong, 277500, People’s Republic of China
| | - Jun Fang
- Anesthesiology Department, Tengzhou Central People’s Hospital Affiliated to Xuzhou Medical University, Tengzhou, Shandong, 277500, People’s Republic of China
| | - Linqiang Gong
- Gastroenterology Department, Tengzhou Central People’s Hospital Affiliated to Xuzhou Medical University, Tengzhou, Shandong, 277500, People’s Republic of China
| | - Yazhou Zhang
- Foot and Ankle Surgery Department, Tengzhou Central People’s Hospital Affiliated to Xuzhou Medical University, Tengzhou, Shandong, 277500, People’s Republic of China
| | - Shanshan Wei
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Jining, Shandong, 272000, People’s Republic of China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People’s Hospital Affiliated to Xuzhou Medical University, Tengzhou, Shandong, 277500, People’s Republic of China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Jining, Shandong, 272000, People’s Republic of China
| |
Collapse
|
5
|
Lin YS, Sun Z, Shen LS, Gong RH, Chen JW, Xu Y, Yu H, Chen S, Chen GQ. Arnicolide D induces endoplasmic reticulum stress-mediated oncosis via ATF4 and CHOP in hepatocellular carcinoma cells. Cell Death Discov 2024; 10:134. [PMID: 38472168 DOI: 10.1038/s41420-024-01911-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
Endoplasmic reticulum (ER) stress can trigger various cell death mechanisms beyond apoptosis, providing promise in cancer treatment. Oncosis, characterized by cellular swelling and increased membrane permeability, represents a non-apoptotic form of cell death. In our study, we discovered that Arnicolide D (AD), a natural sesquiterpene lactone compound, induces ER stress-mediated oncosis in hepatocellular carcinoma (HCC) cells, and this process is reactive oxygen species (ROS)-dependent. Furthermore, we identified the activation of the PERK-eIF2α-ATF4-CHOP pathway during ER stress as a pivotal factor in AD-induced oncosis. Notably, the protein synthesis inhibitor cycloheximide (CHX) was found to effectively reverse AD-induced oncosis, suggesting ATF4 and CHOP may hold crucial roles in the induction of oncosis by AD. These proteins play a vital part in promoting protein synthesis during ER stress, ultimately leading to cell death. Subsequent studies, in where we individually or simultaneously knocked down ATF4 and CHOP in HCC cells, provided further confirmation of their indispensable roles in AD-induced oncosis. Moreover, additional animal experiments not only substantiated AD's ability to inhibit HCC tumor growth but also solidified the essential role of ER stress-mediated and ROS-dependent oncosis in AD's therapeutic potential. In summary, our research findings strongly indicate that AD holds promise as a therapeutic agent for HCC by its ability to induce oncosis.
Collapse
Affiliation(s)
- Yu-Shan Lin
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Zhiwei Sun
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing, 400065, China
| | - Rui-Hong Gong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong, China
| | - Jia-Wen Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 100193, Beijing, China
| | - Yanfeng Xu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Haiyang Yu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, and State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China.
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong, China.
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 100193, Beijing, China.
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong, China.
| | - Guo-Qing Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China.
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong, China.
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong, China.
| |
Collapse
|
6
|
Xue K, Zhao Y, Sun S, Li Y, Qi Z. A near-infrared aggregation-induced emission photosensitizer targeting mitochondria for depleting Cu 2+ to trigger light-activated cancer cells oncosis. Bioorg Chem 2024; 143:107020. [PMID: 38176374 DOI: 10.1016/j.bioorg.2023.107020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024]
Abstract
Abnormally high levels of copper in tumors stimulate malignant proliferation and migration of cancer cells, which proposes a formidable challenge for the thorough therapy of malignant tumors. In this work, we developed a reliable, mitochondria-targeted near-infrared aggregation-induced emission fluorescent probe, TTQ-Th, whose thiourea moiety specifically could recognize mitochondria even both upon loss of mitochondrial membrane potential or in fixated cells, and can capture copper overexpressed by tumor cells, leading to severe copper deficiency. In parallel, TTQ-Th can generate sufficient reactive oxygen species (ROS) upon photoexcitation, while copper deficiency inhibits expression of related copper-based enzymes, resulting in a decline in ATP production. Such energy deficiency, combined with reduced MMP and elevated oxidative stress can lead to critical cell oncosis. Both in vitro and intracellular experiments can illustrate that the elevated ROS has remarkable damage to tumor cells and contributes to the elimination of the primary tumor, while copper deficiency further hinder tumor cell migration and induces G0/G1 cell cycle arrest in a dose-dependent manner, which is an efficacious strategy for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Ke Xue
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Yongfei Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Saidong Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Yuanhang Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Zhengjian Qi
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China.
| |
Collapse
|
7
|
Zhang D, Yuan R, Pan J, Fan Q, Sun K, Xu Z, Gao X, Wang Q, He J, Ye Y, Mu Z, Leng J, Gao H. Dihydrotanshinone Triggers Porimin-Dependent Oncosis by ROS-Mediated Mitochondrial Dysfunction in Non-Small-Cell Lung Cancer. Int J Mol Sci 2023; 24:11953. [PMID: 37569328 PMCID: PMC10419281 DOI: 10.3390/ijms241511953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer death. Non-small-cell lung cancer (NSCLC) accounts for the majority of lung cancer diagnoses. Dihydrotanshinone (DHT) is a compound extract from Salvia miltiorrhiza, which has favorable anti-inflammatory and anti-cancer activities. However, the role of DHT in NSCLC has not been fully studied. The anti-cancer drugs used for treating lung cancer often lead to apoptosis; however, the drug resistance of apoptosis restricts the effect of these drugs. Oncosis is a passive form of cell death that is different from apoptosis. It is characterized by cell swelling, and Porimin is a specific marker for oncosis. In this study, the role of DHT in mediating oncosis in A549 cells was investigated. In vitro, the MTS assay was used to detect cell activity after DHT treatment. Microscopy and electron microscopy were used to observe cell morphology changes. Western blotting was used to detect protein expression. Flow cytometry was used to detect intracellular reactive oxygen species (ROS) level, calcium ion (Ca2+) level, and cell mortality. The intracellular Lactic dehydrogenase (LDH) level was detected by an LDH detection kit after DHT treatment. The ATP level was detected using an ATP detection kit. In vivo, Lewis lung cancer (LLC) xenograft mice were used to evaluate the anti-tumor effect of DHT. Hematoxylin and eosin (HE) staining was used to detect the pathology of lung cancer tumors. The detection of Porimin in the tumor tissues of the mice after DHT administration was assessed by immunohistochemistry (IHC). The results of this study showed that DHT treatment changed the cell morphology; destroyed the mitochondrial structure; increased the expression of Porimin; increased the levels of LDH, ROS, and Ca2+; decreased the mitochondrial membrane potential and ATP level; and played an anti-tumor role in vitro by mediating oncosis in A549 cells. The in vivo studies showed that DHT could effectively inhibit tumor growth. The results of protein detection and IHC detection in the tumor tissues showed that the expression of Porimin was increased and that oncosis occurred in the tumor tissues of mice. DHT triggered Porimin-dependent oncosis by ROS-mediated mitochondrial dysfunction in NSCLC. The in vivo studies showed that DHT could inhibit tumor growth in LLC xenograft mice by triggering oncosis. This study indicates the potential for DHT to treat NSCLC.
Collapse
Affiliation(s)
- Dongjie Zhang
- College of Basic Medical, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Renyikun Yuan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jiaping Pan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Qiumei Fan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Kaili Sun
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhipeng Xu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xiang Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Qinqin Wang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jia He
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Yaqing Ye
- College of Basic Medical, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhengrong Mu
- College of Basic Medical, Guangxi Medical University, Nanning 530200, China
| | - Jing Leng
- College of Basic Medical, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
8
|
Nakao H, Yamaguchi T, Kawabata K, Higashi K, Nonaka M, Tuiji M, Nagai Y, Toyoda H, Yamaguchi Y, Kawasaki N, Kawasaki T. Characterization of novel antibodies that recognize sialylated keratan sulfate and lacto-N-fucopentaose I on human induced pluripotent cells: comparison with existing antibodies. Glycobiology 2023; 33:150-164. [PMID: 36373215 DOI: 10.1093/glycob/cwac074] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
This report describes the isolation and characterization of two new antibodies, R-6C (IgM) and R-13E (IgM), which were generated in C57BL/6 mice (Mus musculus) using the Tic (JCRB1331) human induced pluripotent cell (hiPSC) line as an antigen, and their comparisons with two existing antibodies, R-10G (IgG1) and R-17F (IgG1). Their epitopes were studied by western blotting after various glycosidase digestions, binding analyses using enzyme-linked immunosorbent assays (ELISAs) and microarrays with various synthetic oligosaccharides. The minimum epitope structures identified were: Siaα2-3Galβ1-3GlcNAc(6S)β1-3Galβ1-4GlcNAc(6S)β1 (R-6C), Fucα1-2Galβ1-3GlcNAcβ1-3Galβ1 (R-13E), Galβ1-4GlcNAc(6S)β1-3Galβ1-4GlcNAc(6S)β1 (R-10G), and Fucα1-2Galβ1-3GlcNAβ1-3Galβ1-4Glc (lacto-N-fucopentaose I) (R-17F). Most glycoprotein epitopes are expressed as O-glycans. The common feature of these epitopes is the presence of an N-acetyllactosamine type 1 structure (Galβ1-3GlcNAc) at their nonreducing termini, followed by a type 2 structure (Galβ1-4GlcNAc); this arrangement comprises a type 1-type 2 motif. This motif is also shared by TRA-1-60, a traditional onco-fetal antigen. In contrast, the R-10G epitope has a type 2-type 2 motif. Among these antibodies, R-17F and R-13E exhibit cytotoxic activity toward hiPSCs. R-17F and R-13E exhibit extremely high similarity in the amino acid sequences in their complementarity-determining regions (CDRs), which is consistent with their highly similar glycan recognition. These antibodies are excellent tools for investigating the biological functions of glycoconjugates in hiPSCs/hESCs; they could be useful for the selection, isolation and selective killing of such undifferentiated pluripotent stem cells.
Collapse
Affiliation(s)
- Hiromi Nakao
- Glycobiotechnology Laboratory, Research Organization of Science and Technology, Ritsumeikan University, Noji-Higashi 1-1-1, Kusatsu, Shiga 525-8577, Japan
| | - Tomoko Yamaguchi
- Laboratory of Cell Model for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Saito-Asagi 7-6-8, Ibaraki, Osaka 567-0085, Japan
| | - Kenji Kawabata
- Laboratory of Cell Model for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Saito-Asagi 7-6-8, Ibaraki, Osaka 567-0085, Japan
| | - Katsuaki Higashi
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Shogoin-Kawaharacho 53, Sakyo-ku, Kyoto, Kyoto 606-8507, Japan
| | - Motohiro Nonaka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Shogoin-Kawaharacho 53, Sakyo-ku, Kyoto, Kyoto 606-8507, Japan
| | - Makoto Tuiji
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yuko Nagai
- Laboratory of Bio-analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga 525-8577, Japan
| | - Hidenao Toyoda
- Laboratory of Bio-analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga 525-8577, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Komatsushima 4-4-1, Aobaku, Sendai, Miyagi 981-8558, Japan
| | - Nobuko Kawasaki
- Glycobiotechnology Laboratory, Research Organization of Science and Technology, Ritsumeikan University, Noji-Higashi 1-1-1, Kusatsu, Shiga 525-8577, Japan
| | - Toshisuke Kawasaki
- Glycobiotechnology Laboratory, Research Organization of Science and Technology, Ritsumeikan University, Noji-Higashi 1-1-1, Kusatsu, Shiga 525-8577, Japan.,Laboratory of Cell Model for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Saito-Asagi 7-6-8, Ibaraki, Osaka 567-0085, Japan
| |
Collapse
|
9
|
Pomlok K, Pata S, Kulaphisit M, Pangnuchar R, Wipasa J, Smith DR, Kasinrerk W, Lithanatudom P. An IgM monoclonal antibody against domain 1 of CD147 induces non-canonical RIPK-independent necroptosis in a cell type specific manner in hepatocellular carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119295. [PMID: 35598753 DOI: 10.1016/j.bbamcr.2022.119295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
CD147/Basigin/EMMPRIN is overexpressed in several cancerous tissues and it has been shown to induce matrix metalloproteinases (MMPs) whose expression is associated with cancer metastasis. Thus, targeting CD147 with monoclonal antibodies (mAbs) potentially has therapeutic applications in cancer immunotherapy. Here, we report the use of anti-CD147 mAbs targeting domain 1 of CD147, namely M6-1D4 (IgM), M6-1F3 (IgM), M6-2F9 (IgM) and M6-1E9 (IgG2a), against several human cancer cell lines. Strikingly, IgM but not IgG mAbs against CD147, especially clone M6-1D4, induced acute cellular swelling, and this phenomenon appeared to be specifically found with hepatocellular carcinoma (HCC) cells. Furthermore, molecular investigation upon treating HepG2 cells with M6-1D4 showed unfolded protein response (UPR) activation, autophagosome accumulation, and cell cycle arrest, but without classic apoptosis related features. More interestingly, prolonged M6-1D4 treatment (24 h) resulted in irreversible oncosis leading to necroptosis. Furthermore, treatment with a mixed lineage kinase domain-like psuedokinase (MLKL) inhibitor and partial knockout of MLKL resulted in reduced sensitivity to necroptosis in M6-1D4-treated HepG2 cells. Surprisingly however, the observed necroptotic signaling axis appeared to be non-canonical as it was independent of receptor-interacting serine/threonine-protein kinase (RIPK) phosphorylation. In addition, no cytotoxic effect on human dermal fibroblast (HDF) was observed after incubation with M6-1D4. Taken together, this study provides clues to target CD147 in HCC using mAbs, as well as sheds new light on a novel strategy to kill cancerous cells by the induction of necroptosis.
Collapse
Affiliation(s)
- Kumpanat Pomlok
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Ph.D.'s Degree Program in Biology (International Program), Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Supansa Pata
- Clinical Immunology Branch, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Mattapong Kulaphisit
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Ph.D.'s Degree Program in Biology (International Program), Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Rachan Pangnuchar
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraprapa Wipasa
- Center for Molecular and Cell Biology for Infectious Diseases, Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Watchara Kasinrerk
- Clinical Immunology Branch, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pathrapol Lithanatudom
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Bioresources for Agriculture, Industry and Medicine, Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
10
|
Wang K, Shao X, Tian Z, Liu L, Zhang C, Tan C, Zhang J, Ling P, Liu F, Chen Q, Diao J, Mao Z. A Continuous Add-On Probe Reveals the Nonlinear Enlargement of Mitochondria in Light-Activated Oncosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004566. [PMID: 34197052 PMCID: PMC8425930 DOI: 10.1002/advs.202004566] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/03/2021] [Indexed: 05/25/2023]
Abstract
Oncosis, depending on DNA damage and mitochondrial swelling, is an important approach for treating cancer and other diseases. However, little is known about the behavior of mitochondria during oncosis, due to the lack of probes for in situ visual illumination of the mitochondrial membrane and mtDNA. Herein, a mitochondrial lipid and mtDNA dual-labeled probe, MitoMN, and a continuous add-on assay, are designed to image the dynamic process of mitochondria in conditions that are unobservable with current mitochondrial probes. Meanwhile, the MitoMN can induce oncosis in a light-activated manner, which results in the enlargement of mitochondria and the death of cancer cells. Using structured illumination microscopy (SIM), MitoMN-stained mitochondria with a dual-color response reveals, for the first time, how swelled mitochondria interacts and fuses with each other for a nonlinear enlargement to accelerate oncosis into an irreversible stage. With this sign of irreversible oncosis revealed by MitoMN, oncosis can be segregated into three stages, including before oncosis, initial oncosis, and accelerated oncosis.
Collapse
Affiliation(s)
- Kang‐Nan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Xintian Shao
- Institute of Materia MedicaShandong First Medical University & Shandong Academy of Medical SciencesJinan250000P. R. China
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
| | - Zhiqi Tian
- Department of Molecular Genetics, Biochemistry, and MicrobiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
| | - Liu‐Yi Liu
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Chengying Zhang
- Institute of Materia MedicaShandong First Medical University & Shandong Academy of Medical SciencesJinan250000P. R. China
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
- School of Pharmaceutical SciencesShandong UniversityJinan250101P. R. China
| | - Cai‐Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Jie Zhang
- Advanced Medical Research Institute/Translational Medicine Core Facility of Advanced Medical Research InstituteShandong UniversityJinan250101P. R. China
| | - Peixue Ling
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
- School of Pharmaceutical SciencesShandong UniversityJinan250101P. R. China
| | - Fei Liu
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
- School of Pharmaceutical SciencesShandong UniversityJinan250101P. R. China
| | - Qixin Chen
- Institute of Materia MedicaShandong First Medical University & Shandong Academy of Medical SciencesJinan250000P. R. China
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
| | - Jiajie Diao
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
| | - Zong‐Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| |
Collapse
|
11
|
Ji S, Yang X, Chen X, Li A, Yan D, Xu H, Fei H. Structure-tuned membrane active Ir-complexed oligoarginine overcomes cancer cell drug resistance and triggers immune responses in mice. Chem Sci 2020; 11:9126-9133. [PMID: 34094193 PMCID: PMC8161536 DOI: 10.1039/d0sc03975f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/21/2022] Open
Abstract
The development of chemotherapy, an important cancer treatment modality, is hindered by the frequently found drug-resistance phenomenon. Meanwhile, researchers have been enthused lately by the synergistic use of chemotherapy with emerging immunotherapeutic treatments. In an effort to address both of the two unmet needs, reported herein is a study on a series of membrane active iridium(iii) complexed oligoarginine peptides with a new cell death mechanism capable of overcoming drug resistance as well as stimulating immunological responses. A systematic structure-activity relationship study elucidated the interdependent effects of three structural factors, i.e., hydrophobicity, topology and cationicity, on the regulation of the cytotoxicity of the Ir(iii)-oligoarginine peptides. With the most prominent toxicities, Ir-complexed octaarginines (R8) were found to display a progressive oncotic cell death featuring cell membrane-penetration and eruptive cytoplasmic content release. Consequently, this membrane-centric death mechanism showed promising potential in overcoming multiple chemical drug-resistance of cancer cells. More interestingly, the eruptive mode of cell death proved to be immunogenic by stimulating the dendritic cell maturation and inflammatory factor accumulation in mice tumours. Taking these mechanisms together, this work demonstrates that membrane active compounds may become the next generation chemotherapeutics because of their combined advantages.
Collapse
Affiliation(s)
- Shuangshuang Ji
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Xiuzhu Yang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Xiaolong Chen
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Ang Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Doudou Yan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing 100005 PR China
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing 100005 PR China
| | - Hao Fei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| |
Collapse
|
12
|
Vankemmelbeke M, McIntosh RS, Chua JX, Kirk T, Daniels I, Patsalidou M, Moss R, Parsons T, Scott D, Harris G, Ramage JM, Spendlove I, Durrant LG. Engineering the Human Fc Region Enables Direct Cell Killing by Cancer Glycan-Targeting Antibodies without the Need for Immune Effector Cells or Complement. Cancer Res 2020; 80:3399-3412. [PMID: 32532823 DOI: 10.1158/0008-5472.can-19-3599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/25/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022]
Abstract
Murine IgG3 glycan-targeting mAb often induces direct cell killing in the absence of immune effector cells or complement via a proinflammatory mechanism resembling oncotic necrosis. This cancer cell killing is due to noncovalent association between Fc regions of neighboring antibodies, resulting in enhanced avidity. Human isotypes do not contain the residues underlying this cooperative binding mode; consequently, the direct cell killing of mouse IgG3 mAb is lost upon chimerization or humanization. Using the Lewisa/c/x -targeting 88mAb, we identified the murine IgG3 residues underlying the direct cell killing and increased avidity via a series of constant region shuffling and subdomain swapping approaches to create improved ("i") chimeric mAb with enhanced tumor killing in vitro and in vivo. Constant region shuffling identified a major CH3 and a minor CH2 contribution, which was further mapped to discontinuous regions among residues 286-306 and 339-378 that, when introduced in 88hIgG1, recapitulated the direct cell killing and avidity of 88mIgG3. Of greater interest was the creation of a sialyl-di-Lewisa-targeting i129G1 mAb via introduction of these selected residues into 129hIgG1, converting it into a direct cell killing mAb with enhanced avidity and significant in vivo tumor control. The human iG1 mAb, termed Avidimabs, retained effector functions, paving the way for the proinflammatory direct cell killing to promote antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity through relief of immunosuppression. Ultimately, Fc engineering of human glycan-targeting IgG1 mAb confers proinflammatory direct cell killing and enhanced avidity, an approach that could be used to improve the avidity of other mAb with therapeutic potential. SIGNIFICANCE: Fc engineering enhances avidity and direct cell killing of cancer-targeting anti-glycan antibodies to create superior clinical candidates for cancer immunotherapy.
Collapse
Affiliation(s)
- Mireille Vankemmelbeke
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom.,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Richard S McIntosh
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Jia Xin Chua
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom.,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Thomas Kirk
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom.,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Ian Daniels
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Marilena Patsalidou
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Robert Moss
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Tina Parsons
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - David Scott
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, United Kingdom
| | - Gemma Harris
- Research Complex at Harwell, Rutherford Appleton Laboratory, Didcot, United Kingdom
| | - Judith M Ramage
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Ian Spendlove
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| | - Lindy G Durrant
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom. .,Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, United Kingdom
| |
Collapse
|
13
|
Yu J, Zhong B, Xiao Q, Du L, Hou Y, Sun HS, Lu JJ, Chen X. Induction of programmed necrosis: A novel anti-cancer strategy for natural compounds. Pharmacol Ther 2020; 214:107593. [PMID: 32492512 DOI: 10.1016/j.pharmthera.2020.107593] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2020] [Indexed: 02/08/2023]
Abstract
Cell death plays a critical role in organism development and the pathogenesis of diseases. Necrosis is considered a non-programmed cell death in an extreme environment. Recent advances have provided solid evidence that necrosis could be programmed and quite a few types of programmed necrosis, such as necroptosis, ferroptosis, pyroptosis, paraptosis, mitochondrial permeability transition-driven necrosis, and oncosis, have been identified. The specific biomarkers, detailed signaling, and precise pathophysiological importance of programmed necrosis are yet to be clarified, but these forms of necrosis provide novel strategies for the treatment of various diseases, including cancer. Natural compounds are a unique source of lead compounds for the discovery of anti-cancer drugs. Natural compounds can induce both apoptosis and programmed necrosis. In this review, we summarized the recent progress of programmed necrosis and introduced their natural inducers. Noptosis, which is a novel type of programmed necrosis that is strictly dependent on NAD(P)H: quinone oxidoreductase 1-derived oxidative stress was proposed. Furthermore, the anti-cancer strategies that take advantage of programmed necrosis and the main concerns from the scientific community in this regard were discussed.
Collapse
Affiliation(s)
- Jie Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Bingling Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Qingwen Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Lida Du
- Department of Surgery, University of Toronto, Ontario, Canada
| | - Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hong-Shuo Sun
- Department of Surgery, University of Toronto, Ontario, Canada
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
14
|
Tan HL, Choo A. Opportunities for Antibody Discovery Using Human Pluripotent Stem Cells: Conservation of Oncofetal Targets. Int J Mol Sci 2019; 20:E5752. [PMID: 31731794 PMCID: PMC6888136 DOI: 10.3390/ijms20225752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Pluripotent stem cells (PSCs) comprise both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). The application of pluripotent stem cells is divided into four main areas, namely: (i) regenerative therapy, (ii) the study and understanding of developmental biology, (iii) drug screening and toxicology and (iv) disease modeling. In this review, we describe a new opportunity for PSCs, the discovery of new biomarkers and generating antibodies against these biomarkers. PSCs are good sources of immunogen for raising monoclonal antibodies (mAbs) because of the conservation of oncofetal antigens between PSCs and cancer cells. Hence mAbs generated using PSCs can potentially be applied in two different fields. First, these mAbs can be used in regenerative cell therapy to characterize the PSCs. In addition, the mAbs can be used to separate or eliminate contaminating or residual undifferentiated PSCs from the differentiated cell product. This step is critical as undifferentiated PSCs can form teratomas in vivo. The mAbs generated against PSCs can also be used in the field of oncology. Here, novel targets can be identified and the mAbs developed as targeted therapy to kill the cancer cells. Conversely, as new and novel oncofetal biomarkers are discovered on PSCs, cancer mAbs that are already approved by the FDA can be repurposed for regenerative medicine, thus expediting the route to the clinics.
Collapse
Affiliation(s)
- Heng Liang Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138668, Singapore;
| | - Andre Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138668, Singapore;
- Department of Biochemical Engineering, National University of Singapore, Singapore 117575, Singapore
| |
Collapse
|
15
|
Li L, Wang J, Li Y, Radford DC, Yang J, Kopeček J. Broadening and Enhancing Functions of Antibodies by Self-Assembling Multimerization at Cell Surface. ACS NANO 2019; 13:11422-11432. [PMID: 31553883 PMCID: PMC6812323 DOI: 10.1021/acsnano.9b04868] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Monoclonal antibody therapy has offered treatment benefits. Nonetheless, a lack of efficacy still exists, partially because monovalent binding of antibodies to specific receptors fails to translate into an active response. Here, we report a pretargeting-postassembly approach that exploits the selective Watson-Crick base pairing properties of oligonucleotides and multivalently tethers receptor-prebound antibodies to albumin at the cell surface. We demonstrate that this two-step self-assembling strategy allows sequential actions of receptor binding and clustering that broadens and strengthens the functions of antibodies. We show that anti-CD20 obinutuzumab (OBN) modified with one morpholino oligonucleotide (OBN-MORF1) maintains the feature of naked OBN antibody upon CD20 binding, and results in actin redistribution, homotypic adhesion, and lysosome-mediated cell death. Consecutive treatment with albumin grafted with multiple copies of a complementary morpholino oligonucleotide (HSA-(MORF2)x) hybridizes with surface-attached OBN-MORF1, manipulates CD20 clustering, and engages additional signals to induce calcium influx and caspase-related apoptosis. With the two types of different mechanisms collaborating in one system, the simple design exerted a notable survival extension of mice bearing disseminated B-cell lymphomas.
Collapse
Affiliation(s)
- Lian Li
- Department
of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled
Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jiawei Wang
- Department
of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled
Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
| | - Yachao Li
- Department
of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled
Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
| | - D. Christopher Radford
- Department
of Biomedical Engineering, University of
Utah, Salt Lake City, Utah 84112, United
States
| | - Jiyuan Yang
- Department
of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled
Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- E-mail:
| | - Jindřich Kopeček
- Department
of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled
Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department
of Biomedical Engineering, University of
Utah, Salt Lake City, Utah 84112, United
States
- E-mail:
| |
Collapse
|
16
|
Tan HL, Tan BZ, Goh WXT, Cua S, Choo A. In vivo surveillance and elimination of teratoma-forming human embryonic stem cells with monoclonal antibody 2448 targeting annexin A2. Biotechnol Bioeng 2019; 116:2996-3005. [PMID: 31388993 PMCID: PMC6790577 DOI: 10.1002/bit.27135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022]
Abstract
This study describes the use of a previously reported chimerised monoclonal antibody (mAb), ch2448, to kill human embryonic stem cells (hESCs) in vivo and prevent or delay the formation of teratomas. ch2448 was raised against hESCs and was previously shown to effectively kill ovarian and breast cancer cells in vitro and in vivo. The antigen target was subsequently found to be Annexin A2, an oncofetal antigen expressed on both embryonic cells and cancer cells. Against cancer cells, ch2448 binds and kills via antibody‐dependent cell‐mediated cytotoxicity (ADCC) and/or antibody‐drug conjugate (ADC) routes. Here, we investigate if the use of ch2448 can be extended to hESC. ch2448 was found to bind specifically to undifferentiated hESC but not differentiated progenitors. Similar to previous study using cancer cells, ch2448 kills hESC in vivo either indirectly by eliciting ADCC or directly as an ADC. The treatment with ch2448 post‐transplantation eliminated the in vivo circulating undifferentiated cells and prevented or delayed the formation of teratomas. This surveillance role of ch2448 adds an additional layer of safeguard to enhance the safety and efficacious use of pluripotent stem cell‐derived products in regenerative medicine. Thereby, translating the use of ch2448 in the treatment of cancers to a proof of concept study in hESC (or pluripotent stem cell [PSC]), we show that mAbs can also be used to eliminate teratoma forming cells in vivo during PSC‐derived cell therapies. We propose to use this strategy to complement existing methods to eliminate teratoma‐forming cells in vitro. Residual undifferentiated cells may escape in vitro removal methods and be introduced into patients together with the differentiated cells.
Collapse
Affiliation(s)
- Heng Liang Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Bao Zhu Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Winfred Xi Tai Goh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Simeon Cua
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Andre Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| |
Collapse
|
17
|
Tan HL, Yong C, Tan BZ, Fong WJ, Padmanabhan J, Chin A, Ding V, Lau A, Zheng L, Bi X, Yang Y, Choo A. Conservation of oncofetal antigens on human embryonic stem cells enables discovery of monoclonal antibodies against cancer. Sci Rep 2018; 8:11608. [PMID: 30072783 PMCID: PMC6072701 DOI: 10.1038/s41598-018-30070-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 06/26/2018] [Indexed: 12/26/2022] Open
Abstract
Monoclonal antibodies (mAbs) are used as targeted therapies against cancers. These mAbs kill cancer cells via various mechanisms of actions. In this study, human embryonic stem cells (hESCs) was used as the immunogen to generate a panel of antibodies. From this panel of mAbs, A19 was found to bind both hESC and various cancer cell lines. The antigen target of A19 was identified as Erbb-2 and glycan analysis showed that A19 binds to a N-glycan epitope on the antigen. A19 was elucidated to internalize into cancer cells following binding to Erbb-2 and hence developed as an antibody-drug conjugate (ADC). Using ADC as the mechanism of action, A19 was able to kill cancer cells in vitro and delayed the onset of tumour formation in mice xenograft model. When compared to Herceptin, A19 binds to different isoforms of Erbb-2 and does not compete with Herceptin for the same epitope. Hence, A19 has the potential to be developed as an alternative targeted therapeutic agent for cancers expressing Erbb-2.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/immunology
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Female
- Human Embryonic Stem Cells/immunology
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Heng Liang Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore.
| | - Charlene Yong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Bao Zhu Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Wey Jia Fong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Jayanthi Padmanabhan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Angela Chin
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Vanessa Ding
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Ally Lau
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Lu Zheng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Andre Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| |
Collapse
|
18
|
Oncosis-like cell death is induced by berberine through ERK1/2-mediated impairment of mitochondrial aerobic respiration in gliomas. Biomed Pharmacother 2018; 102:699-710. [PMID: 29604589 DOI: 10.1016/j.biopha.2018.03.132] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/12/2022] Open
Abstract
Gliomas, the most common primary malignant brain tumor, exhibit high metabolic activity. The targeting of metabolism alterations, particularly in mitochondria, is emerging as an efficient approach for curing cancers. Here, we showed that berberine, a natural compound that is used as an antibacterial agent, could reduce cellular viability and induce oncosis-like death, characterized by cell swelling, cytoplasmic vacuoles and plasma membrane blebbing, in gliomas, and that these effects were correlated with intracellular adenosine triphosphate (ATP) depletion. We also found that berberine induced autophagy as a protective effect and decreased the oxygen consumption rate (OCR), which could inhibit mitochondrial aerobic respiration by repressing phosphorylated extracellular regulated protein kinases (p-ERK1/2). Furthermore, the down-regulation of mitochondrial p-ERK1/2 by berberine inhibited aerobic respiration and led to glycolysis, an inefficient energy production pathway. In addition, berberine reduced tumor growth and inhibited Ki-67 and p-ERK1/2 expression in vivo. The results demonstrate that berberine, which represses aerobic oxidation in mitochondria and decreases their energy production efficiency, decreases metabolic activity by reducing ERK1/2 activity.
Collapse
|
19
|
AML-specific cytotoxic antibodies in patients with durable graft-versus-leukemia responses. Blood 2017; 131:131-143. [PMID: 29061569 DOI: 10.1182/blood-2017-02-768762] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 09/30/2017] [Indexed: 12/30/2022] Open
Abstract
Most patients with acute myeloid leukemia (AML) can only be cured when allogeneic hematopoietic stem-cell transplantation induces a graft-versus-leukemia immune response (GVL). Although the role of T cells and natural killer cells in tumor immunology has been established, less is known about the contribution of B cells. From B cells of high-risk patients with AML with potent and lasting GVL responses, we isolated monoclonal antibodies directed against antigens expressed on the cell surface of AML cells but not on normal hematopoietic and nonhematopoietic cells. A number of these donor-derived antibodies recognized the U5 snRNP200 complex, a component of the spliceosome that in normal cells is found in the cell. In AML however, the U5 snRNP200 complex is exposed on the cell membrane of leukemic blasts. U5 snRNP200 complex-specific antibodies induced death of AML cells in an Fc receptor-dependent way in the absence of cytotoxic leukocytes or complement. In an AML mouse model, treatment with U5 snRNP200 complex-specific antibodies led to significant tumor growth inhibition. Thus, donor-derived U5 snRNP200 complex-recognizing AML-specific antibodies may contribute to antitumor responses.
Collapse
|
20
|
Kashyap MK, Amaya-Chanaga CI, Kumar D, Simmons B, Huser N, Gu Y, Hallin M, Lindquist K, Yafawi R, Choi MY, Amine AA, Rassenti LZ, Zhang C, Liu SH, Smeal T, Fantin VR, Kipps TJ, Pernasetti F, Castro JE. Targeting the CXCR4 pathway using a novel anti-CXCR4 IgG1 antibody (PF-06747143) in chronic lymphocytic leukemia. J Hematol Oncol 2017. [PMID: 28526063 DOI: 10.1186/s13045-017-0435-x,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The CXCR4-CXCL12 axis plays an important role in the chronic lymphocytic leukemia (CLL)-microenvironment interaction. Overexpression of CXCR4 has been reported in different hematological malignancies including CLL. Binding of the pro-survival chemokine CXCL12 with its cognate receptor CXCR4 induces cell migration. CXCL12/CXCR4 signaling axis promotes cell survival and proliferation and may contribute to the tropism of leukemia cells towards lymphoid tissues and bone marrow. Therefore, we hypothesized that targeting CXCR4 with an IgG1 antibody, PF-06747143, may constitute an effective therapeutic approach for CLL. METHODS Patient-derived primary CLL-B cells were assessed for cytotoxicity in an in vitro model of CLL microenvironment. PF-06747143 was analyzed for cell death induction and for its potential to interfere with the chemokine CXCL12-induced mechanisms, including migration and F-actin polymerization. PF-06747143 in vivo efficacy was determined in a CLL murine xenograft tumor model. RESULTS PF-06747143, a novel-humanized IgG1 CXCR4 antagonist antibody, induced cell death of patient-derived primary CLL-B cells, in presence or absence of stromal cells. Moreover, cell death induction by the antibody was independent of CLL high-risk prognostic markers. The cell death mechanism was dependent on CXCR4 expression, required antibody bivalency, involved reactive oxygen species production, and did not require caspase activation, all characteristics reminiscent of programmed cell death (PCD). PF-06747143 also induced potent B-CLL cytotoxicity via Fc-driven antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity activity (CDC). PF-06747143 had significant combinatorial effect with standard of care (SOC) agents in B-CLL treatment, including rituximab, fludarabine (F-ara-A), ibrutinib, and bendamustine. In a CLL xenograft model, PF-06747143 decreased tumor burden and improved survival as a monotherapy, and in combination with bendamustine. CONCLUSIONS We show evidence that PF-06747143 has biological activity in CLL primary cells, supporting a rationale for evaluation of PF-06747143 for the treatment of CLL patients.
Collapse
Affiliation(s)
- Manoj K Kashyap
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Carlos I Amaya-Chanaga
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Deepak Kumar
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Brett Simmons
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Nanni Huser
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Yin Gu
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Max Hallin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.,Present Address: Mirati Therapeutics, San Diego, CA, USA
| | - Kevin Lindquist
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Rolla Yafawi
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, CA, USA
| | - Michael Y Choi
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ale-Ali Amine
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Laura Z Rassenti
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cathy Zhang
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Shu-Hui Liu
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Tod Smeal
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.,Present Address: Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Valeria R Fantin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.,Present Address: ORIC Pharmaceuticals, South San Francisco, CA, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Flavia Pernasetti
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.
| | - Januario E Castro
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA. .,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
21
|
Kashyap MK, Amaya-Chanaga CI, Kumar D, Simmons B, Huser N, Gu Y, Hallin M, Lindquist K, Yafawi R, Choi MY, Amine AA, Rassenti LZ, Zhang C, Liu SH, Smeal T, Fantin VR, Kipps TJ, Pernasetti F, Castro JE. Targeting the CXCR4 pathway using a novel anti-CXCR4 IgG1 antibody (PF-06747143) in chronic lymphocytic leukemia. J Hematol Oncol 2017; 10:112. [PMID: 28526063 PMCID: PMC5438492 DOI: 10.1186/s13045-017-0435-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The CXCR4-CXCL12 axis plays an important role in the chronic lymphocytic leukemia (CLL)-microenvironment interaction. Overexpression of CXCR4 has been reported in different hematological malignancies including CLL. Binding of the pro-survival chemokine CXCL12 with its cognate receptor CXCR4 induces cell migration. CXCL12/CXCR4 signaling axis promotes cell survival and proliferation and may contribute to the tropism of leukemia cells towards lymphoid tissues and bone marrow. Therefore, we hypothesized that targeting CXCR4 with an IgG1 antibody, PF-06747143, may constitute an effective therapeutic approach for CLL. METHODS Patient-derived primary CLL-B cells were assessed for cytotoxicity in an in vitro model of CLL microenvironment. PF-06747143 was analyzed for cell death induction and for its potential to interfere with the chemokine CXCL12-induced mechanisms, including migration and F-actin polymerization. PF-06747143 in vivo efficacy was determined in a CLL murine xenograft tumor model. RESULTS PF-06747143, a novel-humanized IgG1 CXCR4 antagonist antibody, induced cell death of patient-derived primary CLL-B cells, in presence or absence of stromal cells. Moreover, cell death induction by the antibody was independent of CLL high-risk prognostic markers. The cell death mechanism was dependent on CXCR4 expression, required antibody bivalency, involved reactive oxygen species production, and did not require caspase activation, all characteristics reminiscent of programmed cell death (PCD). PF-06747143 also induced potent B-CLL cytotoxicity via Fc-driven antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity activity (CDC). PF-06747143 had significant combinatorial effect with standard of care (SOC) agents in B-CLL treatment, including rituximab, fludarabine (F-ara-A), ibrutinib, and bendamustine. In a CLL xenograft model, PF-06747143 decreased tumor burden and improved survival as a monotherapy, and in combination with bendamustine. CONCLUSIONS We show evidence that PF-06747143 has biological activity in CLL primary cells, supporting a rationale for evaluation of PF-06747143 for the treatment of CLL patients.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- CHO Cells
- Cell Death/drug effects
- Cricetulus
- Female
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/therapeutic use
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice, Inbred BALB C
- Mice, SCID
- Reactive Oxygen Species/immunology
- Receptors, CXCR4/analysis
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/immunology
- Signal Transduction/drug effects
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Manoj K Kashyap
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Carlos I Amaya-Chanaga
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Deepak Kumar
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Brett Simmons
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Nanni Huser
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Yin Gu
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Max Hallin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
- Present Address: Mirati Therapeutics, San Diego, CA, USA
| | - Kevin Lindquist
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Rolla Yafawi
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, CA, USA
| | - Michael Y Choi
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ale-Ali Amine
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Laura Z Rassenti
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cathy Zhang
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Shu-Hui Liu
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Tod Smeal
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
- Present Address: Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Valeria R Fantin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
- Present Address: ORIC Pharmaceuticals, South San Francisco, CA, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Flavia Pernasetti
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.
| | - Januario E Castro
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|