1
|
Bando H, Kumagai S, Kotani D, Mishima S, Irie T, Itahashi K, Tanaka Y, Habu T, Fukaya S, Kondo M, Tsushima T, Hara H, Kadowaki S, Kato K, Chin K, Yamaguchi K, Kageyama SI, Hojo H, Nakamura M, Tachibana H, Wakabayashi M, Fukui M, Fuse N, Koyama S, Mano H, Nishikawa H, Shitara K, Yoshino T, Kojima T. Atezolizumab following definitive chemoradiotherapy in patients with unresectable locally advanced esophageal squamous cell carcinoma - a multicenter phase 2 trial (EPOC1802). NATURE CANCER 2025; 6:445-459. [PMID: 39972105 PMCID: PMC11949839 DOI: 10.1038/s43018-025-00918-1] [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] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025]
Abstract
Platinum-based definitive chemoradiotherapy (dCRT) is the standard treatment for patients with unresectable locally advanced esophageal squamous cell carcinoma (ESCC) that invades the aorta, vertebral body or trachea; however, complete response rates remain low (11-25%), leading to poor survival. To evaluate the additive efficacy of the anti-PD-L1 antibody drug atezolizumab, we conducted a phase 2, multicenter, single-arm trial of 1 year of atezolizumab treatment following dCRT in 40 patients with unresectable locally advanced ESCC recruited from seven Japanese centers (UMIN000034373). The confirmed complete response (cCR) rate (primary end point) of the first consecutive 38 patients was 42.1% (90% CI 28.5-56.7%). Regarding the secondary end points, the median progression-free survival and 12-month progression-free survival rates of all 40 patients were 3.2 months and 29.6%, respectively, and the preliminary median overall survival with short-term follow-up and 12-month overall survival rate were 31.0 months and 65.8%, respectively. Other secondary end points evaluated included the cCR rate determined by an investigator's assessment in the locoregionally recurrent ESCC cohort, cCR rate determined by central assessment, overall response rate and incidence of adverse events. No treatment-related death occurred during the study. Atezolizumab monotherapy after dCRT resulted in a promising cCR rate, although long-term survival data are required.
Collapse
Affiliation(s)
- Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Shogo Kumagai
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
- Division of Cellular Signaling, Research Institute, National Cancer Center, Tokyo, Japan
| | - Daisuke Kotani
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Saori Mishima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takuma Irie
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
| | - Kota Itahashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
| | - Yosuke Tanaka
- Division of Cellular Signaling, Research Institute, National Cancer Center, Tokyo, Japan
| | - Takumi Habu
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Sayuri Fukaya
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaki Kondo
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Tsushima
- Division of Gastrointestinal Oncology Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroki Hara
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Ken Kato
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Keisho Chin
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shun-Ichiro Kageyama
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hidehiro Hojo
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masaki Nakamura
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hidenobu Tachibana
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masashi Wakabayashi
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Makoto Fukui
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Nozomu Fuse
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shohei Koyama
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, Research Institute, National Cancer Center, Tokyo, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Kashiwa, Japan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Kindai University Faculty of Medicine, Osaka, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
2
|
Syed Altaf RR, Mohan A, Palani N, Mendonce KC, Monisha P, Rajadesingu S. A review of innovative design strategies: Artificial antigen presenting cells in cancer immunotherapy. Int J Pharm 2025; 669:125053. [PMID: 39667594 DOI: 10.1016/j.ijpharm.2024.125053] [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: 09/22/2024] [Revised: 11/07/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Developing nanocarriers that can carry medications directly to tumors is an exciting development in cancer nanomedicine. The efficacy of this intriguing therapeutic approach is, however, compromised by intricate and immunosuppressive circumstances that arise concurrently with the onset of cancer. The artificial antigen presenting cell (aAPC), a micro or nanoparticle based device that mimics an antigen presenting cell by providing crucial signal proteins to T lymphocytes to activate them against cancer, is one cutting-edge method for cancer immunotherapy. This review delves into the critical design considerations for aAPCs, particularly focusing on particle size, shape, and the non-uniform distribution of T cell activating proteins on their surfaces. Adequate surface contact between T cells and aAPCs is essential for activation, prompting engineers to develop nano-aAPCs with microscale contact areas through techniques such as shape modification and nanoparticle clustering. Additionally, we explore recommendations for future advancements in this field.
Collapse
Affiliation(s)
- Rabiya Riffath Syed Altaf
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Agilandeswari Mohan
- Department of BioChemistry, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Naveen Palani
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Keren Celestina Mendonce
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - P Monisha
- PG & Research Department of Physics, Sri Sarada College for Women, Salem - 636016, Tamil Nadu, India
| | - Suriyaprakash Rajadesingu
- Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
3
|
Li Z, Jiang S, Wang J, Li W, Yang J, Liu W, Gao H, Huang Y, Ruan S. Peptide-drug conjugates repolarize glioblastoma-associated macrophages to resensitize chemo-immunotherapy of glioblastoma. SCIENCE ADVANCES 2025; 11:eadr8841. [PMID: 39823328 PMCID: PMC11740939 DOI: 10.1126/sciadv.adr8841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
The prevalent tumor-supporting glioblastoma-associated macrophages (GAMs) promote glioblastoma multiforme (GBM) progression and resistance to multiple therapies. Repolarizing GAMs from tumor-supporting to tumor-inhibiting phenotype may troubleshoot. However, sufficient accumulation of drugs at the GBM site is restricted by blood-brain barrier (BBB). Herein, we designed peptide-drug conjugates (PDCs) by conjugating camptothecin or resiquimod to a tandem peptide composed of matrix metalloproteinase 2-responsive peptide and angiopep-2 via disulfonyl-ethyl carbonate/carbamate (MAPDCs). The mixed self-assembly MAPDCs could recognize low-density lipoprotein receptor-related protein 1 (LRP1) to facilitate BBB transport. Once reaching the GBM site, the responsive peptide would be cleaved to shed the angiopep-2, blocking abluminal LRP1-mediated brain-to-blood efflux and enhancing drug retention. Sequentially, drugs are released under the high level of intracellular glutathione. In vivo studies demonstrated that MAPDCs repolarized GAMs, boosted immune response, and resensitized chemotherapeutic toxicity, offering a much-improved anti-GBM effect. The effectiveness of MAPDCs validates GAMs as therapeutic target and PDCs as versatile brain delivery system with high design flexibility.
Collapse
Affiliation(s)
- Zhi Li
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Shaoping Jiang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jie Wang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Wenpei Li
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jun Yang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Weimin Liu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Huile Gao
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuanyu Huang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Shaobo Ruan
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
4
|
da Silva LM, Martins MR, Dos Santos RL, Da Silva JPA, Lima CAC, Forones NM, Torres LC. Evaluation of soluble co-inhibitors and co-stimulators levels of the immune response in gastric cancer. J Surg Oncol 2024. [PMID: 38946193 DOI: 10.1002/jso.27747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Co-inhibitor and co-stimulator mediators trigger actions that result in immunological homeostasis and are being evaluated as potential therapeutic targets in gastric cancer (GC). OBJECTIVE To evaluate the soluble levels of sPD-1, sPD-L1, sPD-L2, sTIM-3, sGal9, sGITR, and sGITRL in GC patients. METHODS The cross-sectional study was carried out at the Hospital de Cancer de Pernambuco, Brazil between 2017 and 2018. A total of 74 GC patients and 30 healthy controls were included. RESULTS Low levels of sPD1 (p = 0.0179), sPDL2 (p = 0.0003), and sGal9 (p < 0.0001), and higher levels of sPDL1 (p = 0.004), sTIM-3 (p = 0.0072), sGITR (p = 0.0179), and sGITRL (p = 0.0055) compared to the control group. High sPD-1, sTIM-3, and sGal9 levels in stage IV compared I/II and III (p < 0.05). High sPDL1, sGal9, and sGITRL levels in esophagogastric junction compared to body and Pylorus/Antrum groups (p < 0.05). No significant differences were observed in sPD1, sPDL1, sPDL2, sTIM3, sGal9, sGITR, and sGITRL levels between the intestinal, diffuse, and mixed GC groups. Low sGITR levels in GC patients who died within the first 24 months compared to the who survived (p = 0.0332). CONCLUSIONS There is an association of sPD1, sTIM-3, and sGal9 with disease progression and sGITR with death, these mediators may be potential prognostic biomarkers in GC.
Collapse
Affiliation(s)
- Luciana Mata da Silva
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Medicine, Postgraduate Program in Translational Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Mário Rino Martins
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Medicine, Postgraduate Program in Translational Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Rogerio Luiz Dos Santos
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
| | - Jeronimo Paulo Assis Da Silva
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Medicine, Postgraduate Program in Translational Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Cecilia Araujo Carneiro Lima
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Medicine, Postgraduate Program in Translational Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Nora Manoukian Forones
- Department of Digestive Surgery, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Leuridan Cavalcante Torres
- Translational Research Laboratory, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Medicine, Postgraduate Program in Translational Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
5
|
de Castro NM, Moura F, Hada AL, Garcia D, Victor EDS, Schvartsman G, Carvalho L, Fernandes MLC, Martins RDS, da Silva EF, dos Santos SSMB, Taniwaki L, Taranto P, Pontes J, Beal JR, Dutra ACP, de Oliveira JB, Araujo SEA, Usón PLS. Prognostic value of programmed cell death ligand 1 (PD-L1) expression in patients with stage III non-small cell lung cancer under different treatment types: a retrospective study. EINSTEIN-SAO PAULO 2024; 22:eAO0575. [PMID: 38922219 PMCID: PMC11196088 DOI: 10.31744/einstein_journal/2024ao0575] [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: 05/21/2023] [Accepted: 09/19/2023] [Indexed: 06/27/2024] Open
Abstract
OBJECTIVE Currently programmed cell death protein 1 (PD-1) inhibitors in combination with other therapies are being evaluated to determine their efficacy in cancer treatment. However, the effect of PD-ligand (L) 1 expression on disease outcomes in stage III (EC III) non-small cell lung cancer is not completely understood. Therefore, this study aimed to assess the influence of PD-L1 expression on the outcomes of EC III non-small cell lung cancer. METHODS This study was conducted on patients diagnosed with EC III non-small cell lung cancer who underwent treatment at a tertiary care hospital. PD-L1 expression was determined using immunohistochemical staining, all patients expressed PD-L1. Survival was estimated using the Kaplan-Meier method. Relationships between variables were assessed using Cox proportional regression models. RESULTS A total of 49 patients (median age=69 years) with EC III non-small cell lung cancer and PD-L1 expression were evaluated. More than half of the patients were men, and most were regular smokers. The patients were treated with neoadjuvant chemotherapy, surgery, or sequential or combined chemotherapy and radiotherapy. The median progression-free survival of the entire cohort was 14.2 months, and the median overall survival was 20 months. There was no significant association between PD-L1 expression and disease progression, clinical characteristics, or overall survival. CONCLUSIONS PD-L1 expression was not correlated with EC III non-small cell lung cancer outcomes. Whether these findings differ from the association with immune checkpoint inhibitors remains to be addressed in future studies.
Collapse
Affiliation(s)
- Nicoly Marques de Castro
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Fernando Moura
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Aline Lury Hada
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Diogo Garcia
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Elivane da Silva Victor
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Gustavo Schvartsman
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Leonardo Carvalho
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Rodrigo de Souza Martins
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Elaine Ferreira da Silva
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Letícia Taniwaki
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Patrícia Taranto
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Janaina Pontes
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Juliana Rodrigues Beal
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Ana Carolina Pereira Dutra
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - João Bosco de Oliveira
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | | | - Pedro Luiz Serrano Usón
- Hospital Israelita Albert EinsteinSão PauloSPBrazil Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
6
|
Grant CE, Flis AL, Toulabi L, Zingone A, Rossi E, Aploks K, Sheppard H, Ryan BM. DRD1 suppresses cell proliferation and reduces EGFR activation and PD-L1 expression in NSCLC. Mol Oncol 2024; 18:1631-1648. [PMID: 38572507 PMCID: PMC11161724 DOI: 10.1002/1878-0261.13608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/28/2023] [Accepted: 02/06/2024] [Indexed: 04/05/2024] Open
Abstract
Dopamine (DA) acts in various key neurological and physiological processes as both a neurotransmitter and circulating hormone. Over the past several decades, the DA signaling network has been shown to regulate the progression of several types of solid tumors, and considerable evidence has shown it is a druggable pathway in the cancer cell context. However, the specific activity and effect of these pathway components appears to be tissue-type and cell-context-dependent. In the present study, expression and methylation of dopamine receptor D1 (DRD1) were measured using RNA sequencing (RNAseq) and reverse transcription polymerase chain reaction (RT-PCR) in non-small cell lung cancer (NSCLC) samples, and validated using publicly available datasets, including The Cancer Genome Atlas (TCGA). In vitro and in vivo functional experiments were performed for cell proliferation and tumor growth, respectively. Mechanistic analyses of the transcriptome and kinome in DRD1-modulated cells informed further experiments, which characterized the effects on the epidermal growth factor receptor (EGFR) pathway and programmed cell death 1 ligand 1 (PD-L1) proteins. Through these experiments, we identified the DRD1 gene as a negative regulator of disease progression in NSCLC. We show that DRD1, as well as other DA pathway components, are expressed in normal human lung tissue, and that loss of DRD1 expression through promoter hypermethylation is a common feature in NSCLC patients and is associated with worse survival. At the cellular level, DRD1 affects proliferation by inhibiting the activation of EGFR and mitogen-activated protein kinase 1/2 (ERK1/2). Interestingly, we also found that DRD1 regulates the expression of PD-L1 in lung cancer cells. Taken together, these results suggest that DRD1 methylation may constitute a biomarker of poor prognosis in NSCLC patients while other components of this pathway could be targeted to improve response to EGFR- and PD-L1-targeted therapies.
Collapse
Affiliation(s)
- Christopher E. Grant
- Laboratory of Human Carcinogenesis, Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Amy L. Flis
- Laboratory of Human Carcinogenesis, Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Leila Toulabi
- Laboratory of Human Carcinogenesis, Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Adriana Zingone
- Laboratory of Human Carcinogenesis, Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Emily Rossi
- Laboratory of Human Carcinogenesis, Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Krist Aploks
- Laboratory of Human Carcinogenesis, Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Heather Sheppard
- Veterinary Pathology CoreSt. Jude Children's Research HospitalMemphisTNUSA
| | - Bríd M. Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| |
Collapse
|
7
|
Han R, Rao X, Zhou H, Lu L. Synergistic Immunoregulation: harnessing CircRNAs and PiRNAs to Amplify PD-1/PD-L1 Inhibition Therapy. Int J Nanomedicine 2024; 19:4803-4834. [PMID: 38828205 PMCID: PMC11144010 DOI: 10.2147/ijn.s461289] [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: 01/24/2024] [Accepted: 05/12/2024] [Indexed: 06/05/2024] Open
Abstract
The utilization of PD-1/PD-L1 inhibitors marks a significant advancement in cancer therapy. However, the efficacy of monotherapy is still disappointing in a substantial subset of patients, necessitating the exploration of combinational strategies. Emerging from the promising results of the KEYNOTE-942 trial, RNA-based therapies, particularly circRNAs and piRNAs, have distinguished themselves as innovative sensitizers to immune checkpoint inhibitors (ICIs). These non-coding RNAs, notable for their stability and specificity, were once underrecognized but are now known for their crucial roles in regulating PD-L1 expression and bolstering anti-cancer immunity. Our manuscript offers a comprehensive analysis of selected circRNAs and piRNAs, elucidating their immunomodulatory effects and mechanisms, thus underscoring their potential as ICIs enhancers. In conjunction with the recent Nobel Prize-awarded advancements in mRNA vaccine technology, our review highlights the transformative implications of these findings for cancer treatment. We also discuss the prospects of circRNAs and piRNAs in future therapeutic applications and research. This study pioneers the synergistic application of circRNAs and piRNAs as novel sensitizers to augment PD-1/PD-L1 inhibition therapy, demonstrating their unique roles in regulating PD-L1 expression and modulating immune responses. Our findings offer a groundbreaking approach for enhancing the efficacy of cancer immunotherapy, opening new avenues for treatment strategies. This abstract aims to encapsulate the essence of our research and the burgeoning role of these non-coding RNAs in enhancing PD-1/PD-L1 inhibition therapy, encouraging further investigation into this promising field.
Collapse
Affiliation(s)
- Rui Han
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- Department of Chinese Medicine, Naval Medical University, Shanghai, People’s Republic of China
| | - Xiwu Rao
- Department of Oncology, The First Hospital Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People’s Republic of China
| | - Huiling Zhou
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People’s Republic of China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, USA
- School of Medicine, Center for Biomedical Data Science, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
8
|
Zhao Y, Yu Y, Li X, Guo A. CCAAT enhancer binding protein delta activates vesicle associated membrane protein 3 transcription to enhance chemoresistance and extracellular PD-L1 expression in triple-negative breast cancer. J Exp Clin Cancer Res 2024; 43:115. [PMID: 38627816 PMCID: PMC11020785 DOI: 10.1186/s13046-024-03041-8] [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: 01/24/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Chemoresistance and immunosuppression are two major obstacles in the current anti-cancer treatments. This study investigates the involvements of a CCAAT enhancer binding protein delta (CEBPD)/vesicle associated membrane protein 3 (VAMP3) axis in paclitaxel (PTX) resistance and immune evasion in triple-negative breast cancer (TNBC). METHODS PTX resistance-related genes were screened by bioinformatics. CEBPD and VAMP3 expression in clinical TNBC samples was examined by immunohistochemistry. Three PTX-resistant TNBC cell lines (MDA-MB-231/PTX, MDA-MB-468/PTX and MDA-MB-453/PTX) were generated, and their drug resistance was analyzed. Autophagy of cells was analyzed by immunofluorescence staining. Interaction between CEBPD and VAMP3 promoter was identified by immunoprecipitation and luciferase assays. The extracellular expression of programmed cell death-ligand 1 (PD-L1) in TNBC cells was detected. Extracellular vesicles (EVs) from TNBC cells were isolated to examine their effects on CD8+ T cell exhaustion. RESULTS CEBPD and VAMP3 were upregulated in chemo-resistant tissue samples and in PTX-resistant TNBC cells. The CEBPD downregulation enhanced PTX sensitivity of cells. However, further upregulation of VAMP3 in cells restored PTX resistance, which was likely due to the activation of autophagy, as the autophagy antagonist chloroquine enhanced PTX sensitivity of cells. CEBPD was found to bind to the VAMP3 promoter to activate its transcription. The CEBPD/VAMP3 axis also increased the PD-L1 expression in the conditioned medium of TNBC cells. The TNBC cell-derived EVs increased the exhaustion of co-cultured CD8+ T cells. CONCLUSION This study provides novel evidence that CEBPD plays a key role in enhancing PTX resistance in TNBC cells across various subtypes through VAMP3-mediated autophagy activation. Additionally, the CEBPD/VAMP3 axis also increases extracellular PD-L1 level, delivered by cancer cell-derived EVs, to suppress CD8+ T cell-mediated anti-tumor immune response. These significant observations may provide new insights into the treatment of TNBC, suggesting CEBPD and VAMP3 as promising targets to overcome treatment resistance.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Breast Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, Liaoning, 110001, P.R. China
| | - Yangyang Yu
- Department of Radiation Oncology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, Liaoning, 110001, P.R. China
| | - Xiangmin Li
- Department of Oncology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, P.R. China.
| | - Ayao Guo
- Department of Breast Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, Liaoning, 110001, P.R. China.
| |
Collapse
|
9
|
Zhou S, Wang B, Wei Y, Dai P, Chen Y, Xiao Y, Xia H, Chen C, Yin W. PD-1 inhibitor combined with Docetaxel exerts synergistic anti-prostate cancer effect in mice by down-regulating the expression of PI3K/AKT/NFKB-P65/PD-L1 signaling pathway. Cancer Biomark 2024; 40:47-59. [PMID: 38306024 PMCID: PMC11307039 DOI: 10.3233/cbm-230090] [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: 05/31/2023] [Accepted: 12/10/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND Docetaxel is a yew compound antitumor agent with accurate antitumor efficacy, but its application is limited due to the high and serious adverse effects, and finding effective combination therapy options is a viable strategy. Immune checkpoint inhibitors have become hotspots in enhancing anti-tumor immunity by blocking immune checkpoint signaling pathways, but their response rate to monotherapy use is not high and the efficacy is minimal. OBJECTIVE To explore the anti-tumor effects and mechanisms of the combination of PD-1 inhibitors and Docetaxel through in vivo experiments and develop a feasible combination treatment for the therapy of prostate cancer. METHODS Tumor-bearing mice were subcutaneously injected with 0.1 ml RM-1 cells. Treatment were taken when the tumor growed up to 3 mm, after which the tumor and spleen were removed to test the antitumor effect with Flow cytometric (FACS) analysis, Immunohistochemistry, Western Blot. RESULTS In this experiment, we found that PD-1 inhibitors combined with Docetaxel had a synergistic effect on mouse prostate cancer, inhibited the growth of prostate cancer, improved survival and reduced adverse reactions, increased spleen and tumor infiltrative CD4+ and CD8+ T cells, especially in group combination with low-dose Docetaxel, and were related to the PI3K/AKT/NFKB-P65/PD-L1 signaling pathway. CONCLUSION Our study confirms that PD-1 inhibitors in combination with Docetaxel are a viable combination strategy and provide a safe and effective combination option for the clinical treatment of prostate cancer.
Collapse
Affiliation(s)
- Sixu Zhou
- College of Chemistry and Biological Engineering, Yichun University, Yichun, Jiangxi, China
| | - Baogui Wang
- College of Chemistry and Biological Engineering, Yichun University, Yichun, Jiangxi, China
| | - Yingying Wei
- College of Chemistry and Biological Engineering, Yichun University, Yichun, Jiangxi, China
| | - Peiru Dai
- College of Chemistry and Biological Engineering, Yichun University, Yichun, Jiangxi, China
| | - Yan Chen
- College of Chemistry and Biological Engineering, Yichun University, Yichun, Jiangxi, China
| | - Yingyi Xiao
- College of Chemistry and Biological Engineering, Yichun University, Yichun, Jiangxi, China
| | - Hongmei Xia
- The People’s Hospital of Yichun Affiliated to Clinical Medicine School of Yichun University in Jiangxi Province, Yichun, Jiangxi, China
| | - Chunlin Chen
- College of Chemistry and Biological Engineering, Yichun University, Yichun, Jiangxi, China
| | - Weihua Yin
- The People’s Hospital of Yichun Affiliated to Clinical Medicine School of Yichun University in Jiangxi Province, Yichun, Jiangxi, China
| |
Collapse
|
10
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
11
|
Katz MHG, Petroni GR, Bauer T, Reilley MJ, Wolpin BM, Stucky CC, Bekaii-Saab TS, Elias R, Merchant N, Dias Costa A, Lenehan P, Cardot-Ruffino V, Rodig S, Pfaff K, Dougan SK, Nowak JA, Varadhachary GR, Slingluff CL, Rahma O. Multicenter randomized controlled trial of neoadjuvant chemoradiotherapy alone or in combination with pembrolizumab in patients with resectable or borderline resectable pancreatic adenocarcinoma. J Immunother Cancer 2023; 11:e007586. [PMID: 38040420 PMCID: PMC10693876 DOI: 10.1136/jitc-2023-007586] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a challenging target for immunotherapy because it has an immunosuppressive tumor microenvironment. Neoadjuvant chemoradiotherapy can increase tumor-infiltrating lymphocyte (TIL) density, which may predict overall survival (OS). We hypothesized that adding programmed cell death protein 1 (PD-1) blockade to chemoradiotherapy would be well tolerated and increase TILs among patients with localized PDAC. METHODS Patients were randomized 2:1 to Arm A (receiving pembrolizumab plus chemoradiotherapy (capecitabine and external beam radiation)) or Arm B (receiving chemoradiotherapy alone) before anticipated pancreatectomy. Primary endpoints were (1) incidence and severity of adverse events during neoadjuvant therapy and (2) density of TILs in resected tumor specimens. TIL density was assessed using multiplexed immunofluorescence histologic examination. RESULTS Thirty-seven patients were randomized to Arms A (n=24) and B (n=13). Grade ≥3 adverse events related to neoadjuvant treatment were experienced by 9 (38%) and 4 (31%) patients in Arms A and B, respectively, with one patient experiencing dose-limiting toxicity in Arm A. Seventeen (71%) and 7 (54%) patients in Arms A and B, respectively, underwent pancreatectomy. Median CD8+ T-cell densities in Arms A and B were 67.4 (IQR: 39.2-141.8) and 37.9 (IQR: 22.9-173.4) cells/mm2, respectively. Arms showed no noticeable differences in density of CD8+Ki67+, CD4+, or CD4+FOXP3+ regulatory T cells; M1-like and M2-like macrophages; or granulocytes. Median OS durations were 27.8 (95% CI: 17.1 to NR) and 24.3 (95% CI: 12.6 to NR) months for Arms A and B, respectively. CONCLUSIONS Adding pembrolizumab to neoadjuvant chemoradiotherapy was safe. However, no convincing effect on CD8+ TILs was observed.
Collapse
Affiliation(s)
- Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gina R Petroni
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Todd Bauer
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Matthew J Reilley
- Division of Hematology and Oncology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Tanios S Bekaii-Saab
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clin, Phoenix, Arizona, USA
| | - Rawad Elias
- Hartford HealthCare Cancer Institute, Plainville, Connecticut, USA
| | - Nipun Merchant
- Department of Surgery, University of Miami, Coral Gables, Florida, USA
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Lenehan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Victoire Cardot-Ruffino
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Scott Rodig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Andrew Nowak
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Gauri R Varadhachary
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Craig L Slingluff
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Osama Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Huang S, Xiong C, Tan K. Neoadjuvant PD-1/PD-L1 axis blockade for patients with head and neck squamous cell carcinoma. Am J Otolaryngol 2023; 44:103985. [PMID: 37442083 DOI: 10.1016/j.amjoto.2023.103985] [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: 04/25/2023] [Revised: 07/04/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common type of cancer, and approximately 64 % are in a locally advanced stage at diagnosis. Therefore, neoadjuvant therapy is of great importance. However, traditional neoadjuvant strategies for HNSCC have shown limited efficacy and high complications. And it is urgent to explore new neoadjuvant approaches. With the breakthrough progress of PD-1/PD-L1 axis blockade in recurrent/metastatic HNSCC, neoadjuvant PD-1/PD-L1 axis blockade is gradually showing positive prospects for HNSCC. This study found that the combination of PD-1/PD-L1 axis blockade and chemotherapy or radiotherapy are potential with the overall response rate (ORR) of 45.0 %-96.7 % and 47.6 %-56.7 %, the pathological complete response (pCR) of 16.7 %-42.3 % and 33.3 %-100.0 %, and the main pathological response (MPR) of 26.9 %-74.1 % and 60.0 %-100.0 %, respectively. But the combination of PD-1/PD-L1 axis blockade and CTLA-4 blockade is worth questioning. And we also found pCR and MPR can be early indicators for long-term prognosis and provide five directions for neoadjuvant PD-1/PD-L1 axis blockade in the future.
Collapse
Affiliation(s)
- Shuang Huang
- Department of Stomatology, The Affiliated Shapingba Hospital of Chongqing University, Chongqing, China.
| | - Chuang Xiong
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Kui Tan
- Department of Stomatology, Chongqing University Jiangjin Hospital, Chongqing, China
| |
Collapse
|
13
|
Lin Y, Liang HW, Liu Y, Pan XB. Nivolumab adjuvant therapy for esophageal cancer: a review based on subgroup analysis of CheckMate 577 trial. Front Immunol 2023; 14:1264912. [PMID: 37860010 PMCID: PMC10582756 DOI: 10.3389/fimmu.2023.1264912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Esophageal cancer is the sixth most common cancer worldwide. Approximately 50% of patients have locally advanced disease. The CROSS and NEOCRTEC5010 trials have demonstrated that neoadjuvant chemoradiotherapy followed by surgery is the standard treatment for patients with resectable disease. However, a pathological complete response is frequently not achieved, and most patients have a poor prognosis. The CheckMate 577 trial demonstrates that nivolumab adjuvant therapy improves disease-free survival in patents without a pathological complete response. However, there are still numerous clinical questions of concern that remain controversial based on the results of the subgroup analysis. In this review, we aim to offer constructive suggestions addressing the clinical concerns raised in the CheckMate 577 trial.
Collapse
Affiliation(s)
- Yan Lin
- Department of Gastroenterology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Huan-Wei Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yang Liu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xin-Bin Pan
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| |
Collapse
|
14
|
Fan Y, Zhan M, Liang J, Yang X, Zhang B, Shi X, Hu Y. Programming Injectable DNA Hydrogels Yields Tumor Microenvironment-Activatable and Immune-Instructive Depots for Augmented Chemo-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302119. [PMID: 37541435 PMCID: PMC10582419 DOI: 10.1002/advs.202302119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/05/2023] [Indexed: 08/06/2023]
Abstract
Injectable hydrogels have attracted increasing attention for promoting systemic antitumor immune response through the co-delivery of chemotherapeutics and immunomodulators. However, the biosafety and bioactivity of conventional hydrogel depots are often impaired by insufficient possibilities for post-gelling injection and means for biofunction integration. Here, an unprecedented injectable stimuli-responsive immunomodulatory depot through programming a super-soft DNA hydrogel adjuvant is reported. This hydrogel system encoded with adenosine triphosphate aptamers can be intratumorally injected in a gel formulation and then undergoes significant molecular conformation change to stimulate the distinct release kinetics of co-encapsulated therapeutics. In this scenario, doxorubicin is first released to induce immunogenic cell death that intimately works together with the polymerized cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN) in gel scaffold for effectively recruiting and activating dendritic cells. The polymerized CpG ODN not only enhances tumor immunogenicity but minimizes free CpG-induced splenomegaly. Furthermore, the subsequently released anti-programmed cell death protein ligand 1 (aPDL1) blocks the corresponding immune inhibitory checkpoint molecule on tumor cells to sensitize antitumor T-cell immunity. This work thus contributes to the first proof-of-concept demonstration of a programmable super-soft DNA hydrogel system that perfectly matches the synergistic therapeutic modalities based on chemotherapeutic toxicity, in situ vaccination, and immune checkpoint blockade.
Collapse
Affiliation(s)
- Yu Fan
- Department of Polymeric MaterialsSchool of Materials Science and EngineeringTongji UniversityShanghai201804P. R. China
| | - Mengsi Zhan
- College of Biological Science and Medical EngineeringDonghua UniversityShanghai201620P. R. China
| | - Junhao Liang
- Department of Polymeric MaterialsSchool of Materials Science and EngineeringTongji UniversityShanghai201804P. R. China
| | - Xingsen Yang
- Department of Polymeric MaterialsSchool of Materials Science and EngineeringTongji UniversityShanghai201804P. R. China
| | - Beibei Zhang
- Department of Polymeric MaterialsSchool of Materials Science and EngineeringTongji UniversityShanghai201804P. R. China
| | - Xiangyang Shi
- College of Biological Science and Medical EngineeringDonghua UniversityShanghai201620P. R. China
| | - Yong Hu
- Department of Polymeric MaterialsSchool of Materials Science and EngineeringTongji UniversityShanghai201804P. R. China
| |
Collapse
|
15
|
Moore AM, Nooruddin Z, Reveles KR, Datta P, Whitehead JM, Franklin K, Alkadimi M, Williams MH, Williams RA, Smith S, Reichelderfer R, Cotarla I, Brannman L, Frankart A, Mulrooney T, Hsieh K, Simmons DJ, Jones X, Frei CR. Durvalumab Treatment Patterns for Patients with Unresectable Stage III Non-Small Cell Lung Cancer in the Veterans Health Administration (VHA): A Nationwide, Real-World Study. Curr Oncol 2023; 30:8411-8423. [PMID: 37754526 PMCID: PMC10529719 DOI: 10.3390/curroncol30090611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Durvalumab is approved for the treatment of adults with unresectable stage III non-small cell lung cancer (NSCLC) post-chemoradiotherapy (CRT). This real-world study describes patient characteristics and durvalumab treatment patterns (number of doses and therapy duration; treatment initiation delays, interruptions, discontinuations, and associated reasons) among VHA-treated patients. METHODS This was a retrospective cohort study of adults with unresectable stage III NSCLC receiving durvalumab at the VHA between 1 January 2017 and 30 June 2020. Patient characteristics and treatment patterns were presented descriptively. RESULTS A total of 935 patients were included (median age: 69 years; 95% males; 21% Blacks; 46% current smokers; 16% ECOG performance scores ≥ 2; 50% squamous histology). Durvalumab initiation was delayed in 39% of patients (n = 367). Among the 200 patients with recorded reasons, delays were mainly due to physician preference (20%) and CRT toxicity (11%). Overall, patients received a median (interquartile range) of 16 (7-24) doses of durvalumab over 9.0 (2.9-11.8) months. Treatment interruptions were experienced by 19% of patients (n = 180), with toxicity (7.8%) and social reasons (2.6%) being the most cited reasons. Early discontinuation occurred in 59% of patients (n = 551), largely due to disease progression (24.2%) and toxicity (18.2%). CONCLUSIONS These real-world analyses corroborate PACIFIC study results in terms of the main reasons for treatment discontinuation in a VHA population with worse prognostic factors, including older age, predominantly male sex, and poorer performance score. One of the main reasons for durvalumab initiation delays, treatment interruptions, or discontinuations was due to toxicities. Patients could benefit from improved strategies to prevent, identify, and manage CRT and durvalumab toxicities timely and effectively.
Collapse
Affiliation(s)
- Amanda M. Moore
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (A.M.M.); (K.R.R.); (X.J.)
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
| | - Zohra Nooruddin
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Kelly R. Reveles
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (A.M.M.); (K.R.R.); (X.J.)
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Paromita Datta
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Jennifer M. Whitehead
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Kathleen Franklin
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Munaf Alkadimi
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | | | - Ryan A. Williams
- MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.W.); (R.A.W.)
| | - Sarah Smith
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Renee Reichelderfer
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Ion Cotarla
- AstraZeneca US Medical Affairs, Gaithersburg, MD 20878, USA; (I.C.); (T.M.); (K.H.); (D.J.S.)
| | - Lance Brannman
- College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA;
| | - Andrew Frankart
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Tiernan Mulrooney
- AstraZeneca US Medical Affairs, Gaithersburg, MD 20878, USA; (I.C.); (T.M.); (K.H.); (D.J.S.)
| | - Kristin Hsieh
- AstraZeneca US Medical Affairs, Gaithersburg, MD 20878, USA; (I.C.); (T.M.); (K.H.); (D.J.S.)
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel J. Simmons
- AstraZeneca US Medical Affairs, Gaithersburg, MD 20878, USA; (I.C.); (T.M.); (K.H.); (D.J.S.)
| | - Xavier Jones
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (A.M.M.); (K.R.R.); (X.J.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| | - Christopher R. Frei
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (A.M.M.); (K.R.R.); (X.J.)
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (Z.N.); (P.D.); (J.M.W.); (K.F.); (M.A.); (S.S.)
- Audie L. Murphy Veterans Hospital, South Texas Veterans Health Care System, San Antonio, TX 78229, USA;
| |
Collapse
|
16
|
Loh J, Low JL, Sachdeva M, Low PQ, Wong RSJ, Huang Y, Chia PL, Soo RA. Management of Oncogene Driven Locally Advanced Unresectable Non-small Cell Lung Cancer. Expert Rev Anticancer Ther 2023; 23:913-926. [PMID: 37551698 DOI: 10.1080/14737140.2023.2245140] [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: 12/11/2022] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION The current standard of care of locally advanced non-small cell lung cancer (LA-NSCLC) is concurrent chemoradiation, followed by consolidation durvalumab. However, there is evidence that the efficacy of chemoradiation and also immunotherapy in many oncogene-positive LA-NSCLC are attenuated, and dependent on the subgroup. AREAS COVERED We will firstly review the outcomes of standard-of-care therapy in oncogene-driven LA-NSCLC. We looked at various oncogene driven subgroups and the tumor microenvironment that may explain differential response. Finally, we review the role of targeted therapy in the treatment of LA-NSCLC. EXPERT OPINION Each oncogene-positive subgroup should be treated as its own entity, and continued efforts should be undertaken to incorporate targeted therapy, which is likely to yield superior survival outcomes if trial design can be optimized and toxicities can be managed.
Collapse
Affiliation(s)
- Jerold Loh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Jia Li Low
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Manavi Sachdeva
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Peter Qj Low
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Rachel Su Jen Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Yiqing Huang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Puey Ling Chia
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| |
Collapse
|
17
|
Wang J, Hu F, Yu P, Wang J, Liu Z, Bao Q, Zhang W, Wen J. Sorafenib inhibits doxorubicin-induced PD-L1 upregulation to improve immunosuppressive microenvironment in Osteosarcoma. J Cancer Res Clin Oncol 2023; 149:5127-5138. [PMID: 36348018 DOI: 10.1007/s00432-022-04458-4] [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: 08/19/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE Although undergoing conventional chemotherapy significantly improves the prognosis of Osteosarcoma, chemoresistance and failure of therapy is still a significant challenge. Furthermore, conventional chemotherapy, like doxorubicin, would upregulate the expression of programmed death-ligand 1 (PD-L1) which caused an immunosuppressive microenvironment and unsatisfied treatment result in Osteosarcoma. Thus, it is urgent to explore a strategy to overcome this disadvantage. METHODS Human Osteosarcoma cell line MG63 and mouse Osteosarcoma cell line K7 were included in this study. Subcutaneous tumor model was used by injection of K7 cells in BALB/C mice to test the effect of doxorubicin and sorafenib on tumor growth. PD-L1 expression was tested in vitro (flow cytometry, western blot and PCR) and in vivo (flow cytometry and immunohistochemistry). Proportion of immune cells (CD4, CD8, Tregs, and cytotoxic T lymphocytes) in vivo was analyzed with flow cytometry. RESULTS Combination of sorafenib and doxorubicin inhibited tumor growth significantly in vivo. Doxorubicin increased PD-L1 expression in vitro and in vivo, while sorafenib inhibited doxorubicin-induced PD-L1 upregulation in vitro and in vivo. Proportion of interferon-γ-secreting CD8 + T lymphocytes in tumor tissue was increased significantly when sorafenib was combined with doxorubicin, while proportion of CD4, CD8, and Tregs was not significantly changed. Extracellular signal-regulated kinases (ERK) pathway could be one of the key mechanisms by which doxorubicin induced upregulation of PD-L1 in Osteosarcoma cells. CONCLUSION Combination of sorafenib and conventional chemotherapeutic reagents is a potent strategy to improve treatment effectiveness by modulating tumor microenvironment in Osteosarcoma through increasing proportion of cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Jizhuang Wang
- Department of Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangqiong Hu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, China
| | - Pei Yu
- Department of Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, China
| | - Zhuochao Liu
- Department of Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiyuan Bao
- Department of Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weibin Zhang
- Department of Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junxiang Wen
- Department of Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Arcovito G, Palomba A, Gallo O, Franchi A. The Histological Background of Recurrence in Laryngeal Squamous Cell Carcinoma: An Insight into the Modifications of Tumor Microenvironment. Cancers (Basel) 2023; 15:3259. [PMID: 37370868 DOI: 10.3390/cancers15123259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/04/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Recurrent laryngeal carcinoma presents differences from the primary tumor that largely depend on the treatment. In this article, we review the histologic and molecular treatment-induced changes that may affect the diagnosis of recurrent laryngeal carcinoma, the assessment of predictive markers, and the response to treatment with immune checkpoint inhibitors. Radiotherapy induces profound modifications that are strictly related to necrosis of different tissue components, fibrosis, and damage of the tumor vessels. Postradiotherapy recurrent/persistent laryngeal squamous cell carcinoma typically presents a discohesive growth pattern within a fibrotic background associated with significant changes of the tumor immune microenvironment, with both important immunosuppressive and immunostimulatory effects. Overall, the increase of immunoregulatory cells and immune checkpoints such as CTLA-4, TIM-3, PD-1, and PD-L1 induced by radiotherapy and chemotherapy strongly supports the use of immune checkpoint inhibitors in recurrent/persistent laryngeal carcinoma. Future studies aiming to identify predictive factors of the response to immune checkpoint inhibitors should consider such treatment-induced modifications.
Collapse
Affiliation(s)
- Giorgia Arcovito
- Section of Pathology, Department of Translational Research, University of Pisa, 56126 Pisa, Italy
| | - Annarita Palomba
- Unit of Histopathology and Molecular Diagnostic, Azienda Ospedaliera Universitaria Careggi, 50139 Florence, Italy
| | - Oreste Gallo
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
| | - Alessandro Franchi
- Section of Pathology, Department of Translational Research, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
19
|
Zhang L, Wang J, Cui H, Zheng H, Yin X, Lin J, Wang Y, Zhao Y, Li H, Chen Q. Simultaneous Knockdown of Immune Suppressive Markers by Tumor Microenvironment-Responsive Multifaceted Prodrug Nanomedicine. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12864-12881. [PMID: 36856003 DOI: 10.1021/acsami.3c00986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Tumors managing to exempt from immune clearance are attributable to their overexpressed immune suppressive molecules (CD47, PD-L1, etc.). Leadingly, the checkpoint blockade-based chemoimmunotherapy by means of knockdown of these immunosuppressive checkpoints, together with immunogenetic chemotherapeutics, is perceived to be a valid therapeutic strategy for improving anti-tumor outcomes. Herein, chemotherapeutic camptothecin was covalently introduced into an intriguing multifaceted nanomedicine. Note that the elaborated nanomedicine was chemically engineered to enable targeted transportation to the tumors via systemic administration, possessing intelligent responsiveness to sequential extracellular and intracellular microenvironments in the targeted tumors for prompted transcellular endocytosis owing to enzymolysis by the tumor-enriched matrix metalloproteinases and the selective liberation of cytocidal camptothecin in the cell interiors owing to thiolysis by glutathione. In addition, this chemotherapeutic nanomedicine allowed facile encapsulation of the negatively charged RNA interference payloads. Consequently, aiming for treatment of intractable triple-negative breast tumors, we attempted the small interfering RNA (siRNA) payloads aiming for CD47 and PD-L1 into the aforementioned nanomedicine. The subsequent investigations demonstrated drastic knockdown of these vital immune suppressive checkpoints by this siRNA-encapsulating chemotherapeutic nanomedicine, conducing to the reversal of the immune checkpoint suppressive microenvironment of triple-negative 4T1 tumors. Namely, the inhibited proceedings of the innate and adaptive anti-tumor immunities were revived, as supported by observation of the activated infiltration and retention of CD68+ macrophages and CD4+ and CD8+ lymphocytes into the tumors. Eventually, most potent anti-tumor efficacies were accomplished by systemic administration of this chemoimmunotherapeutic nanomedicine, which verified the amplified contribution from anti-tumor immunities by means of knockdown of the immune suppressive molecules to the ultimate anti-tumor efficacies. Note that the upregulation of the immune suppressive molecules was constantly reported in a variety of clinical therapies; hence, our facile chemoimmunotherapeutic platform should be emphasized in clinical translation for seeking improved therapeutic outcomes.
Collapse
Affiliation(s)
- Liuwei Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
| | - Hongyan Cui
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
| | - Haonan Zheng
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
| | - Xiaolan Yin
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
| | - Jiaqi Lin
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
| | - Yue Wang
- Department of Gastric Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning 110042, China
| | - Yan Zhao
- Department of Gastric Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning 110042, China
| | - Haidong Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
| | - Qixian Chen
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, P. R. China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning 110042, China
| |
Collapse
|
20
|
Kciuk M, Kołat D, Kałuzińska-Kołat Ż, Gawrysiak M, Drozda R, Celik I, Kontek R. PD-1/PD-L1 and DNA Damage Response in Cancer. Cells 2023; 12:530. [PMID: 36831197 PMCID: PMC9954559 DOI: 10.3390/cells12040530] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
The application of immunotherapy for cancer treatment is rapidly becoming more widespread. Immunotherapeutic agents are frequently combined with various types of treatments to obtain a more durable antitumor clinical response in patients who have developed resistance to monotherapy. Chemotherapeutic drugs that induce DNA damage and trigger DNA damage response (DDR) frequently induce an increase in the expression of the programmed death ligand-1 (PD-L1) that can be employed by cancer cells to avoid immune surveillance. PD-L1 exposed on cancer cells can in turn be targeted to re-establish the immune-reactive tumor microenvironment, which ultimately increases the tumor's susceptibility to combined therapies. Here we review the recent advances in how the DDR regulates PD-L1 expression and point out the effect of etoposide, irinotecan, and platinum compounds on the anti-tumor immune response.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Damian Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Mateusz Gawrysiak
- Department of Immunology and Allergy, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, 91-347 Lodz, Poland
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| |
Collapse
|
21
|
Aslan V, Yazici O, Özdemir N. Increased Tumor Mutation Burden Levels and Sensitivity of Non-Small Cell Lung Cancer to PD-L1 Blockade. JAMA Oncol 2023; 9:570. [PMID: 36729466 DOI: 10.1001/jamaoncol.2022.7586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Volkan Aslan
- Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Ozan Yazici
- Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Nuriye Özdemir
- Department of Medical Oncology, Gazi University, Ankara, Turkey
| |
Collapse
|
22
|
Sarkar M, Nguyen T, Gundre E, Ogunlusi O, El-Sobky M, Giri B, Sarkar TR. Cancer-associated fibroblasts: The chief architect in the tumor microenvironment. Front Cell Dev Biol 2023; 11:1089068. [PMID: 36793444 PMCID: PMC9923123 DOI: 10.3389/fcell.2023.1089068] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Stromal heterogeneity of tumor microenvironment (TME) plays a crucial role in malignancy and therapeutic resistance. Cancer-associated fibroblasts (CAFs) are one of the major players in tumor stroma. The heterogeneous sources of origin and subsequent impacts of crosstalk with breast cancer cells flaunt serious challenges before current therapies to cure triple-negative breast cancer (TNBC) and other cancers. The positive and reciprocal feedback of CAFs to induce cancer cells dictates their mutual synergy in establishing malignancy. Their substantial role in creating a tumor-promoting niche has reduced the efficacy of several anti-cancer treatments, including radiation, chemotherapy, immunotherapy, and endocrine therapy. Over the years, there has been an emphasis on understanding CAF-induced therapeutic resistance in order to enhance cancer therapy results. CAFs, in the majority of cases, employ crosstalk, stromal management, and other strategies to generate resilience in surrounding tumor cells. This emphasizes the significance of developing novel strategies that target particular tumor-promoting CAF subpopulations, which will improve treatment sensitivity and impede tumor growth. In this review, we discuss the current understanding of the origin and heterogeneity of CAFs, their role in tumor progression, and altering the tumor response to therapeutic agents in breast cancer. In addition, we also discuss the potential and possible approaches for CAF-mediated therapies.
Collapse
Affiliation(s)
- Mrinmoy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Tristan Nguyen
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Esheksha Gundre
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Olajumoke Ogunlusi
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Mohanad El-Sobky
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, English Bazar, India
| | - Tapasree Roy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
23
|
Hassan M, Trung V, Bedi D, Shaddox S, Gunturu D, Yates C, Datta P, Samuel T. Interference with pathways activated by topoisomerase inhibition alters the surface expression of PD-L1 and MHC I in colon cancer cells. Oncol Lett 2022; 25:41. [PMID: 36589674 PMCID: PMC9773314 DOI: 10.3892/ol.2022.13628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/25/2022] [Indexed: 12/13/2022] Open
Abstract
Topoisomerase inhibitors are clinically used to treat various cancer types, including colorectal cancer. These drugs also activate signaling pathways that modulate cell survival and immune cell functions. Immunotherapy is promising for certain tumors, including microsatellite instable colorectal cancer, but not for microsatellite stable colorectal cancer. The reasons for this lack of responsiveness are largely unknown. Understanding how colorectal cancer cell-surface proteins interact with tumor-resident immune cells may offer an opportunity to identify molecules that, if targeted, may render tumor cells visible to immune cells. The present study used flow cytometry, fluorescent staining and immunoblotting to examine if inhibition of pathways activated by topoisomerase-targeting drugs may modulate the outcomes of treatment through effects on cell cycle arrest and apoptosis, and by altering surface expression levels of programmed death-ligand 1 (PD-L1) or major histocompatibility complex protein I (MHC I). Inhibition of either NF-κB or DNA-damage response (DDR) potently enhanced cell death in combination with topoisomerase inhibition, while only NF-κB inhibition increased MHC I. PD-L1 upregulation was moderately affected by NF-κB or DDR inhibitors, while both topoisomerase inhibitors and DNA damaging agents may enhance the surface expression of MHC I molecules on colon cancer cells. Such enhanced expression of MHC I may be suppressed by inhibitors of ataxia-telangiectasia mutated or checkpoint kinase kinases. Additionally, adaptive tolerance to topoisomerase inhibition caused altered cell cycle response, and reduced the expression levels of both PD-L1 and MHC I on both microsatellite instable and stable colon cancer cell lines. Therefore, targeted modulation of DDR pathways, PD-L1, MHC I or other immune regulators in colon cancer cells may make them more visible to immune cells and enable rational combination of conventional therapy with immunotherapy.
Collapse
Affiliation(s)
- Mohamed Hassan
- Tuskegee University College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL 36088, USA
| | - Vu Trung
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Deepa Bedi
- Tuskegee University College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL 36088, USA
| | - Sage Shaddox
- Tuskegee University College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL 36088, USA
| | - Dilip Gunturu
- Tuskegee University College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL 36088, USA
| | - Clayton Yates
- Tuskegee University College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL 36088, USA
| | - Pran Datta
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA,Dr Pran Datta, Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, 1824 6th Avenue South, WTI 520C, Birmingham, AL 35294, USA, E-mail:
| | - Temesgen Samuel
- Tuskegee University College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL 36088, USA,Correspondence to: Dr Temesgen Samuel, Tuskegee University College of Veterinary Medicine and Center for Biomedical Research, 1200 W. Montgomery Road, Tuskegee, AL 36088, USA, E-mail:
| |
Collapse
|
24
|
Voronova V, Vislobokova A, Mutig K, Samsonov M, Peskov K, Sekacheva M, Materenchuk M, Bunyatyan N, Lebedeva S. Combination of immune checkpoint inhibitors with radiation therapy in cancer: A hammer breaking the wall of resistance. Front Oncol 2022; 12:1035884. [PMID: 36544712 PMCID: PMC9760959 DOI: 10.3389/fonc.2022.1035884] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
Immuno-oncology is an emerging field in the treatment of oncological diseases, that is based on recruitment of the host immune system to attack the tumor. Radiation exposure may help to unlock the potential of the immune activating agents by enhancing the antigen release and presentation, attraction of immunocompetent cells to the inflammation site, and eliminating the tumor cells by phagocytosis, thereby leading to an overall enhancement of the immune response. Numerous preclinical studies in mouse models of glioma, murine melanoma, extracranial cancer, or colorectal cancer have contributed to determination of the optimal radiotherapy fractionation, as well as the radio- and immunotherapy sequencing strategies for maximizing the antitumor activity of the treatment regimen. At the same time, efficacy of combined radio- and immunotherapy has been actively investigated in clinical trials of metastatic melanoma, non-small-cell lung cancer and renal cell carcinoma. The present review summarizes the current advancements and challenges related to the aforementioned treatment approach.
Collapse
Affiliation(s)
- Veronika Voronova
- Department of Pharmacological Modeling, M&S Decisions LLC, Moscow, Russia
| | - Anastasia Vislobokova
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kerim Mutig
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mikhail Samsonov
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kirill Peskov
- Department of Pharmacological Modeling, M&S Decisions LLC, Moscow, Russia,MID3 Research Center, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,Artificial Intelligence Research Center, STU Sirius, Sochi, Russia
| | - Marina Sekacheva
- World-Class Research Center “Digital biodesign and personalized healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Materenchuk
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Natalya Bunyatyan
- Institute of Professional Education, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,Federal State Budgetary Institution “Scientific Centre for Expert Evaluation of Medicinal Products” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Svetlana Lebedeva
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,Institute of Professional Education, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,*Correspondence: Svetlana Lebedeva,
| |
Collapse
|
25
|
Song P, Lu Z, Jiang T, Han W, Chen X, Zhao X. Chitosan coated pH/redox-responsive hyaluronic acid micelles for enhanced tumor targeted co-delivery of doxorubicin and siPD-L1. Int J Biol Macromol 2022; 222:1078-1091. [DOI: 10.1016/j.ijbiomac.2022.09.245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
|
26
|
Mulgaonkar A, Elias R, Woolford L, Guan B, Nham K, Kapur P, Christie A, Tcheuyap VT, Singla N, Bowman IA, Stevens C, Hao G, Brugarolas J, Sun X. ImmunoPET Imaging with 89Zr-Labeled Atezolizumab Enables In Vivo Evaluation of PD-L1 in Tumorgraft Models of Renal Cell Carcinoma. Clin Cancer Res 2022; 28:4907-4916. [PMID: 36074149 PMCID: PMC9669181 DOI: 10.1158/1078-0432.ccr-22-1547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/18/2022] [Accepted: 09/06/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Immune checkpoint inhibitors (ICI) targeting the programmed cell death protein 1 and its ligand (PD-1/PD-L1) have transformed the treatment paradigm for metastatic renal cell carcinoma (RCC). However, response rates to ICIs as single agents or in combination vary widely and predictive biomarkers are lacking. Possibly related to the heterogeneity and dynamic nature of PD-L1 expression, tissue-based methods have shown limited value. Immuno-positron emission tomography (immunoPET) may enable noninvasive, comprehensive, and real-time PD-L1 detection. Herein, we systematically examined the performance of immunoPET for PD-L1 detection relative to IHC in an RCC patient-derived tumorgraft (TG) platform. EXPERIMENTAL DESIGN Eight independent RCC TGs with a wide range of PD-L1 expression (0%-85%) were evaluated by immunoPET. Uptake of 89Zr-labeled atezolizumab ([89Zr]Zr-DFO-ATZ) was compared with PD-L1 expression in tumors by IHC through double-blind analyses. Clinical outcomes of ICI-treated patients whose TGs were examined were analyzed to evaluate the clinical role of immunoPET in RCC. RESULTS ImmunoPET with [89Zr]Zr-DFO-ATZ (day 6/7 postinjection) revealed a statistically significant association with PD-L1 IHC assays (P = 0.0014; correlation ρXY = 0.78). Furthermore, immunoPET can be used to assess the heterogeneous distribution of PD-L1 expression. Finally, studies in the corresponding patients (n = 4) suggest that PD-L1 signal may influence ICI responsiveness. CONCLUSIONS ImmunoPET with [89Zr]Zr-DFO-ATZ may enable a thorough and dynamic assessment of PD-L1 across sites of disease. The power of immunoPET to predict ICI response in RCC is being explored in an ongoing clinical trial (NCT04006522).
Collapse
Affiliation(s)
- Aditi Mulgaonkar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Roy Elias
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Internal Medicine, Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Layton Woolford
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Internal Medicine, Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bing Guan
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kien Nham
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alana Christie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vanina T. Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Internal Medicine, Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nirmish Singla
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - I. Alex Bowman
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Internal Medicine, Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christina Stevens
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Internal Medicine, Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guiyang Hao
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Internal Medicine, Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA,Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
27
|
Chang CH, Chang YC. Comparing the Therapeutic Efficacies of Lung Cancer: Network Meta-Analysis Approaches. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14324. [PMID: 36361201 PMCID: PMC9657796 DOI: 10.3390/ijerph192114324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND In recent years, reduction of nuclear power generation and the use of coal-fired power for filling the power supply gap might have increased the risk of lung cancer. This study aims to explore the most effective treatment for different stages of lung cancer patients. METHODS We searched databases to investigate the treatment efficacy of lung cancer. The network meta-analysis was used to explore the top three effective therapeutic strategies among all collected treatment methodologies. RESULTS A total of 124 studies were collected from 115 articles with 171,757 participants in total. The results of network meta-analyses showed that the best top three treatments: (1) in response rate, for advanced lung cancer were Targeted + Targeted, Chemo + Immuno, and Targeted + Other Therapy with cumulative probabilities 82.9, 80.8, and 69.3%, respectively; for non-advanced lung cancer were Chemoradio + Targeted, Chemoradi + Immuno, and Chemoradio + Other Therapy with cumulative probabilities 69.0, 67.8, and 60.7%, respectively; (2) in disease-free control rate, for advanced lung cancer were Targeted + Others, Chemo + Immuno, and Targeted + Targeted Therapy with cumulative probabilities 93.4, 91.5, and 59.4%, respectively; for non-advanced lung cancer were Chemo + Surgery, Chemoradio + Targeted, and Surgery Therapy with cumulative probabilities 80.1, 71.5, and 43.1%, respectively. CONCLUSION The therapeutic strategies with the best effectiveness will be different depending on the stage of lung cancer patients.
Collapse
Affiliation(s)
- Chuan-Hsin Chang
- Research Center for Chinese Herbal Medicine, Graduate Institute of Healthy Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Yue-Cune Chang
- Department of Mathematics, Tamkang University, New Taipei City 25137, Taiwan
| |
Collapse
|
28
|
Programmed death-ligand 1 expression in the immune compartment of colon carcinoma. Mod Pathol 2022; 35:1740-1748. [PMID: 35773332 DOI: 10.1038/s41379-022-01128-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/08/2022]
Abstract
Programmed cell death ligand 1 (PD-L1) on tumor cells is a significant prognostic biomarker for a number of malignancies, although less is known about the significance of PD-L1 positive immune cells in colon carcinoma. The purpose of this study is to evaluate the role of PD-L1 in a large cohort of colon carcinomas to identify patterns of PD-L1 expression in the tumor microenvironment and its correlation with other key immune subsets to better understand the impact of these immune cells. We assessed 1218 colon carcinomas on representative tissue microarray sections, gathered relevant clinicopathologic information, and performed immunohistochemical staining for mismatch repair proteins, CD8, CD163, LAG3, PD-L1, FoxP3, and BRAF V600E. We then performed automated quantification; manual quantification was used for PD-L1 tumor cells and immune cells. Dual PD-L1/PU.1 immunostain was also performed. The majority of PD-L1 positive cells expressed PU.1 thus representing tumor-associated macrophages. Based on the median number of PD-L1 positive immune cells (7.6/mm2), we classified tumors into two classes: (1) PD-L1 immune cell low and (2) PD-L1 immune cell high. PD-L1 immune cell high colon carcinomas showed favorable prognostic pathologic features including less frequent extramural venous invasion (p = 0.0001) and lower AJCC stage (p = 0.0001); they were also more commonly associated with deficient mismatch repair (dMMR) (p = 0.0001) and BRAF V600E reactivity. PD-LI immune cell high tumors were associated with high CD8, CD163, and FoxP3 positive cells (p = 0.0001, respectively). PD-L1 immune cell high and LAG3 high colon carcinomas were associated with improved disease-specific survival (p = 0.0001 and 0.001, respectively). PD-L1 expression on tumor cells was not associated with disease-specific survival. On multivariate analysis of chemotherapy naïve stage 2 colon carcinomas, only extramural venous invasion (p = 0.002), perineural invasion (p = 0.001) and PD-L1 immune cell expression (p = 0.032) correlated with disease-specific survival. Resected colonic carcinomas with high expression of PD-L1 and LAG3 proteins on immune cells were associated with improved prognosis in colon carcinoma. The mechanism underlying the improved prognosis of colon carcinomas bearing high numbers of immunoregulatory cells needs further investigation.
Collapse
|
29
|
Wang Z, Li B, Li S, Lin W, Wang Z, Wang S, Chen W, Shi W, Chen T, Zhou H, Yinwang E, Zhang W, Mou H, Chai X, Zhang J, Lu Z, Ye Z. Metabolic control of CD47 expression through LAT2-mediated amino acid uptake promotes tumor immune evasion. Nat Commun 2022; 13:6308. [PMID: 36274066 PMCID: PMC9588779 DOI: 10.1038/s41467-022-34064-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 10/12/2022] [Indexed: 12/25/2022] Open
Abstract
Chemotherapy elicits tumor immune evasion with poorly characterized mechanisms. Here, we demonstrate that chemotherapy markedly enhances the expression levels of CD47 in osteosarcoma tissues, which are positively associated with patient mortality. We reveal that macrophages in response to chemotherapy secrete interleukin-18, which in turn upregulates expression of L-amino acid transporter 2 (LAT2) in tumor cells for substantially enhanced uptakes of leucine and glutamine, two potent stimulators of mTORC1. The increased levels of leucine and enhanced glutaminolysis activate mTORC1 and subsequent c-Myc-mediated transcription of CD47. Depletion of LAT2 or treatment of tumor cells with a LAT inhibitor downregulates CD47 with enhanced macrophage infiltration and phagocytosis of tumor cells, and sensitizes osteosarcoma to doxorubicin treatment in mice. These findings unveil a mutual regulation between macrophage and tumor cells that plays a critical role in tumor immune evasion and underscore the potential to intervene with the LAT2-mediated amino acid uptake for improving cancer therapies.
Collapse
Affiliation(s)
- Zenan Wang
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Binghao Li
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Shan Li
- grid.13402.340000 0004 1759 700XDepartment of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Wenlong Lin
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XInstitute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Zhan Wang
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Shengdong Wang
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Weida Chen
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Wei Shi
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Tao Chen
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Hao Zhou
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Eloy Yinwang
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Wenkan Zhang
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Haochen Mou
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Xupeng Chai
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Jiahao Zhang
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| | - Zhimin Lu
- grid.13402.340000 0004 1759 700XDepartment of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XZhejiang University Cancer Center, Hangzhou, Zhejiang China
| | - Zhaoming Ye
- grid.13402.340000 0004 1759 700XDepartment of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China ,grid.13402.340000 0004 1759 700XOrthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang China ,grid.412465.0Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang China
| |
Collapse
|
30
|
Zhang S, Sui M, Zhang Z, Su Y. Brusatol From Brucea javanica Suppresses Arsenic Trioxide-Induced PD-L1 Upregulation Through Inhibition of NRF2 in Leukemia Cells. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221132699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Overexpression of programed death-ligand 1 (PD-L1) is associated with poor prognosis in leukemia. Moreover, antitumor pharmaceuticals have been shown to induce immunoresistance, leading to reduced efficacy. Previous studies have indicated that arsenic trioxide (ATO) promotes immune evasion by inducing PD-L1 expression in solid tumors; however, little is known about its role in leukemia. A proportion of patients with acute promyelocytic leukemia were resistant to ATO therapy. Thus, this study aimed to investigate the effect of ATO on the expression of PD-L1 in leukemia cells and the underlying mechanism mediated through the nuclear factor erythroid 2 related factor (NRF2) protein. Brusatol, extracted from Brucea javanica, was selected as a unique NRF2 inhibitor, and we evaluated the possibility of using a regimen combining ATO/Brusatol in leukemia therapy. Promyelocytic NB4 and lymphocytic Jurkat cells were treated with ATO and brusatol either alone or in combination. We found that ATO significantly upregulated the expression of PD-L1 in NB4 and Jurkat cells at both the protein and mRNA levels compared with its expression in the untreated cell group. Mechanistically, ATO increased nuclear NRF2 expression and the extent of NRF2 binding to the PD-L1 promoter. Pharmacological inhibition of NRF2 by brusatol significantly blocked this effect, thereby reducing ATO-induced PD-L1 expression. In addition, the combination of brusatol and ATO showed stronger cytotoxicity than ATO alone indicated by cell counting kit-8 assay. Therefore, brusatol may further enhance the antileukemia effect of ATO not only by inhibiting ATO-induced PD-L1 expression but also by enhancing ATO-induced cytotoxicity. Our study provides a rationale for the clinical application of ATO/brusatol combination therapy.
Collapse
Affiliation(s)
- Shunji Zhang
- First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Meijuan Sui
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Zhuo Zhang
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yanhua Su
- First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
31
|
Bote H, Mesas A, Baena J, Herrera M, Paz-Ares L. Emerging immune checkpoint inhibitors for the treatment of non-small cell lung cancer. Expert Opin Emerg Drugs 2022; 27:289-300. [PMID: 36203360 DOI: 10.1080/14728214.2022.2113377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Over the last decade, immune checkpoint inhibitors (ICIs) have impacted on the standard therapy for patients with non-small cell lung cancer (NSCLC). ICIs first showed efficacy in patients with advanced disease who had progressed after chemotherapy, later reaching the first-line therapy context alone, in combination with chemotherapy, and/or with dual-immunotherapy regimens. AREAS COVERED Most of their benefit is, however, restricted to just 20% of patients due to primary or emergence of acquired resistance. In this review, we will describe the role of new emerging ICIs in the current panorama of NSCLC therapeutic approaches, not only in metastatic disease but also in locally advanced stage disease, with specific focus on those drugs under investigation in Phase 2/3 clinical trials. EXPERT OPINION Several new ICIs are now under investigation to optimize NSCLC patient management; these are usually used in combination with other well-known agents, such as 'traditional' ICIs and chemotherapy, or with other newly developed drugs. Identification of better biomarkers will provide personalized treatment approaches to overcome patient-specific immune resistance.
Collapse
Affiliation(s)
- Helena Bote
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Health Research Institute Hospital Universitario 12 de Octbure (i+12)/Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Andrés Mesas
- Department of Medical Oncology, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Javier Baena
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Health Research Institute Hospital Universitario 12 de Octbure (i+12)/Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Mercedes Herrera
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Health Research Institute Hospital Universitario 12 de Octbure (i+12)/Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Health Research Institute Hospital Universitario 12 de Octbure (i+12)/Spanish National Cancer Research Center (CNIO), Madrid, Spain.,CIBERONC, Madrid, Spain.,Department of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
32
|
Remon J, Levy A, Singh P, Hendriks LEL, Aldea M, Arrieta O. Current challenges of unresectable stage III NSCLC: are we ready to break the glass ceiling of the PACIFIC trial? Ther Adv Med Oncol 2022; 14:17588359221113268. [PMID: 35923929 PMCID: PMC9340398 DOI: 10.1177/17588359221113268] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023] Open
Abstract
Consolidation anti-programmed death-ligand 1 has become a new standard of care in unresectable stage III non-small cell lung cancer (NSCLC) following chemo-radiotherapy (CTRT), based on the results of two phase III trials. Advances remain however needed, in particular to reduce the risk of distant relapse and for treatment personalization. Newer strategies are currently being tested, including consolidation with dual immune checkpoint inhibitors (ICIs), concurrent chemo-radioimmunotherapy and (chemo)-immunotherapy induction before CTRT. One randomized phase II reported better outcomes with a double ICI consolidation as compared with durvalumab alone. Three nonrandomized phase II trials also suggested that concurrent ICI-CTRT was feasible. Within this review, we summarize the current evidence, highlight ongoing trials and discuss challenges that will ideally lead to a cure for more patients with unresectable stage III NSCLC.
Collapse
Affiliation(s)
- Jordi Remon
- Department of Medical Oncology, Centro Integral
Oncológico Clara Campal (HM-CIOCC), Hospital HM Nou Delfos, HM Hospitales,
Avinguda de Vallcarca, 151, Barcelona 08023, Spain
| | - Antonin Levy
- Department of Radiation Oncology, International
Center for Thoracic Cancers (CICT), Université Paris-Saclay, Gustave Roussy,
Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine,
Le Kremlin-Bicêtre, France
| | - Pawan Singh
- Department of Pulmonary and Critical Care
Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical
Sciences, Rohtak, Haryana, India
| | - Lizza E. L. Hendriks
- Department of Pulmonary Diseases, GROW–School
for Oncology and Reproduction, Maastricht University Medical Center,
Maastricht, The Netherlands
| | - Mihaela Aldea
- Department of Medical Oncology, International
Center for Thoracic Cancers (CICT), Université Paris-Saclay, Gustave Roussy,
Gustave Roussy, Villejuif, France
| | - Oscar Arrieta
- Thoracic Oncology Unit and Laboratory of
Personalized Medicine, Instituto Nacional de Cancerología, Mexico City,
Mexico
| |
Collapse
|
33
|
Choi YJ, Cho YL, Chae JR, Park JY, Cho H, Kang WJ. In vivo positron emission tomography imaging for PD-L1 expression in cancer using aptamer. Biochem Biophys Res Commun 2022; 620:105-112. [DOI: 10.1016/j.bbrc.2022.06.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/19/2022] [Indexed: 11/24/2022]
|
34
|
Davern M, Donlon NE, O' Connell F, Sheppard AD, Hayes C, King R, Temperley H, Butler C, Bhardwaj A, Moore J, Bracken-Clarke D, Donohoe C, Ravi N, Reynolds JV, Maher SG, Conroy MJ, Lysaght J. Cooperation between chemotherapy and immune checkpoint blockade to enhance anti-tumour T cell immunity in oesophageal adenocarcinoma. Transl Oncol 2022; 20:101406. [PMID: 35366537 PMCID: PMC8976141 DOI: 10.1016/j.tranon.2022.101406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/15/2022] [Accepted: 03/20/2022] [Indexed: 10/27/2022] Open
Abstract
Response rates to immune checkpoint blockade (ICB) remain low in oesophageal adenocarcinoma (OAC). Combining ICB with immunostimulatory chemotherapies to boost response rates is an attractive approach for converting 'cold' tumours into 'hot' tumours. This study profiled immune checkpoint (IC) expression on circulating and tumour-infiltrating T cells in OAC patients and correlated these findings with clinical characteristics. The effect of first-line chemotherapy regimens (FLOT and CROSS) on anti-tumour T cell immunity was assessed to help guide design of ICB and chemotherapy combinations in the first-line setting. The ability of ICB to enhance lymphocyte-mediated cytolysis of OAC cells in the absence and presence of post-FLOT and post-CROSS chemotherapy tumour cell secretome was assessed by a CCK-8 assay. Expression of ICs on T cells positively correlated with higher grade tumours and a subsequent poor response to neoadjuvant treatment. First-line chemotherapy regimens substantially altered IC expression profiles of T cells increasing PD-1, A2aR, KLRG-1, PD-L1, PD-L2 and CD160 and decreasing TIM-3 and LAG-3. In addition, pro-inflammatory T cell cytokine profiles were enhanced by first-line chemotherapy regimens. T cell activation status was significantly altered; both chemotherapy regimens upregulated co-stimulatory markers ICOS and CD69 yet downregulated co-stimulatory marker CD27. However, ICB attenuated chemotherapy-induced downregulation of CD27 on T cells and promoted differentiation of effector memory T cells into a terminally differentiated state. Importantly, dual nivolumab-ipilimumab treatment increased lymphocyte-mediated cytolysis of OAC cells, an effect further enhanced in the presence of post-FLOT tumour cell secretome. These findings justify a rationale to administer ICBs concurrently with first-line chemotherapies.
Collapse
Affiliation(s)
- Maria Davern
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Noel E Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Fiona O' Connell
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Andrew D Sheppard
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Conall Hayes
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Ross King
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Hugo Temperley
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Christine Butler
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Anshul Bhardwaj
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Jenny Moore
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Dara Bracken-Clarke
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Claire Donohoe
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Narayanasamy Ravi
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - John V Reynolds
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Stephen G Maher
- Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Melissa J Conroy
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, St. James's Hospital campus, Dublin 8, Ireland.
| |
Collapse
|
35
|
Aslan V, Sütcüoğlu O, Özet A, Özdemir N, Yazıcı O. Abscobal Effect of Balstilimab and Zalifrelimab Combination as Second-Line Treatment for Advanced Cervical Cancer. J Clin Oncol 2022; 40:2177-2178. [PMID: 35486885 DOI: 10.1200/jco.22.00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Volkan Aslan
- Volkan Aslan, MD, Osman Sütcüoğlu, MD, Ahmet Özet, MD, Nuriye Özdemir, MD, and Ozan Yazıcı, MD, Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Osman Sütcüoğlu
- Volkan Aslan, MD, Osman Sütcüoğlu, MD, Ahmet Özet, MD, Nuriye Özdemir, MD, and Ozan Yazıcı, MD, Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Ahmet Özet
- Volkan Aslan, MD, Osman Sütcüoğlu, MD, Ahmet Özet, MD, Nuriye Özdemir, MD, and Ozan Yazıcı, MD, Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Nuriye Özdemir
- Volkan Aslan, MD, Osman Sütcüoğlu, MD, Ahmet Özet, MD, Nuriye Özdemir, MD, and Ozan Yazıcı, MD, Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Ozan Yazıcı
- Volkan Aslan, MD, Osman Sütcüoğlu, MD, Ahmet Özet, MD, Nuriye Özdemir, MD, and Ozan Yazıcı, MD, Department of Medical Oncology, Gazi University, Ankara, Turkey
| |
Collapse
|
36
|
Zhao Y, Feng H, Tian J, Li B, Wang C, Ge L, Wang JK, Yang K, Yu Q. Consolidation treatments after chemoradiotherapy in patients with locally advanced inoperable non-small cell lung cancer: a systematic review and network meta-analysis protocol. BMJ Open 2022; 12:e060900. [PMID: 35414564 PMCID: PMC9006802 DOI: 10.1136/bmjopen-2022-060900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTION Concurrent chemoradiotherapy (CCRT) is the standard of care for inoperable locally advanced non-small cell lung cancer. To further improve prognosis, the use of consolidation treatments after CCRT has been explored extensively. Although durvalumab is the only consolidation treatment recommended by national clinical practice guidelines, there have been many studies exploring the effectiveness of other agents. However, until now, no studies have compared all agents systematically, and no studies have provided evidence for the optimal combination of different CCRTs and consolidation treatments regimens. This systematic review will evaluate the comparative clinical efficacy of consolidation therapies after CCRT as well as various combinations of CCRTs and consolidation therapies. METHODS AND ANALYSIS PubMed, the Cochrane Controlled Register of Trials (CENTRAL), EMBASE and ClinicalTrials.gov will be searched for relevant information. The estimated end date for the search will be 3 February 2022. Each stage of the review, including the study section, data extraction and risk of bias and quality of evidence assessments, will be performed in duplicate. We will include randomised controlled trials that included participants who received CCRT and consolidation treatment in at least one treatment arm. The primary endpoints will be overall survival and progression-free survival. Tumour response, health-related quality of life, disease-free survival and treatment-related toxicity will be presented as secondary outcomes. Both traditional meta-analysis and network meta-analysis (NMA) with the Bayesian approach will be conducted. Subgroup analyses and meta-regression will be completed to investigate heterogeneity, and sensitivity analyses will be conducted to assess the robustness of the findings. ETHICS AND DISSEMINATION Ethical approval and patient consent are not required as this study is a meta-analysis based on published studies. The results of this study will be submitted to a peer-reviewed journal for publication. In case of any changes in the protocol, protocol amendments will be updated in PROSPERO and explanations of these modifications will be described in the final report of this review. The results of this systematic review and NMA will be published in a peer-reviewed journal. PROSPERO REGISTRATION NUMBER CRD42021239433.
Collapse
Affiliation(s)
- Ye Zhao
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Haiming Feng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
| | - Jinhui Tian
- Evidence Based Medicine Center, Lanzhou University, Lanzhou, Gansu, China
| | - Bin Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
| | - Cheng Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
| | - Long Ge
- Evidence Based Medicine Center, Lanzhou University, Lanzhou, Gansu, China
| | - Jian Kai Wang
- Department of Radiotherapy, Gansu Province People's Hospital, Lanzhou, Gansu, China
| | - Kehu Yang
- Evidence Based Medicine Center, Lanzhou University, Lanzhou, Gansu, China
| | - Qin Yu
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
37
|
Wu L, Wang Y, Li B, Wan G, Liang L, Li T, Lang J, Wang Q. Toripalimab in Combination With Induction Chemotherapy and Subsequent Chemoradiation as First-Line Treatment in Patients With Advanced/Metastatic Esophageal Carcinoma: Protocol for a Single-Arm, Prospective, Open-Label, Phase II Clinical Trial (TR-EAT). Front Oncol 2022; 12:878851. [PMID: 35463350 PMCID: PMC9024160 DOI: 10.3389/fonc.2022.878851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitor therapy combined with chemotherapy is safe and effective in treating advanced esophageal carcinoma; however, some patients still experience tumor progression and/or metastasis. Whether the addition of radiotherapy to immunotherapy combined with chemotherapy improves the prognosis of patients with advanced/metastatic esophageal carcinoma needs to be investigated. In the present study, we developed a protocol for our clinical trial indicating that toripalimab combined with induction chemotherapy followed by chemoradiotherapy can safely prolong survival in patients with stage IV esophageal carcinoma. This open-label, single-arm, phase II trial will include patients with unresectable stage IV esophageal squamous cell carcinoma who have not received prior systemic therapy. The patients will be treated with two cycles of toripalimab (240 mg, 1 day before chemotherapy, Q3W) combined with induction chemotherapy (paclitaxel, 135–175 mg/m2 + carboplatin, area under the curve = 4–6, day 1, intravenous, Q3W). Thereafter, they will undergo two cycles of the aforementioned treatment with concurrent radiotherapy (30–50 Gy in 15–25 fractions), followed by toripalimab (240 mg, day 1, Q3W) for 1 year. The primary outcome measure will be progression-free survival; the secondary outcome measures will include the objective response rate, disease control rate, duration of remission, 1- and 2-year overall survival rates, safety and tolerability, and changes in health-related quality of life. The study protocol was approved by the Ethics Committee of Sichuan Cancer Hospital (SCCHEC-02-2021-021). The trial is underway in accordance with the Declaration of Helsinki.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinyi Lang
- *Correspondence: Qifeng Wang, ; Jinyi Lang,
| | | |
Collapse
|
38
|
Zhao S, Xu B, Ma W, Chen H, Jiang C, Cai J, Meng X. DNA Damage Repair in Brain Tumor Immunotherapy. Front Immunol 2022; 12:829268. [PMID: 35095931 PMCID: PMC8792754 DOI: 10.3389/fimmu.2021.829268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 12/22/2021] [Indexed: 12/01/2022] Open
Abstract
With the gradual understanding of tumor development, many tumor therapies have been invented and applied in clinical work, and immunotherapy has been widely concerned as an emerging hot topic in the last decade. It is worth noting that immunotherapy is nowadays applied under too harsh conditions, and many tumors are defined as “cold tumors” that are not sensitive to immunotherapy, and brain tumors are typical of them. However, there is much evidence that suggests a link between DNA damage repair mechanisms and immunotherapy. This may be a breakthrough for the application of immunotherapy in brain tumors. Therefore, in this review, first, we will describe the common pathways of DNA damage repair. Second, we will focus on immunotherapy and analyze the mechanisms of DNA damage repair involved in the immune process. Third, we will review biomarkers that have been or may be used to evaluate immunotherapy for brain tumors, such as TAMs, RPA, and other molecules that may provide a precursor assessment for the rational implementation of immunotherapy for brain tumors. Finally, we will discuss the rational combination of immunotherapy with other therapeutic approaches that have an impact on the DNA damage repair process in order to open new pathways for the application of immunotherapy in brain tumors, to maximize the effect of immunotherapy on DNA damage repair mechanisms, and to provide ideas and guidance for immunotherapy in brain tumors.
Collapse
Affiliation(s)
- Shihong Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Boya Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
39
|
Zhang J, Pan S, Jian C, Hao L, Dong J, Sun Q, Jin H, Han X. Immunostimulatory Properties of Chemotherapy in Breast Cancer: From Immunogenic Modulation Mechanisms to Clinical Practice. Front Immunol 2022; 12:819405. [PMID: 35069604 PMCID: PMC8766762 DOI: 10.3389/fimmu.2021.819405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) is the most common malignancy among females. Chemotherapy drugs remain the cornerstone of treatment of BC and undergo significant shifts over the past 100 years. The advent of immunotherapy presents promising opportunities and constitutes a significant complementary to existing therapeutic strategies for BC. Chemotherapy as a cytotoxic treatment that targets proliferation malignant cells has recently been shown as an effective immune-stimulus in multiple ways. Chemotherapeutic drugs can cause the release of damage-associated molecular patterns (DAMPs) from dying tumor cells, which result in long-lasting antitumor immunity by the key process of immunogenic cell death (ICD). Furthermore, Off-target effects of chemotherapy on immune cell subsets mainly involve activation of immune effector cells including natural killer (NK) cells, dendritic cells (DCs), and cytotoxic T cells, and depletion of immunosuppressive cells including Treg cells, M2 macrophages and myeloid-derived suppressor cells (MDSCs). Current mini-review summarized recent large clinical trials regarding the combination of chemotherapy and immunotherapy in BC and addressed the molecular mechanisms of immunostimulatory properties of chemotherapy in BC. The purpose of our work was to explore the immune-stimulating effects of chemotherapy at the molecular level based on the evidence from clinical trials, which might be a rationale for combinations of chemotherapy and immunotherapy in BC.
Collapse
Affiliation(s)
- Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen Jian
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Li Hao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jie Dong
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Qingqing Sun
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
40
|
Fan Z, Wu C, Chen M, Jiang Y, Wu Y, Mao R, Fan Y. The generation of PD-L1 and PD-L2 in cancer cells: From nuclear chromatin reorganization to extracellular presentation. Acta Pharm Sin B 2022; 12:1041-1053. [PMID: 35530130 PMCID: PMC9069407 DOI: 10.1016/j.apsb.2021.09.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/27/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
The immune checkpoint blockade (ICB) targeting on PD-1/PD-L1 has shown remarkable promise in treating cancers. However, the low response rate and frequently observed severe side effects limit its broad benefits. It is partially due to less understanding of the biological regulation of PD-L1. Here, we systematically and comprehensively summarized the regulation of PD-L1 from nuclear chromatin reorganization to extracellular presentation. In PD-L1 and PD-L2 highly expressed cancer cells, a new TAD (topologically associating domain) (chr9: 5,400,000-5,600,000) around CD274 and CD273 was discovered, which includes a reported super-enhancer to drive synchronous transcription of PD-L1 and PD-L2. The re-shaped TAD allows transcription factors such as STAT3 and IRF1 recruit to PD-L1 locus in order to guide the expression of PD-L1. After transcription, the PD-L1 is tightly regulated by miRNAs and RNA-binding proteins via the long 3'UTR. At translational level, PD-L1 protein and its membrane presentation are tightly regulated by post-translational modification such as glycosylation and ubiquitination. In addition, PD-L1 can be secreted via exosome to systematically inhibit immune response. Therefore, fully dissecting the regulation of PD-L1/PD-L2 and thoroughly detecting PD-L1/PD-L2 as well as their regulatory networks will bring more insights in ICB and ICB-based combinational therapy.
Collapse
Key Words
- 3′-UTR, 3′-untranslated region
- ADAM17, a disintegrin and metalloprotease 17
- APCs, antigen-presenting cells
- AREs, adenylate and uridylate (AU)-rich elements
- ATF3, activating transcription factor 3
- CD273/274, cluster of differentiation 273/274
- CDK4, cyclin-dependent kinase 4
- CMTM6, CKLF like MARVEL transmembrane domain containing 6
- CSN5, COP9 signalosome subunit 5
- CTLs, cytotoxic T lymphocytes
- EMT, epithelial to mesenchymal transition
- EpCAM, epithelial cell adhesion molecule
- Exosome
- FACS, fluorescence-activated cell sorting
- GSDMC, Gasdermin C
- GSK3β, glycogen synthase kinase 3 beta
- HSF1, heat shock transcription factor 1
- Hi-C, high throughput chromosome conformation capture
- ICB, immune checkpoint blockade
- IFN, interferon
- IL-6, interleukin 6
- IRF1, interferon regulatory factor 1
- Immune checkpoint blockade
- JAK, Janus kinase 1
- NFκB, nuclear factor kappa B
- NSCLC, non-small cell lung cancer
- OTUB1, OTU deubiquitinase, ubiquitin aldehyde binding 1
- PARP1, poly(ADP-ribose) polymerase 1
- PD-1, programmed cell death-1
- PD-L1
- PD-L1, programmed death-ligand 1
- PD-L2
- PD-L2, programmed death ligand 2
- Post-transcriptional regulation
- Post-translational regulation
- SP1, specificity protein 1
- SPOP, speckle-type POZ protein
- STAG2, stromal antigen 2
- STAT3, signal transducer and activator of transcription 3
- T2D, type 2 diabetes
- TADs, topologically associating domains
- TFEB, transcription factor EB
- TFs, transcription factors
- TNFα, tumor necrosis factor-alpha
- TTP, tristetraprolin
- Topologically associating domain
- Transcription
- UCHL1, ubiquitin carboxy-terminal hydrolase L1
- USP22, ubiquitin specific peptidase 22
- dMMR, deficient DNA mismatch repair
- irAEs, immune related adverse events
Collapse
Affiliation(s)
- Zhiwei Fan
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Changyue Wu
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Miaomiao Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Yongying Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Yuanyuan Wu
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Yihui Fan
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
41
|
PD-1 blockade enhances chemotherapy toxicity in oesophageal adenocarcinoma. Sci Rep 2022; 12:3259. [PMID: 35228614 PMCID: PMC8885636 DOI: 10.1038/s41598-022-07228-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/01/2022] [Indexed: 12/17/2022] Open
Abstract
Chemotherapy upregulates immune checkpoint (IC) expression on the surface of tumour cells and IC-intrinsic signalling confers a survival advantage against chemotherapy in several cancer-types including oesophageal adenocarcinoma (OAC). However, the signalling pathways mediating chemotherapy-induced IC upregulation and the mechanisms employed by ICs to protect OAC cells against chemotherapy remain unknown. Longitudinal profiling revealed that FLOT-induced IC upregulation on OE33 OAC cells was sustained for up to 3 weeks post-treatment, returning to baseline upon complete tumour cell recovery. Pro-survival MEK signalling mediated FLOT-induced upregulation of PD-L1, TIM-3, LAG-3 and A2aR on OAC cells promoting a more immune-resistant phenotype. Single agent PD-1, PD-L1 and A2aR blockade decreased OAC cell viability, proliferation and mediated apoptosis. Mechanistic insights demonstrated that blockade of the PD-1 axis decreased stem-like marker ALDH and expression of DNA repair genes. Importantly, combining single agent PD-1, PD-L1 and A2aR blockade with FLOT enhanced cytotoxicity in OAC cells. These findings reveal novel mechanistic insights into the immune-independent functions of IC-intrinsic signalling in OAC cells with important clinical implications for boosting the efficacy of the first-line FLOT chemotherapy regimen in OAC in combination with ICB, to not only boost anti-tumour immunity but also to suppress IC-mediated promotion of key hallmarks of cancer that drive tumour progression.
Collapse
|
42
|
Alhesa A, Awad H, Bloukh S, Al-Balas M, El-Sadoni M, Qattan D, Azab B, Saleh T. PD-L1 expression in breast invasive ductal carcinoma with incomplete pathological response to neoadjuvant chemotherapy. Int J Immunopathol Pharmacol 2022; 36:3946320221078433. [PMID: 35225058 PMCID: PMC8891930 DOI: 10.1177/03946320221078433] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives: To investigate the expression of programmed death-ligand 1 (PD-L1) in breast cancer in association with incomplete pathological response (PR) to neoadjuvant chemotherapy (NAC). Methods PD-L1 expression was evaluated using immunohistochemistry in post-operative, post-NAC samples of 60 patients (n = 60) diagnosed with breast invasive ductal carcinoma with incomplete PR to NAC, including 31 matched pre-NAC and post-NAC samples (n = 31). PD-L1 protein expression was assessed using three scoring approaches, including the tumor proportion score (TPS), the immune cell score (ICS), and the combined tumor and immune cell score (combined positive score, CPS) with a 1% cut-off. Results In the post-operative, post-NAC samples (n = 60), positive expression rate of PD-L1 was observed in 18.3% (11/60) of cases by TPS, 31.7% (19/60) by ICS, and 25% (15/60) by CPS. In matched samples, positive expression rate of PD-L1 was observed in 19.3% (6/31) of patients by TPS, 51.6% (16/31) by ICS, and 19.3% (6/31) by CPS in pre-NAC specimens, while it was observed in 22.6% (7/31) of matched post-NAC samples by TPS, 22.6% (7/31) by ICS, and 19.3% (6/31) by CPS. In the matched samples, there was a significant decrease in PD-L1 immunoexpression using ICS in post-NAC specimens (McNemar’s, p = 0.020), while no significant differences were found using TPS and CPS between pre- and post-NAC samples (p = 1.000, p = 0.617; respectively). PD-L1 immunoexpression determined by TPS or CPS was only significantly associated with ER status (p = 0.022, p = 0.021; respectively), but not with other clinicopathological variables. We could not establish a correlation between PD-L1 expression and the overall survival rate (p > 0.05). There were no significant differences in the tumor infiltrating lymphocytes count between the paired pre- and post-NAC samples (t = 0.581, p = 0.563 or Wilcoxon’s Signed Rank test; z = -0.625, p = 0.529). Conclusion Our findings indicate that PD-L1 protein expression in infiltrating immune cells was significantly reduced in breast tumors that developed incomplete PR following the exposure to NAC.
Collapse
Affiliation(s)
- Ahmad Alhesa
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Heyam Awad
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Sarah Bloukh
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mahmoud Al-Balas
- Department of General and Specialized Surgery, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Mohammed El-Sadoni
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Duaa Qattan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Bilal Azab
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
43
|
Geum DH, Hwang DS, Lee CH, Cho SD, Jang MA, Ryu MH, Kim UK. PD-L1 Expression Correlated with Clinicopathological Factors and Akt/Stat3 Pathway in Oral SCC. Life (Basel) 2022; 12:life12020238. [PMID: 35207525 PMCID: PMC8875073 DOI: 10.3390/life12020238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 11/28/2022] Open
Abstract
Programmed cell death ligand 1 (PD-L1) is an immune checkpoint molecule that inhibits immune responses. The physiological and prognostic role of the PD-L1 signaling pathway in the oral maxillofacial region is unclear. This study aimed to investigate the role of PD-L1 in the progression of oral squamous cell carcinoma (OSCC). Furthermore, clinicopathological factors related to PD-L1 expression were examined in patients with OSCC through immunohistochemistry (IHC) of tissue sections and through an in vitro study in OSCC cells. The medical records, radiographic findings, and mortality referrals of 81 patients obtained from the National Statistical Office were reviewed. IHC was performed on tissue specimens of these patients to determine the expression levels of PD-L1, which showed significant statistical differences based on age, tumor size, TNM stage, cervical lymph node metastasis, and locoregional recurrence. Patients with a high PD-L1 expression had significantly poorer survival rates. Multivariate analysis using the Cox proportional model confirmed the high relative risk ratio for high PD-L1 expression, TNM stage, and neck node metastasis, all of which were significantly associated with a poor prognosis in patients with OSCC. The in vitro study showed that SAS and YD38 cells transfected with PD-L1 siRNA had significantly increased apoptosis, reduced proliferative capacity, and tumorigenicity.
Collapse
Affiliation(s)
- Dong-Ho Geum
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, 49, Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Korea; (D.-H.G.); (D.-S.H.)
| | - Dae-Seok Hwang
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, 49, Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Korea; (D.-H.G.); (D.-S.H.)
| | - Chang-Hun Lee
- Department of Pathology, School of Medicine, Pusan National University, 49, Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Korea;
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Korea;
| | - Min-A Jang
- Dental and Life Science Institute, Pusan National University, 49, Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Korea;
| | - Mi-Heon Ryu
- Department of Oral Pathology, Dental and Life Science Institute, School of Dentistry, Pusan National University, 49, Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Korea
- Correspondence: (M.-H.R.); (U.-K.K.); Tel.: +82-51-510-8251 (M.-H.R.); +82-55-360-5112 (U.-K.K.); Fax: +82-51-510-8249 (M.-H.R.); +82-55-360-5104 (U.-K.K.)
| | - Uk-Kyu Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, 49, Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Korea; (D.-H.G.); (D.-S.H.)
- Correspondence: (M.-H.R.); (U.-K.K.); Tel.: +82-51-510-8251 (M.-H.R.); +82-55-360-5112 (U.-K.K.); Fax: +82-51-510-8249 (M.-H.R.); +82-55-360-5104 (U.-K.K.)
| |
Collapse
|
44
|
Safety and efficacy of durvalumab with R-CHOP or R 2-CHOP in untreated, high-risk DLBCL: a phase 2, open-label trial. Int J Hematol 2021; 115:222-232. [PMID: 34797531 DOI: 10.1007/s12185-021-03241-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022]
Abstract
Patients with high-risk diffuse large B-cell lymphoma (DLBCL) have poor outcomes following first-line cyclophosphamide, doxorubicin, vincristine, prednisone, and rituximab (R-CHOP). Evidence shows chemotherapy and immune checkpoint blockade can increase antitumor efficacy. This study investigated durvalumab, a programmed death-ligand 1 inhibitor, combined with R-CHOP or lenalidomide + R-CHOP (R2-CHOP) in newly diagnosed high-risk DLBCL. Patients received durvalumab 1125 mg every 21 days for 2-8 cycles + R-CHOP (non-activated B-cell [ABC] subtype) or R2-CHOP (ABC), then durvalumab consolidation (1500 mg every 28 days). Of 46 patients, 43 received R-CHOP and three R2-CHOP. All patients had the high-risk disease; 14 (30.4%) and eight (17.4%) had double- or triple-hit DLBCL, respectively. Following induction, 20/37 (54.1%) patients receiving durvalumab + R-CHOP achieved complete response (CR), and seven (18.9%) partial response (PR); 25 (67.6% [95% CI 50.2-82.0]) continued to consolidation and were progression-free at 12 months. Among efficacy-evaluable patients with double- or triple-hit DLBCL (n = 12), five achieved CR and five PR. Adverse events were generally consistent with R-CHOP. Correlative analyses did not identify conclusive biomarkers of response. Durvalumab + R-CHOP is feasible in DLBCL with no new safety signals, but the combination provided no greater benefit than R-CHOP.
Collapse
|
45
|
Xu J, Zheng Q, Cheng X, Hu S, Zhang C, Zhou X, Sun P, Wang W, Su Z, Zou T, Song Z, Xia Y, Yi X, Gao Y. Chemo-photodynamic therapy with light-triggered disassembly of theranostic nanoplatform in combination with checkpoint blockade for immunotherapy of hepatocellular carcinoma. J Nanobiotechnology 2021; 19:355. [PMID: 34717654 PMCID: PMC8557521 DOI: 10.1186/s12951-021-01101-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignant tumor with high rate of metastasis and recurrence. Although immune checkpoint blockade (ICB) has emerged as a promising type of immunotherapy in advanced HCC, treatment with ICB alone achieves an objective remission rate less than 20%. Thus, combination therapy strategies is needed to improve the treatment response rate and therapeutic effect. METHODS A light-triggered disassembly of nanoplatform (TB/PTX@RTK) co-loaded an aggregation induced emission (AIE) photosensitizer (TB) and paclitaxel (PTX) was prepared for on-command drug release and synergistic chemo-photodynamic therapy (chemo-PDT). Nano-micelles were characterized for drug loading content, hydrodynamic size, absorption and emission spectra, reactive oxygen species production, and PTX release from micelles. The targeted fluorescence imaging of TB/PTX@RTK micelles and the synergistic anti-tumor efficacy of TB/PTX@RTK micelles-mediated chemo-PDT combined with anti-PD-L1 were assessed both in vitro and in vivo. RESULTS The TB/PTX@RTK micelles could specifically accumulate at the tumor site through cRGD-mediated active target and facilitate image-guided PDT for tumor ablation. Once irradiated by light, the AIE photosensitizer of TB could produce ROS for PDT, and the thioketal linker could be cleaved by ROS to precise release of PTX in tumor cells. Chemo-PDT could not only synergistically inhibit tumor growth, but also induce immunogenic cell death and elicit anti-tumor immune response. Meanwhile, chemo-PDT significantly upregulated the expression of PD-L1 on tumor cell surface which could efficiently synergize with anti-PD-L1 monoclonal antibodies to induce an abscopal effect, and establish long-term immunological memory to inhibit tumor relapse and metastasis. CONCLUSION Our results suggest that the combination of TB/PTX@RTK micelle-mediated chemo-PDT with anti-PD-L1 monoclonal antibodies can synergistically enhance systemic anti-tumor effects, and provide a novel insight into the development of new nanomedicine with precise controlled release and multimodal therapy to enhance the therapeutic efficacy of HCC.
Collapse
Affiliation(s)
- Jianjun Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qichang Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang Cheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xing Zhou
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ping Sun
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weimin Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhe Su
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tianhao Zou
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yun Xia
- Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaoqing Yi
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, College of Pharmacy, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, China.
| | - Yang Gao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
46
|
Majidi M, Safaee S, Amini M, Baghbanzadeh A, Hajiasgharzadeh K, Hashemzadeh S, Sandoghchian Shotorbani S, Mokhtarzadeh A, Baradaran B. The effects of chemotherapeutic drugs on PD-L1 gene expression in breast cancer cell lines. Med Oncol 2021; 38:147. [PMID: 34687372 DOI: 10.1007/s12032-021-01556-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/24/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common cancer among women in terms of prevalence and mortality, and chemotherapy is one of the most effective treatments at higher stages. However, resistance to chemotherapy is the main obstacle in the treatment of this cancer. Accumulated evidence identified the PD-L1 protein as an essential protein in the development of different cancers. Abnormal expression of this protein in various tumor cells is linked to cancer development and inhibiting the function of immune cells, which correlated with reduced beneficial effects of chemotherapy drugs. In the present study, the effects of common chemotherapy drugs including doxorubicin, paclitaxel, and docetaxel on the expression of the PD-L1 gene were investigated by qRT-PCR before and after the treatment with these drugs in MD231, MD468, SKBR3 breast cancer cell lines. Also, the MTT test was applied to examine the effects of drugs on the growth and proliferation of cancer cells considering PD-L1 expression. The expression of the PD-L1 gene increased after 24 and 48 h of treatment with chemotherapy drugs. The obtained results indicate the enhancing effects of chemotherapy drugs on PD-L1 gene expression, which have a suppressive effect on the immune system against breast cancer. The use of these drugs as the first line of chemotherapy in triple-negative breast cancer is not recommended. However, there is still a need for further experimental and clinical research on the exact effects of these drugs on undesired immune cells exhaustion in breast cancer therapy.
Collapse
Affiliation(s)
- Misagh Majidi
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran
| | - Sahar Safaee
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahriar Hashemzadeh
- Department of Thoracic Surgery, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
47
|
Kim R, Kin T. Current and Future Therapies for Immunogenic Cell Death and Related Molecules to Potentially Cure Primary Breast Cancer. Cancers (Basel) 2021; 13:cancers13194756. [PMID: 34638242 PMCID: PMC8507525 DOI: 10.3390/cancers13194756] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary How a cure for primary breast cancer after (neo)adjuvant therapy can be achieved at the molecular level remains unclear. Immune activation by anticancer drugs may contribute to the eradication of residual tumor cells by postoperative (neo)adjuvant chemotherapy. In addition, chemotherapy-induced immunogenic cell death (ICD) may result in long-term immune activation by memory effector T cells, leading to the curing of primary breast cancer. In this review, we discuss the molecular mechanisms by which anticancer drugs induce ICD and immunogenic modifications for antitumor immunity and targeted therapy against damage-associated molecular patterns. Our aim was to gain a better understanding of how to eradicate residual tumor cells treated with anticancer drugs and cure primary breast cancer by enhancing antitumor immunity with immune checkpoint inhibitors and vaccines. Abstract How primary breast cancer can be cured after (neo)adjuvant therapy remains unclear at the molecular level. Immune activation by anticancer agents may contribute to residual tumor cell eradication with postsurgical (neo)adjuvant chemotherapy. Chemotherapy-induced immunogenic cell death (ICD) may result in long-term immune activation with memory effector T cells, leading to a primary breast cancer cure. Anthracycline and taxane treatments cause ICD and immunogenic modulations, resulting in the activation of antitumor immunity through damage-associated molecular patterns (DAMPs), such as adenosine triphosphate, calreticulin, high mobility group box 1, heat shock proteins 70/90, and annexin A1. This response may eradicate residual tumor cells after surgical treatment. Although DAMP release is also implicated in tumor progression, metastasis, and drug resistance, thereby representing a double-edged sword, robust immune activation by anticancer agents and the subsequent acquisition of long-term antitumor immune memory can be essential components of the primary breast cancer cure. This review discusses the molecular mechanisms by which anticancer drugs induce ICD and immunogenic modifications for antitumor immunity and targeted anti-DAMP therapy. Our aim was to improve the understanding of how to eradicate residual tumor cells treated with anticancer drugs and cure primary breast cancer by enhancing antitumor immunity with immune checkpoint inhibitors and vaccines.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima 730-0051, Japan
- Correspondence:
| | - Takanori Kin
- Department of Breast Surgery, Hiroshima City Hospital, 7-33, Moto-machi, Naka-ku, Hiroshima 730-8518, Japan;
| |
Collapse
|
48
|
Umeda Y, Morikawa M, Anzai M, Ameshima S, Kadowaki M, Waseda Y, Shigemi H, Tsujikawa T, Kiyono Y, Okazawa H, Ishizuka T. Predictive value of integrated 18F-FDG PET/MRI in the early response to nivolumab in patients with previously treated non-small cell lung cancer. J Immunother Cancer 2021; 8:jitc-2019-000349. [PMID: 32345624 PMCID: PMC7213911 DOI: 10.1136/jitc-2019-000349] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2020] [Indexed: 12/26/2022] Open
Abstract
Background The early response to treatment with immune-checkpoint inhibitors is difficult to evaluate. We determined whether changes in integrated [18F]-fluoro-2-deoxy-D-glucose positron emission tomography/MRI (18F-FDG PET/MRI) parameters after the first 2 weeks of antiprogrammed death-1 antibody nivolumab therapy could predict the response of patients with non-small cell lung cancer (NSCLC). Methods Twenty-five patients with previously treated NSCLC were enrolled prospectively and underwent 18F-FDG PET/MRI before and at 2 weeks after nivolumab therapy. Changes in maximal standardized uptake value, total lesion glycolysis (ΔTLG) and apparent diffusion coefficient (ΔADC) between the two scans were calculated and evaluated for their associations with the clinical response to therapy. Results The disease control rate was 64%. Patients with non-progressive disease (non-PD) had significantly decreased TLG, increased ADCmean (ie, negative ΔADCmean) and lower ΔTLG+ΔADCmean than patients with PD. Among the parameters tested, receiver operating characteristic curve analysis revealed that a cut-off value of 16.5 for ΔTLG+ΔADCmean had the highest accuracy (92%) for distinguishing between patients with non-PD and PD. A ΔTLG+ΔADCmean value <16.5 was significantly associated with longer median progression-free survival (9.0 vs 1.8 months, p<0.00001) and overall survival (23.6 vs 4.7 months, p=0.0001) compared with ΔTLG+ΔADCmean value ≥16.5. A multivariate Cox model revealed that ≥16.5 ΔTLG+ΔADCmean was an independent predictor of shorter progression-free survival (HR 37.7) and overall survival (HR 9.29). Conclusions A combination of ΔTLG and ΔADCmean measured by integrated 18F-FDG PET/MRI may have value as a predictor of the response and survival of patients with NSCLC following nivolumab therapy. Trial registration number UMIN 000020707.
Collapse
Affiliation(s)
- Yukihiro Umeda
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Miwa Morikawa
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masaki Anzai
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Shingo Ameshima
- Department of Internal Medicine, Sakai Municipal Mikuni Hospital, Fukui, Japan
| | - Maiko Kadowaki
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yuko Waseda
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hiroko Shigemi
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tetsuya Tsujikawa
- Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| | - Yasushi Kiyono
- Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| | - Hidehiko Okazawa
- Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| | - Tamotsu Ishizuka
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
49
|
Shirasawa M, Yoshida T, Shimoda Y, Takayanagi D, Shiraishi K, Kubo T, Mitani S, Matsumoto Y, Masuda K, Shinno Y, Okuma Y, Goto Y, Horinouchi H, Ichikawa H, Kohno T, Yamamoto N, Matsumoto S, Goto K, Watanabe SI, Ohe Y, Motoi N. Differential Immune-Related Microenvironment Determines Programmed Cell Death Protein-1/Programmed Death-Ligand 1 Blockade Efficacy in Patients With Advanced NSCLC. J Thorac Oncol 2021; 16:2078-2090. [PMID: 34419685 DOI: 10.1016/j.jtho.2021.07.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/28/2021] [Accepted: 07/27/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Programmed death-ligand 1 (PD-L1) expression is not a completely reliable predictive marker of the efficacy of anti-programmed cell death protein-1 (PD-1)/PD-L1 therapy in patients with advanced NSCLC. Immune-related tumor microenvironment (TME) is classified into four different types based on the tumor-infiltrating lymphocyte (TIL) status and PD-L1 expression. METHODS We retrospectively reviewed patients with advanced NSCLC treated with anti-PD-1/PD-L1 therapy between 2015 and 2019. We investigated the association between the efficacy of anti-PD-1/PD-L1 therapy, the types of TME based on PD-L1 (clone: 22C3) expression, the density of CD8-positive TILs assessed by immunohistochemistry, and mutational profiles by next-generation sequencing. RESULTS Overall, 228 patients were included in the analysis. The patients were classified into the following four groups: type I: PD-L1High (tumor proportion score ≥ 50%)/TILHigh (≥85/mm2; n = 73); type II: PD-L1Low (tumor proportion score < 50%)/TILLow (<85/mm2; n = 70); type III: PD-L1High/TILLow (n = 37); and type IV: PD-L1Low/TILHigh (n = 48). The objective response rate (ORR) and progression-free survival (PFS) of anti-PD-1/PD-L1 therapy clearly differed according to the different TME types (ORR and PFS; type I: 64%, 14.5 mo; type II: 12%, 2.1 mo; type III: 24%, 3.6 mo; type IV; 41%, 10.8 mo). In patients with PD-L1High tumors, type I tumors had significantly better ORR and PFS than type III tumors (ORR: p < 0.001 and PFS: p < 0.001). The presence of TP53 and KRAS mutation was related to the density of CD8-positive TILs and PD-L1 expression, respectively. CONCLUSIONS Differential types of TME, including PD-L1 expression and TIL status, could accurately predict the efficacy of anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Masayuki Shirasawa
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| | - Yukiko Shimoda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Daisuke Takayanagi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Kubo
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Sachiyo Mitani
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuji Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Masuda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Shinno
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hitoshi Ichikawa
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Shun-Ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Noriko Motoi
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
50
|
Rahma OE, Yothers G, Hong TS, Russell MM, You YN, Parker W, Jacobs SA, Colangelo LH, Lucas PC, Gollub MJ, Hall WA, Kachnic LA, Vijayvergia N, O’Rourke MA, Faller BA, Valicenti RK, Schefter TE, Moxley KM, Kainthla R, Stella PJ, Sigurdson E, Wolmark N, George TJ. Use of Total Neoadjuvant Therapy for Locally Advanced Rectal Cancer: Initial Results From the Pembrolizumab Arm of a Phase 2 Randomized Clinical Trial. JAMA Oncol 2021; 7:1225-1230. [PMID: 34196693 PMCID: PMC8251652 DOI: 10.1001/jamaoncol.2021.1683] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/08/2021] [Indexed: 01/09/2023]
Abstract
IMPORTANCE Total neoadjuvant therapy (TNT) is often used to downstage locally advanced rectal cancer (LARC) and decrease locoregional relapse; however, more than one-third of patients develop recurrent metastatic disease. As such, novel combinations are needed. OBJECTIVE To assess whether the addition of pembrolizumab during and after neoadjuvant chemoradiotherapy can lead to an improvement in the neoadjuvant rectal (NAR) score compared with treatment with FOLFOX (5-fluorouracil, leucovorin, and oxaliplatin) and chemoradiotherapy alone. DESIGN, SETTING, AND PARTICIPANTS In this open-label, phase 2, randomized clinical trial (NRG-GI002), patients in academic and private practice settings were enrolled. Patients with stage II/III LARC with distal location (cT3-4 ≤ 5 cm from anal verge, any N), with bulky disease (any cT4 or tumor within 3 mm of mesorectal fascia), at high risk for metastatic disease (cN2), and/or who were not candidates for sphincter-sparing surgery (SSS) were stratified based on clinical tumor and nodal stages. Trial accrual opened on August 1, 2018, and ended on May 31, 2019. This intent-to-treat analysis is based on data as of August 2020. INTERVENTIONS Patients were randomized (1:1) to neoadjuvant FOLFOX for 4 months and then underwent chemoradiotherapy (capecitabine with 50.4 Gy) with or without intravenous pembrolizumab administered at a dosage of 200 mg every 3 weeks for up to 6 doses before surgery. MAIN OUTCOMES AND MEASURES The primary end point was the NAR score. Secondary end points included pathologic complete response (pCR) rate, SSS, disease-free survival, and overall survival. This report focuses on end points available after definitive surgery (NAR score, pCR, SSS, clinical complete response rate, margin involvement, and safety). RESULTS A total of 185 patients (126 [68.1%] male; mean [SD] age, 55.7 [11.1] years) were randomized to the control arm (CA) (n = 95) or the pembrolizumab arm (PA) (n = 90). Of these patients, 137 were evaluable for NAR score (68 CA patients and 69 PA patients). The mean (SD) NAR score was 11.53 (12.43) for the PA patients (95% CI, 8.54-14.51) vs 14.08 (13.82) for the CA patients (95% CI, 10.74-17.43) (P = .26). The pCR rate was 31.9% in the PA vs 29.4% in the CA (P = .75). The clinical complete response rate was 13.9% in the PA vs 13.6% in the CA (P = .95). The percentage of patients who underwent SSS was 59.4% in the PA vs 71.0% in the CA (P = .15). Grade 3 to 4 adverse events were slightly increased in the PA (48.2%) vs the CA (37.3%) during chemoradiotherapy. Two deaths occurred during FOLFOX: sepsis (CA) and pneumonia (PA). No differences in radiotherapy fractions, FOLFOX, or capecitabine doses were found. CONCLUSIONS AND RELEVANCE Pembrolizumab added to chemoradiotherapy as part of total neoadjuvant therapy was suggested to be safe; however, the NAR score difference does not support further study. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02921256.
Collapse
Affiliation(s)
- Osama E. Rahma
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Medical Oncology, Dana-Farber Cancer Institute/Alliance, Boston, Massachusetts
| | - Greg Yothers
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Theodore S. Hong
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Massachusetts General Hospital, Boston
| | - Marcia M. Russell
- NRG Oncology, Philadelphia, Pennsylvania
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Y. Nancy You
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - William Parker
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Medical Physics, McGill University Health Centre, Montréal, Quebec, Canada
| | | | - Linda H. Colangelo
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Peter C. Lucas
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Pathology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Marc J. Gollub
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William A. Hall
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee
| | - Lisa A. Kachnic
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Columbia University Irving Medical Center, Herbert Irving Comprehensive Cancer Center, New York, New York
- SWOG Cancer Research Network, San Antonio, Texas
| | - Namrata Vijayvergia
- NRG Oncology, Philadelphia, Pennsylvania
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mark A. O’Rourke
- NRG Oncology, Philadelphia, Pennsylvania
- National Cancer Institute Community Oncology Research Program, Prisma Health Cancer Institute, Greenville, South Carolina
| | - Bryan A. Faller
- Missouri Baptist Medical Center, Heartland Cancer Research, National Cancer Institute Community Oncology Research Program, St Louis
| | | | - Tracey E. Schefter
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Radiation Oncology, University of Colorado Cancer Center, Aurora
| | - Katherine M. Moxley
- NRG Oncology, Philadelphia, Pennsylvania
- Section of Gynecologic Oncology, University of Oklahoma Stephenson Cancer Center, Oklahoma City
| | - Radhika Kainthla
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Philip J. Stella
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Medical Oncology, St Joseph Mercy Hospital, Ann Arbor, Michigan
| | - Elin Sigurdson
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Norman Wolmark
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas J. George
- NRG Oncology, Philadelphia, Pennsylvania
- Department of Medicine, University of Florida Health Cancer Center, Gainesville
| |
Collapse
|