251
|
Boissière-Michot F, Chateau MC, Thézenas S, Guiu S, Bobrie A, Jacot W. Correlation of the TIGIT-PVR immune checkpoint axis with clinicopathological features in triple-negative breast cancer. Front Immunol 2022; 13:1058424. [PMID: 36544779 PMCID: PMC9760730 DOI: 10.3389/fimmu.2022.1058424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Background T cell immunoreceptor with Ig and ITIM domains (TIGIT) interacts with poliovirus receptor (PVR) to contribute to cancer immune escape. Recently, TIGIT and PVR have been identified as promising immunotherapy targets. Their gene expression is upregulated in many solid tumors, but their protein expression level is not well documented, particularly in triple negative breast cancer (TNBC), the breast cancer subtype that most benefit from immunotherapy. Methods TIGIT and PVR expression levels were assessed by immunohistochemistry in 243 surgically resected localized TNBC and then their relationship with clinical-pathological features and clinical outcome was analyzed. Results TIGIT expression was observed in immune cells from the tumor microenvironment, whereas PVR was mainly expressed by tumor cells. High TIGIT expression was significantly associated with age (p=0.010), histological grade (p=0.014), non-lobular histology (p=0.024), adjuvant chemotherapy (p=0.006), and various immune cell populations (tumor infiltrating lymphocytes (TILs), CD3+, CD8+, PD-1+ cells; all p<0.0001), PD-L1+ tumor cells (p<0.0001), and PD-L1+ stromal cells (p=0.003). Infiltration by TIGIT+ cells tended to be higher in non-molecular apocrine tumors (p=0.088). PVR was significantly associated with histological grade (p<0.0001), the basal-like (p=0.003) and non-molecular apocrine phenotypes (p=0.039), high TILs infiltration (p=0.011), CD3+ (p=0.002), CD8+ (p=0.024) T cells, and PD-L1 expression in tumor (p=0.003) and stromal cells (p=0.001). In univariate analysis, only known prognostic factors (age, tumor size, lymph node status, adjuvant chemotherapy, TILs and CD3+ T-cell infiltrate) were significantly associated with relapse-free survival (RFS) and overall survival. High TIGIT and PVR expression levels tended to be associated with longer RFS (p=0.079 and 0.045, respectively). The analysis that included only non-molecular apocrine TNBC revealed longer RFS for tumors that strongly expressed TIGIT or PVR (p=0.025 for TIGIT and 0.032 for PVR). Conclusions These results indicated that in TNBC, TIGIT+ cells can easily interact with PVR to exert their inhibitory effects. Their wide expression in TNBC and their association with other immune checkpoint components suggest the therapeutic interest of the TIGIT-PVR axis.
Collapse
Affiliation(s)
- Florence Boissière-Michot
- Translational Research Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,*Correspondence: Florence Boissière-Michot,
| | - Marie-Christine Chateau
- Translational Research Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France
| | - Simon Thézenas
- Biometrics Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France
| | - Séverine Guiu
- Department of Medical Oncology, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Montpellier, France
| | - Angélique Bobrie
- Department of Medical Oncology, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Montpellier, France
| | - William Jacot
- Translational Research Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Department of Medical Oncology, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Montpellier, France,Faculty of Medicine, Montpellier University, Montpellier, France
| |
Collapse
|
252
|
Shahbandi A, Chiu FY, Ungerleider NA, Kvadas R, Mheidly Z, Sun MJS, Tian D, Waizman DA, Anderson AY, Machado HL, Pursell ZF, Rao SG, Jackson JG. Breast cancer cells survive chemotherapy by activating targetable immune-modulatory programs characterized by PD-L1 or CD80. NATURE CANCER 2022; 3:1513-1533. [PMID: 36482233 PMCID: PMC9923777 DOI: 10.1038/s43018-022-00466-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 10/14/2022] [Indexed: 04/16/2023]
Abstract
Breast cancer cells must avoid intrinsic and extrinsic cell death to relapse following chemotherapy. Entering senescence enables survival from mitotic catastrophe, apoptosis and nutrient deprivation, but mechanisms of immune evasion are poorly understood. Here we show that breast tumors surviving chemotherapy activate complex programs of immune modulation. Characterization of residual disease revealed distinct tumor cell populations. The first population was characterized by interferon response genes, typified by Cd274, whose expression required chemotherapy to enhance chromatin accessibility, enabling recruitment of IRF1 transcription factor. A second population was characterized by p53 signaling, typified by CD80 expression. Treating mammary tumors with chemotherapy followed by targeting the PD-L1 and/or CD80 axes resulted in marked accumulation of T cells and improved response; however, even combination strategies failed to fully eradicate tumors in the majority of cases. Our findings reveal the challenge of eliminating residual disease populated by senescent cells expressing redundant immune inhibitory pathways and highlight the need for rational immune targeting strategies.
Collapse
Affiliation(s)
- Ashkan Shahbandi
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Fang-Yen Chiu
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Nathan A Ungerleider
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Raegan Kvadas
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Zeinab Mheidly
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Meijuan J S Sun
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Di Tian
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Daniel A Waizman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ashlyn Y Anderson
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Heather L Machado
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Zachary F Pursell
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Sonia G Rao
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - James G Jackson
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
253
|
He R, Yuan X, Chen Z, Zheng Y. Combined immunotherapy for metastatic triple-negative breast cancer based on PD-1/PD-L1 immune checkpoint blocking. Int Immunopharmacol 2022; 113:109444. [DOI: 10.1016/j.intimp.2022.109444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
|
254
|
Immune Checkpoint Inhibitors and Novel Immunotherapy Approaches for Breast Cancer. Curr Oncol Rep 2022; 24:1801-1819. [PMID: 36255603 DOI: 10.1007/s11912-022-01339-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW To critically review the existing evidence on immune checkpoint inhibitors (ICIs) in early-stage and metastatic breast cancer and discuss emerging strategies in the different breast cancer subtypes. RECENT FINDINGS Immunotherapy has become one of the major milestones in contemporary oncology, revolutionizing the treatment of multiple solid tumors. ICI agents combined with chemotherapy have demonstrated significant efficacy in both early-stage and metastatic triple-negative breast cancer. However, only a subgroup of patients responds to those agents and some associated toxicities, although infrequent, can be life-disabling. Emerging data from immunotherapy studies in advanced hormone receptor-positive (HR +) breast cancer as well as HER2-positive disease are arising with mixed results. Although breast cancer has not classically been considered a hot tumor, ICIs have proven to be effective in a subset of breast cancer patients. However, much remains to be learned, and the identification of new biomarkers beyond PD-L1 expression is essential not only to improve the efficacy of ICI but also to identify patients who can avoid them, together with their toxicities and costs.
Collapse
|
255
|
Passalacqua MI, Rizzo G, Santarpia M, Curigliano G. 'Why is survival with triple negative breast cancer so low? insights and talking points from preclinical and clinical research'. Expert Opin Investig Drugs 2022; 31:1291-1310. [PMID: 36522800 DOI: 10.1080/13543784.2022.2159805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Triple negative breast cancer is typically related to poor prognosis, early metastasis, and high recurrence rate. Intrinsic and extrinsic biological features of TNBC and resistance mechanisms to conventional therapies can support its aggressive behavior, characterizing TNBC how extremely heterogeneous. Novel combination strategies are under investigation, including immunotherapeutic agents, anti-drug conjugates, PARP inhibitors, and various targeting agents, exploring, in the meanwhile, possible predictive biomarkers to correctly select patients for the optimal treatment for their specific subtype. AREAS COVERED This article examines the main malignity characteristics across different subtype, both histological and molecular, and the resistance mechanisms, both primary and acquired, to different drugs explored in the landscape of TNBC treatment, that lead TNBC to still has high mortality rate. EXPERT OPINION The complexity of TNBC is not only the main reason of its aggressivity, but its heterogeneity should be exploited in terms of therapeutics opportunities, combining agents with different mechanism of action, after a correct selection by biologic or molecular biomarkers. The main goal is to understand what TNBC really is and to act selectively on its characteristics, with a personalized anticancer treatment.
Collapse
Affiliation(s)
- Maria Ilenia Passalacqua
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.,Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Graziella Rizzo
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.,Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
256
|
Kudelova E, Smolar M, Holubekova V, Hornakova A, Dvorska D, Lucansky V, Koklesova L, Kudela E, Kubatka P. Genetic Heterogeneity, Tumor Microenvironment and Immunotherapy in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms232314937. [PMID: 36499265 PMCID: PMC9735793 DOI: 10.3390/ijms232314937] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
Heterogeneity of triple-negative breast cancer is well known at clinical, histopathological, and molecular levels. Genomic instability and greater mutation rates, which may result in the creation of neoantigens and enhanced immunogenicity, are additional characteristics of this breast cancer type. Clinical outcome is poor due to early age of onset, high metastatic potential, and increased likelihood of distant recurrence. Consequently, efforts to elucidate molecular mechanisms of breast cancer development, progression, and metastatic spread have been initiated to improve treatment options and improve outcomes for these patients. The extremely complex and heterogeneous tumor immune microenvironment is made up of several cell types and commonly possesses disorganized gene expression. Altered signaling pathways are mainly associated with mutated genes including p53, PIK3CA, and MAPK, and which are positively correlated with genes regulating immune response. Of note, particular immunity-associated genes could be used in prognostic indexes to assess the most effective management. Recent findings highlight the fact that long non-coding RNAs also play an important role in shaping tumor microenvironment formation, and can mediate tumor immune evasion. Identification of molecular signatures, through the use of multi-omics approaches, and effector pathways that drive early stages of the carcinogenic process are important steps in developing new strategies for targeted cancer treatment and prevention. Advances in immunotherapy by remodeling the host immune system to eradicate tumor cells have great promise to lead to novel therapeutic strategies. Current research is focused on combining immune checkpoint inhibition with chemotherapy, PARP inhibitors, cancer vaccines, or natural killer cell therapy. Targeted therapies may improve therapeutic response, eliminate therapeutic resistance, and improve overall patient survival. In the future, these evolving advancements should be implemented for personalized medicine and state-of-art management of cancer patients.
Collapse
Affiliation(s)
- Eva Kudelova
- Clinic of Surgery and Transplant Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Marek Smolar
- Clinic of Surgery and Transplant Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Veronika Holubekova
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Andrea Hornakova
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Dana Dvorska
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Vincent Lucansky
- Biomedical Centre, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Erik Kudela
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
- Correspondence:
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
257
|
Zhang Y, Wang J, Hu T, Wang H, Long M, Liang B. Adverse Events of PD-1 or PD-L1 Inhibitors in Triple-Negative Breast Cancer: A Systematic Review and Meta-Analysis. Life (Basel) 2022; 12:1990. [PMID: 36556355 PMCID: PMC9787874 DOI: 10.3390/life12121990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
(1) Background: This study aimed to develop a comprehensive understanding of the treatment-related adverse events when using PD-1 or PD-L1 inhibitors in triple-negative breast cancer (TNBC). (2) Methods: We conducted a meta-analysis of Phase II/III randomized clinical trials. Studies were searched for using PubMed, Embase, and Cochrane Library from 1 March 1980 till 30 June 2022. Data on adverse events were mainly extracted from ClinicalTrials.gov and published articles. A generalized linear mixed model with the logit transformation was employed to obtain the overall incidence of adverse events across all studies. For serious adverse events with low incidences, the Peto method was used to calculate the odds ratio (OR) and 95% confidence interval (95%CI) in the PD-1 or PD-L1 inhibitors groups compared to the control groups. (3) Results: Nine studies were included in the meta-analysis, including a total of 2941 TNBC patients treated with PD-1 or PD-L1 inhibitors (including atezolizumab, pembrolizumab and durvalumab) and 2339 patients in the control groups. Chemotherapy alone was the control group in all studies. The average incidences of all serious immune-related adverse events of interest (hypothyroidism, hyperthyroidism, pneumonitis, pruritus, rash) were less than 1%, except for adrenal insufficiency (1.70%, 95%CI: 0.50-5.61%) in the PD-1 or PD-L1 groups. PD-1 or PD-L1 inhibitors significantly increased the risk of serious pneumonitis (OR = 2.52, 95%CI: 1.02-6.26), hypothyroidism (OR = 5.92, 95%CI: 1.22-28.86), alanine aminotransferase (ALT) elevation (OR = 1.66, 95%CI: 1.12-2.45), and adrenal insufficiency (OR = 18.81, 95%CI: 3.42-103.40). For non-serious adverse events, the patients treated with PD-1 or PD-L1 inhibitors had higher risk of aspartate aminotransferase (AST) elevation (OR =1.26, 95%CI: 1.02-1.57), hypothyroidism (OR = 3.63, 95%CI: 2.92-4.51), pruritus (OR = 1.84, 95%CI: 1.30-2.59), rash (OR = 1.29, 95%CI: 1.08-1.55), and fever (OR = 1.77, 95%CI: 1.13-2.77), compared with chemotherapy alone. (4) Conclusions: The incidence of serious immune-related adverse events in PD-1 or PD-L1 inhibitors groups is low but significantly higher than in chemotherapy groups. When using PD-1 or PD-L1 inhibitors for the treatment of TNBC, serious pneumonitis, hypothyroidism, ALT elevation, and adrenal insufficiency should be considered. Non-serious adverse events, such as AST elevation, rash, and fever, should also be taken into consideration.
Collapse
Affiliation(s)
- Yixi Zhang
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Jingyuan Wang
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Taobo Hu
- Department of Breast Surgery, Peking University People’s Hospital, Beijing 100044, China
| | - Huina Wang
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Mengping Long
- Department of Pathology, Peking University Cancer Hospital, Beijing 100083, China
| | - Baosheng Liang
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| |
Collapse
|
258
|
Bone Metastasis of Breast Cancer: Molecular Mechanisms and Therapeutic Strategies. Cancers (Basel) 2022; 14:cancers14235727. [PMID: 36497209 PMCID: PMC9738274 DOI: 10.3390/cancers14235727] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Bone metastasis is a common complication of many types of advanced cancer, including breast cancer. Bone metastasis may cause severe pain, fractures, and hypercalcemia, rendering clinical management challenging and substantially reducing the quality of life and overall survival (OS) time of breast cancer patients. Studies have revealed that bone metastasis is related to interactions between tumor cells and the bone microenvironment, and involves complex molecular biological mechanisms, including colonization, osteolytic destruction, and an immunosuppressive bone microenvironment. Agents inhibiting bone metastasis (such as bisphosphate and denosumab) alleviate bone destruction and improve the quality of life of breast cancer patients with bone metastasis. However, the prognosis of these patients remains poor, and the specific biological mechanism of bone metastasis is incompletely understood. Additional basic and clinical studies are urgently needed, to further explore the mechanism of bone metastasis and develop new therapeutic drugs. This review presents a summary of the molecular mechanisms and therapeutic strategies of bone metastasis of breast cancer, aiming to improve the quality of life and prognosis of breast cancer patients and provide a reference for future research directions.
Collapse
|
259
|
Ramnaraign BH, Lee JH, Ali A, Rogers SC, Fabregas JC, Thomas RM, Allegra CJ, Sahin I, DeRemer DL, George TJ, Chatzkel JA. Atezolizumab plus tivozanib for immunologically cold tumor types: the IMMCO-1 trial. Future Oncol 2022; 18:3815-3822. [PMID: 36399037 DOI: 10.2217/fon-2022-0392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Immune checkpoint inhibitor therapy represents a significant advance in cancer care. The interaction between PD-1 and PD-L1 induces immune tolerance and the inhibition of this interaction is an effective treatment strategy for numerous malignancies. Despite its demonstrated potential, immunotherapy is not clinically effective in immunogenically 'cold' tumors such as pancreatic cancer, prostate cancer and neuroendocrine tumors. Through the inhibition of VEGF, it may be possible to potentiate the effect of immune checkpoint blockade in tumors that have traditionally shown a lack of clinical response to immunotherapy. This signal-seeking, single-arm, prospective clinical trial aims to determine the objective response of tivozanib and atezolizumab in advanced immunogenically cold solid tumors. Clinical Trial Registration: NCT05000294 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Brian H Ramnaraign
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Ji-Hyun Lee
- Department of Biostatistics, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Azka Ali
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Sherise C Rogers
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Jesus C Fabregas
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Ryan M Thomas
- Department of Surgery, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Carmen J Allegra
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Ilyas Sahin
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - David L DeRemer
- Department of Pharmacy, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Thomas J George
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Jonathan A Chatzkel
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| |
Collapse
|
260
|
Dai M, Liu M, Yang H, Küçük C, You H. New insights into epigenetic regulation of resistance to PD-1/PD-L1 blockade cancer immunotherapy: mechanisms and therapeutic opportunities. Exp Hematol Oncol 2022; 11:101. [PMID: 36384676 PMCID: PMC9667634 DOI: 10.1186/s40164-022-00356-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Programmed cell death protein 1(PD-1) is a type of immune-inhibitory checkpoint protein, which delivers inhibitory signals to cytotoxic T cells by binding to the programmed death ligand-1 (PD-L1) displayed on the surface of cancer cells. Antibodies blocking PD-1/PD-L1 interaction have been extensively used in treatment of human malignancies and have achieved promising outcomes in recent years. However, gradual development of resistance to PD-1/PD-L1 blockade has decreased the effectiveness of this immunotherapy in cancer patients. The underlying epigenetic mechanisms need to be elucidated for application of novel strategies overcoming this immunotherapy resistance. Epigenetic aberrations contribute to cancerogenesis by promoting different hallmarks of cancer. Moreover, these alterations may lead to therapy resistance, thereby leading to poor prognosis. Recently, the epigenetic regulatory drugs have been shown to decrease the resistance to PD-1/PD-L1 inhibitors in certain cancer patients. Inhibitors of the non-coding RNAs, DNA methyltransferases, and histone deacetylases combined with PD-1/PD-L1 inhibitors have shown considerable therapeutic efficacy against carcinomas as well as blood cancers. Importantly, DNA methylation-mediated epigenetic silencing can inhibit antigen processing and presentation, which promotes cancerogenesis and aggravates resistance to PD-1/PD-L1 blockade immunotherapy. These observations altogether suggest that the combination of the epigenetic regulatory drugs with PD-1/PD-L1 inhibitors may present potential solution to the resistance caused by monotherapy of PD-1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Mengyuan Dai
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Hematology and Oncology, Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan Second Rd., Yuzhong District, 401122, Chongqing, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Hua Yang
- Department of Basic Medicine and Biomedical Engineering, School of Medical, Foshan University, Foshan, China
| | - Can Küçük
- İzmir International Biomedicine and Genome Institute, Dokuz Eylül University, İzmir, Türkiye
- Basic and Translational Research Program, İzmir Biomedicine and Genome Center, İzmir, Türkiye
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, İzmir, Türkiye
| | - Hua You
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Hematology and Oncology, Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan Second Rd., Yuzhong District, 401122, Chongqing, China.
| |
Collapse
|
261
|
Uliano J, Nicolò E, Corvaja C, Taurelli Salimbeni B, Trapani D, Curigliano G. Combination immunotherapy strategies for triple-negative breast cancer: current progress and barriers within the pharmacological landscape. Expert Rev Clin Pharmacol 2022; 15:1399-1413. [DOI: 10.1080/17512433.2022.2142559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jacopo Uliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Eleonora Nicolò
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carla Corvaja
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Medicine, University of Udine, Udine, Italy
| | - Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Clinical and Molecular Medicine, Oncology Unit, “La Sapienza” University of RomeAzienda Ospedaliera Sant’Andrea, Rome, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Medical Oncology, Medical Oncology Dana Farber Cancer Institute, Boston, MA, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
262
|
Liu S, Li J, Gu L, Wu K, Xing H. Nanoparticles for Chemoimmunotherapy Against Triple-Negative Breast Cancer. Int J Nanomedicine 2022; 17:5209-5227. [PMID: 36388877 PMCID: PMC9651025 DOI: 10.2147/ijn.s388075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) exhibits high recurrence and mortality rates because of the lack of effective treatment targets. Surgery and traditional chemotherapy are the primary treatment options. Immunotherapy shows high potential for treating various cancers but exhibits limited efficacy against TNBC as a monotherapy. Chemoimmunotherapy has broad prospects for applications for cancer treatment conferred through the synergistic immunomodulatory and anti-tumor effects of chemotherapy and immunotherapeutic strategies. However, improving the efficacy of synergistic therapy and reducing the side effects of multiple drugs remain to be the main challenges in chemoimmunotherapy against TNBC. Nanocarriers can target both cancer and immune cells, promote drug accumulation, and show minimal toxicity, making them ideal delivery systems for chemotherapeutic and immunotherapeutic agents. In this review, we introduce the immunomodulatory effects of chemotherapy and combined mechanisms of chemoimmunotherapy, followed by a summary of nanoparticle-mediated chemoimmunotherapeutic strategies used for treating TNBC. This up-to-date synthesis of relevant findings in the field merits contemplation, while considering avenues of investigation to enable advances in the field.
Collapse
Affiliation(s)
- Siyan Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Jing Li
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Lin Gu
- Breast Surgery, Jilin Province Tumor Hospital, Changchun, People’s Republic of China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Hua Xing
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
263
|
MYC promotes immune-suppression in triple-negative breast cancer via inhibition of interferon signaling. Nat Commun 2022; 13:6579. [PMID: 36323660 PMCID: PMC9630413 DOI: 10.1038/s41467-022-34000-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
The limited efficacy of immune checkpoint inhibitor treatment in triple-negative breast cancer (TNBC) patients is attributed to sparse or unresponsive tumor-infiltrating lymphocytes, but the mechanisms that lead to a therapy resistant tumor immune microenvironment are incompletely known. Here we show a strong correlation between MYC expression and loss of immune signatures in human TNBC. In mouse models of TNBC proficient or deficient of breast cancer type 1 susceptibility gene (BRCA1), MYC overexpression dramatically decreases lymphocyte infiltration in tumors, along with immune signature remodelling. MYC-mediated suppression of inflammatory signalling induced by BRCA1/2 inactivation is confirmed in human TNBC cell lines. Moreover, MYC overexpression prevents the recruitment and activation of lymphocytes in both human and mouse TNBC co-culture models. Chromatin-immunoprecipitation-sequencing reveals that MYC, together with its co-repressor MIZ1, directly binds promoters of multiple interferon-signalling genes, resulting in their downregulation. MYC overexpression thus counters tumor growth inhibition by a Stimulator of Interferon Genes (STING) agonist via suppressing induction of interferon signalling. Together, our data reveal that MYC suppresses innate immunity and facilitates tumor immune escape, explaining the poor immunogenicity of MYC-overexpressing TNBCs.
Collapse
|
264
|
Castellano LRC, Cruz SBSC, Hier M, Bonan PRF, Alaoui-Jamali MA, da Silva SD. Implications and Emerging Therapeutic Avenues of Inflammatory Response in HPV+ Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:5406. [PMID: 36358823 PMCID: PMC9657300 DOI: 10.3390/cancers14215406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 10/24/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are a heterogeneous group of malignancies which have shown exponential incidence in the last two decades especially due to human papillomavirus (HPV) infection. The HPV family comprises more than 100 types of viruses with HPV16 and HPV18 being the most prevalent strains in HNSCC. Literature data reveal that the mutation profile as well as the response to chemotherapy and radiotherapy are distinct among HPV+ versus HPV-negative tumors. Furthermore, the presence of the virus induces activation of an immune response, in particular the recruitment of specific antiviral T lymphocytes to tumor sites. These T cells when activated produce soluble factors including cytokines and chemokines capable of modifying the local immune tumor microenvironment and impact on tumor response to the treatment. In this comprehensive review we investigated current knowledge on how the presence of an HPV can modify the inflammatory response systemically and within the tumor microenvironment's immunological responses, thereby impacting on disease prognosis and survival. We highlighted the research gaps and emerging approaches necessary to discover novel immunotherapeutic targets for HPV-associated HNSCC.
Collapse
Affiliation(s)
- Lúcio Roberto Cançado Castellano
- Department of Otolaryngology and Head and Neck Surgery and Lady Davis Institutes for Medical Research of the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Human Immunology Research and Education Group, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
- Graduate Program in Dentistry, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Sara Brito Silva Costa Cruz
- Department of Otolaryngology and Head and Neck Surgery and Lady Davis Institutes for Medical Research of the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Human Immunology Research and Education Group, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
- Graduate Program in Dentistry, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Michael Hier
- Department of Otolaryngology and Head and Neck Surgery and Lady Davis Institutes for Medical Research of the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Paulo Rogério Ferreti Bonan
- Human Immunology Research and Education Group, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
- Graduate Program in Dentistry, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Moulay A. Alaoui-Jamali
- Department of Otolaryngology and Head and Neck Surgery and Lady Davis Institutes for Medical Research of the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Sabrina Daniela da Silva
- Department of Otolaryngology and Head and Neck Surgery and Lady Davis Institutes for Medical Research of the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
265
|
Lee DY, Im E, Yoon D, Lee YS, Kim GS, Kim D, Kim SH. Pivotal role of PD-1/PD-L1 immune checkpoints in immune escape and cancer progression: Their interplay with platelets and FOXP3+Tregs related molecules, clinical implications and combinational potential with phytochemicals. Semin Cancer Biol 2022; 86:1033-1057. [PMID: 33301862 DOI: 10.1016/j.semcancer.2020.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 01/27/2023]
Abstract
Immune checkpoint proteins including programmed cell death protein 1 (PD-1), its ligand PD-L1 and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are involved in proliferation, angiogenesis, metastasis, chemoresistance via immune escape and immune tolerance by disturbing cytotoxic T cell activation. Though many clinical trials have been completed in several cancers by using immune checkpoint inhibitors alone or in combination with other agents to date, recently multi-target therapy is considered more attractive than monotherapy, since immune checkpoint proteins work with other components such as surrounding blood vessels, dendritic cells, fibroblasts, macrophages, platelets and extracellular matrix within tumor microenvironment. Thus, in the current review, we look back on research history of immune checkpoint proteins and discuss their associations with platelets or tumor cell induced platelet aggregation (TCIPA) and FOXP3+ regulatory T cells (Tregs) related molecules involved in immune evasion and tumor progression, clinical implications of completed trial results and signaling networks by phytochemicals for combination therapy with immune checkpoint inhibitors and suggest future research perspectives.
Collapse
Affiliation(s)
- Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Eunji Im
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dahye Yoon
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Young-Seob Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Geum-Soog Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Donghwi Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
266
|
Taurelli Salimbeni B, Corvaja C, Valenza C, Zagami P, Curigliano G. The triple negative breast cancer drugs graveyard: a review of failed clinical trials 2017-2022. Expert Opin Investig Drugs 2022; 31:1203-1226. [PMID: 36413823 DOI: 10.1080/13543784.2022.2151433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) accounts for 15-20% of breast cancers (BC) and has the worst prognosis. It is characterized by the absence of both hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2). TNBC has more limited therapeutic options compared to other subtypes, meaning that there is still a long way to go to discover target treatments. AREAS COVERED Our review aims to summarize phase II/III clinical trials enrolling patients with TNBC that have been published between 2017 and 2022 but failed to reach their primary endpoint. We here try to emphasize the limitations and weaknesses noted in negative studies and to point out unexpected results which might be useful to enhance the therapeutic approach to TNBC disease. EXPERT OPINION A deeper understanding of the mechanisms behind TNBC heterogeneity allowed to enhance the knowledge of new prognostic and predictive biomarkers of response. However, it is also through several failed clinical trials that we were able to define new therapeutic approaches which improved TNBC patients' clinical outcomes. Nowadays, we still need to overcome several difficulties to fully recognize different intracellular and extracellular pathways that crosstalk in TNBC and the mechanisms of resistance to identify novel tailored-patients' therapies.
Collapse
Affiliation(s)
- Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Clinical and Molecular Medicine, Oncology Unit, "la Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy
| | - Carla Corvaja
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Medicine, University of Udine, Udine, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| | - Paola Zagami
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| |
Collapse
|
267
|
PD-L1 Expression in Triple-negative Breast Cancer-a Comparative Study of 3 Different Antibodies. Appl Immunohistochem Mol Morphol 2022; 30:726-730. [PMID: 36165931 PMCID: PMC9983741 DOI: 10.1097/pai.0000000000001062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 08/12/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Assessment of programmed death protein-ligand 1 (PD-L1) in triple-negative breast cancer (TNBC) has entered daily practice to identify patients eligible for treatment with immune checkpoint inhibitors. However, different antibodies and different cut-offs for PD-L1 positivity are used, and the interchangeability of these methods is not clear. The aim of our study was to analyze whether different PD-L1 antibodies can be used interchangeably to identify TNBC patients as PD-L1 positive. METHODS A tissue microarray encompassing 147 TNBC cases was immunohistochemically analyzed using 3 different antibodies against PD-L1: SP142, SP263, and E1L3N. PD-L1 positivity was determined as ≥1% of positive tumor-associated immune cells. The staining patterns of the 3 antibodies were compared and correlated with clinicopathological data. RESULTS A total of 84 cases were evaluable for PD-L1 analysis with all 3 antibodies. PD-L1 was positive in 50/84 patients (59.5%) with SP263, in 44/84 (52.4%) with E1L3N, and in 29/84 (34.5%) with SP142. There was no statistical difference between the performance of SP263 and E1L3N, but both antibodies stained significantly more cases than the SP142 antibody. CONCLUSIONS Our results show that the 3 PD-L1 antibodies identify different TNBC patient subgroups as PD-L1 positive and, therefore cannot be used interchangeably. Additional studies are needed to further investigate the use and impact of different PD-L1 antibody clones for predictive selection of TNBC patients for treatment with immune checkpoint inhibitors.
Collapse
|
268
|
Varma R, Wright M, Abraham J, Kruse M. Immune checkpoint inhibition in early-stage triple-negative breast cancer. Expert Rev Anticancer Ther 2022; 22:1225-1238. [PMID: 36278877 DOI: 10.1080/14737140.2022.2139240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Breast cancer cells can evade immune recognition by upregulating programmed death-ligand 1 (PD-L1) leading to decreased T cell function. Anti-PD-1 agents, like pembrolizumab, and anti-PD-L1 agents, such as atezolizumab and durvalumab, in combination with chemotherapy were found to have efficacy in metastatic triple-negative breast cancer (TNBC). With sub-optimal long-term outcomes in early-stage TNBC, this combination of immune checkpoint inhibition with chemotherapy was subsequently investigated. A robust immune microenvironment and extensive tumor antigen exposure in early-stage breast cancer is believed to facilitate response to checkpoint inhibitors. AREAS COVERED This review focuses on studies that assess the role of neoadjuvant immune checkpoint inhibition along with chemotherapy. The results of key phase I, II and III trials using checkpoint inhibitors in early-stage breast cancer (ESBC) are reviewed along with foundational data from metastatic TNBC, including the role of biomarkers in predicting response to immunotherapy. EXPERT OPINION Despite a clear role for neoadjuvant immune checkpoint inhibition in TNBC, many questions remain. The benefit of these agents in the neoadjuvant versus adjuvant setting is unclear and immune-related toxicity is a major concern. Additional studies are needed to elucidate which immune checkpoint inhibitor is most efficacious and best tolerated in early-stage TNBC.
Collapse
Affiliation(s)
- Revati Varma
- Jawaharlal Institute of Post-graduate Medical Education and Research (JIPMER), Puducherry, India
| | - Matthew Wright
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland, Ohio, United States
| | - Jame Abraham
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland, Ohio, United States
| | - Megan Kruse
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland, Ohio, United States
| |
Collapse
|
269
|
Zhang J, Chan DW, Lin SY. Exploiting DNA Replication Stress as a Therapeutic Strategy for Breast Cancer. Biomedicines 2022; 10:2775. [PMID: 36359297 PMCID: PMC9687274 DOI: 10.3390/biomedicines10112775] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 09/19/2023] Open
Abstract
Proliferating cells rely on DNA replication to ensure accurate genome duplication. Cancer cells, including breast cancer cells, exhibit elevated replication stress (RS) due to the uncontrolled oncogenic activation, loss of key tumor suppressors, and defects in the DNA repair machinery. This intrinsic vulnerability provides a great opportunity for therapeutic exploitation. An increasing number of drug candidates targeting RS in breast cancer are demonstrating promising efficacy in preclinical and early clinical trials. However, unresolved challenges lie in balancing the toxicity of these drugs while maintaining clinical efficacy. Furthermore, biomarkers of RS are urgently required to guide patient selection. In this review, we introduce the concept of targeting RS, detail the current therapies that target RS, and highlight the integration of RS with immunotherapies for breast cancer treatment. Additionally, we discuss the potential biomarkers to optimizing the efficacy of these therapies. Together, the continuous advances in our knowledge of targeting RS would benefit more patients with breast cancer.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
270
|
Zhou Z, Tao C, Li J, Tang JCO, Chan ASC, Zhou Y. Chimeric antigen receptor T cells applied to solid tumors. Front Immunol 2022; 13:984864. [PMID: 36389701 PMCID: PMC9659902 DOI: 10.3389/fimmu.2022.984864] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/18/2022] [Indexed: 12/02/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy is novel tumor immunotherapy that enables autologous T to express synthetic receptors to specifically recognize the surface tumor-associated antigens for exerting subsequent antitumor effects, and eliminating the resistance, metastases and recurrence of cancer. Although CAR T cells have exhibited success in eradicating hematologic malignancies, their applications to solid tumors has not yet been achieved due to obstacles such as the immune-suppressor tumor microenvironment and lack of tumor specific target antigens. In this review, we presented advancements in the development of CAR T cell therapy in solid tumors, and offered a brief summary of the challenges, as well as novel engineering and pharmaceutical interventions to overcome these barriers. Looking forward, we discussed the latest studies which are expected to reach the clinicals in the next few years, including CRISPR screens-based CAR modification and CAR T cells driven from progenitor-like T cells. Collectively, this review may inspire researchers and clinicians to develop clinical available strategies of CAR T cell therapies in solid tumor.
Collapse
Affiliation(s)
- Zhongguo Zhou
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Can Tao
- School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Jianting Li
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Johnny Cheuk-on Tang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Kamford Genetics Company Limited, Hong Kong, Hong Kong SAR, China
| | - Albert Sun-chi Chan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Zhou
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
- *Correspondence: Yuanyuan Zhou,
| |
Collapse
|
271
|
Jungles KM, Holcomb EA, Pearson AN, Jungles KR, Bishop CR, Pierce LJ, Green MD, Speers CW. Updates in combined approaches of radiotherapy and immune checkpoint inhibitors for the treatment of breast cancer. Front Oncol 2022; 12:1022542. [PMID: 36387071 PMCID: PMC9643771 DOI: 10.3389/fonc.2022.1022542] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022] Open
Abstract
Breast cancer is the most prevalent non-skin cancer diagnosed in females and developing novel therapeutic strategies to improve patient outcomes is crucial. The immune system plays an integral role in the body’s response to breast cancer and modulating this immune response through immunotherapy is a promising therapeutic option. Although immune checkpoint inhibitors were recently approved for the treatment of breast cancer patients, not all patients respond to immune checkpoint inhibitors as a monotherapy, highlighting the need to better understand the biology underlying patient response. Additionally, as radiotherapy is a critical component of breast cancer treatment, understanding the interplay of radiation and immune checkpoint inhibitors will be vital as recent studies suggest that combined therapies may induce synergistic effects in preclinical models of breast cancer. This review will discuss the mechanisms supporting combined approaches with radiotherapy and immune checkpoint inhibitors for the treatment of breast cancer. Moreover, this review will analyze the current clinical trials examining combined approaches of radiotherapy, immunotherapy, chemotherapy, and targeted therapy. Finally, this review will evaluate data regarding treatment tolerance and potential biomarkers for these emerging therapies aimed at improving breast cancer outcomes.
Collapse
Affiliation(s)
- Kassidy M. Jungles
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Erin A. Holcomb
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ashley N. Pearson
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kalli R. Jungles
- Department of Biology, Saint Mary’s College, Notre Dame, IN, United States
| | - Caroline R. Bishop
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Lori J. Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Michael D. Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
- *Correspondence: Michael D. Green, ; Corey W. Speers,
| | - Corey W. Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Radiation Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Case Comprehensive Cancer Center, Cleveland, OH, United States
- *Correspondence: Michael D. Green, ; Corey W. Speers,
| |
Collapse
|
272
|
Li X, Tang L, Chen Q, Cheng X, Liu Y, Wang C, Zhu C, Xu K, Gao F, Huang J, Wang R, Guan X. Inhibition of MYC suppresses programmed cell death ligand-1 expression and enhances immunotherapy in triple-negative breast cancer. Chin Med J (Engl) 2022; 135:2436-2445. [PMID: 36583862 PMCID: PMC9945371 DOI: 10.1097/cm9.0000000000002329] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cancer immunotherapy has emerged as a promising strategy against triple-negative breast cancer (TNBC). One of the immunosuppressive pathways involves programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1), but many patients derived little benefit from PD-1/PD-L1 checkpoint blockades treatment. Prior research has shown that MYC, a master transcription amplifier highly expressed in TNBC cells, can regulate the tumor immune microenvironment and constrain the efficacy of immunotherapy. This study aims to investigate the regulatory relationship between MYC and PD-L1, and whether a cyclin-dependent kinase (CDK) inhibitor that inhibits MYC expression in combination with anti-PD-L1 antibodies can enhance the response to immunotherapy. METHODS Public databases and TNBC tissue microarrays were used to study the correlation between MYC and PD-L1. The expression of MYC and PD-L1 in TNBCs was examined by quantitative real-time polymerase chain reaction and Western blotting. A patient-derived tumor xenograft (PDTX) model was used to evaluate the influence of a CDK7 inhibitor THZ1 on PD-L1 expression. Cell proliferation and migration were detected by 5-ethynyl-2'-deoxyuridine (EdU) cell proliferation and cell migration assays. Tumor xenograft models were established for in vivo verification. RESULTS A high MYC expression level was associated with a poor prognosis and could alter the proportion of tumor-infiltrating immune cells (TIICs). The positive correlation between MYC and PD-L1 was confirmed by immunostaining samples from 165 TNBC patients. Suppression of MYC in TNBC caused a reduction in the levels of both PD-L1 messenger RNA and protein. In addition, antitumor immune response was enhanced in the TNBC cancer xenograft mouse model with suppression of MYC by CDK7 inhibitor THZ1. CONCLUSIONS The combined therapy of CDK7 inhibitor THZ1 and anti-PD-L1 antibody appeared to have a synergistic effect, which might offer new insight for enhancing immunotherapy in TNBC.
Collapse
Affiliation(s)
- Xintong Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lin Tang
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Qin Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xumin Cheng
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yiqiu Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Cenzhu Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chengjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Kun Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Fangyan Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jinyi Huang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Runtian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
273
|
Avila J, Leone JP. Advances in the Management of Central Nervous System Metastases from Breast Cancer. Int J Mol Sci 2022; 23:12525. [PMID: 36293379 PMCID: PMC9604332 DOI: 10.3390/ijms232012525] [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] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
Central nervous system (CNS) metastases are common in breast cancer (BC) patients and are particularly relevant as new treatments for BC are prolonging survival. Here, we review advances in the treatment of CNS metastases from BC, including radiotherapy, systemic therapies, and the evolving role of immunotherapy. The use of radiotherapy and chemotherapy is the cornerstone of treatment for CNS metastases. However, new targeted therapies have recently been developed, including anti-HER2 agents and antibody-drug conjugates that have presented promising results for the treatment of these patients.
Collapse
Affiliation(s)
- Jorge Avila
- Department of Internal Medicine, St Elizabeth’s Medical Center, 736 Cambridge St., Boston, MA 02135, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - José Pablo Leone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215, USA
| |
Collapse
|
274
|
Sun K, Xu Y, Zhang L, Niravath P, Darcourt J, Patel T, Teh BS, Farach AM, Guerrero C, Mathur S, Sultenfuss MA, Gupta N, Schwartz MR, Haley SL, Nair S, Li X, Nguyen TTA, Butner JD, Ensor J, Mejia JA, Mei Z, Butler EB, Chen SH, Bernicker EH, Chang JC. A Phase 2 Trial of Enhancing Immune Checkpoint Blockade by Stereotactic Radiation and In Situ Virus Gene Therapy in Metastatic Triple-Negative Breast Cancer. Clin Cancer Res 2022; 28:4392-4401. [PMID: 35877117 PMCID: PMC9561553 DOI: 10.1158/1078-0432.ccr-22-0622] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/06/2022] [Accepted: 07/21/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE A Phase 2 trial of stereotactic radiotherapy and in situ cytotoxic virus therapy in patients with metastatic triple-negative breast cancer (mTNBC) followed by pembrolizumab (STOMP) was designed to evaluate dual approach of enhancing single-agent immune checkpoint blockade with adenovirus-mediated expression of herpes-simplex-virus thymidine-kinase (ADV/HSV-tk) plus valacyclovir gene therapy and stereotactic body radiotherapy (SBRT) in patients with mTNBC. PATIENTS AND METHODS In this single-arm, open-label Phase 2 trial, patients with mTNBC were treated with ADV/HSV-tk [5 × 1011 virus particles (vp)] intratumoral injection, followed by SBRT to the injected tumor site, then pembrolizumab (200 mg, every 3 weeks). The primary endpoint was clinical benefit rate [CBR; complete response (CR), partial response (PR), or stable disease (SD) ≥ 24 weeks per RECIST version1.1 at non-irradiated site]. Secondary endpoints included duration on treatment (DoT), overall survival (OS), and safety. Exploratory endpoints included immune response to treatment assessed by correlative tissue and blood-based biomarkers. RESULTS Twenty-eight patients were enrolled and treated. CBR was seen in 6 patients (21.4%), including 2 CR (7.1%), 1 PR (3.6%), and 3 SD (10.7%). Patients with clinical benefit had durable responses, with median DoT of 9.6 months and OS of 14.7 months. The median OS was 6.6 months in the total population. The combination was well tolerated. Correlative studies with Cytometry by Time of Flight (CyTOF) and imaging mass cytometry (IMC) revealed a significant increase of CD8 T cells in responders and of myeloid cells in non-responders. CONCLUSIONS The median OS increased by more than 2-fold in patients with clinical benefit. The therapy is a well-tolerated treatment in heavily pretreated patients with mTNBC. Early detection of increased effector and effector memory CD8 T cells and myeloids correlate with response and non-response, respectively.
Collapse
Affiliation(s)
- Kai Sun
- Houston Methodist Neal Cancer Center, Houston, Texas
| | - Yitian Xu
- Houston Methodist Research Institute, Center for Immunotherapy Research, Houston, Texas
| | - Licheng Zhang
- Houston Methodist Research Institute, Center for Immunotherapy Research, Houston, Texas
| | | | | | - Tejal Patel
- Houston Methodist Neal Cancer Center, Houston, Texas
| | - Bin S. Teh
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas
| | - Andrew M. Farach
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas
| | | | - Sunil Mathur
- Houston Methodist Neal Cancer Center, Houston, Texas
| | | | - Nakul Gupta
- Department of Radiology, Houston Methodist Hospital, Houston, Texas
| | - Mary R. Schwartz
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Susan L. Haley
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Sindhu Nair
- Houston Methodist Neal Cancer Center, Houston, Texas
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Thi Truc Anh Nguyen
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas
| | - Joe Ensor
- Houston Methodist Neal Cancer Center, Houston, Texas
| | | | - Zhuyong Mei
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - E. Brian Butler
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas
| | - Shu-hsia Chen
- Houston Methodist Research Institute, Center for Immunotherapy Research, Houston, Texas
| | | | - Jenny C. Chang
- Houston Methodist Neal Cancer Center, Houston, Texas.,Corresponding Author: Jenny C. Chang, Houston Methodist Research Institute, 6445 Main Street, Floor 24, Houston, TX 77030. Phone: 713-441-9948; Fax: 713-441-8791; E-mail:
| |
Collapse
|
275
|
Myricetin-induced apoptosis in triple-negative breast cancer cells through inhibition of the PI3K/Akt/mTOR pathway. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:248. [PMID: 36209343 DOI: 10.1007/s12032-022-01856-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/17/2022] [Indexed: 10/10/2022]
Abstract
Breast cancer is still a severe origin of malignant demise in females, and its prevalence is rising worldwide. Triple-negative breast cancer (TNBC) is a diversified aggressive breast tumor distinguished by inadequate prognosis, early recurrence, high invasion, and extremely metastasized disease. Chemotherapy is being used to treat it; however, it has low efficacy. On the other hand, with the growing number of corroborations on subtypes of TNBC and molecular biology of tumors, significant advancement in TNBC targeted treatment has been made. Myricetin (MYR), a polyhydroxyflavonol compound widely found in nature, has been shown to possess anticancer effects in various cancers. Though, the mechanisms and impacts of MYR on metastasis of TNBC remain unclear. Early and late apoptotic cell death and cell proliferation inhibition were observed in MYR-treated TNBC cells. MYR modulated cell cycle, pro-angiogenic, and invasion effects via the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Protein kinase B (PKB/also known as AKT) signaling pathways. Moreover, it regulates the expression of MAPK, PI3K/AKT/mTOR, IκB/NF-κB, Hippo, STAT3, GSK-3β, Nrf2/HO-1, TLR, eNOS / NO, ACE, and AChE. Here, we review the anticancer effects of MYR for TNBC and target the PI3K/AKT/mTOR pathway as a therapeutic target for the fruitful treatment of TNBC to summarize MYR's therapeutic potential.
Collapse
|
276
|
Kaleem M, Dalhat MH, Azmi L, Asar TO, Ahmad W, Alghanmi M, Almostadi A, Zughaibi TA, Tabrez S. An Insight into Molecular Targets of Breast Cancer Brain Metastasis. Int J Mol Sci 2022; 23:ijms231911687. [PMID: 36232989 PMCID: PMC9569595 DOI: 10.3390/ijms231911687] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Brain metastasis is one of the major reasons of death in breast cancer (BC) patients, significantly affecting the quality of life, physical activity, and interdependence on several individuals. There is no clear evidence in scientific literature that depicts an exact mechanism relating to brain metastasis in BC patients. The tendency to develop breast cancer brain metastases (BCBMs) differs by the BC subtype, varying from almost half with triple-negative breast cancer (TNBC) (HER2- ER- PR-), one-third with HER2+ (human epidermal growth factor receptor 2-positive, and around one-tenth with luminal subclass (ER+ (estrogen positive) or PR+ (progesterone positive)) breast cancer. This review focuses on the molecular pathways as possible therapeutic targets of BCBMs and their potent drugs under different stages of clinical trial. In view of increased numbers of clinical trials and systemic studies, the scientific community is hopeful of unraveling the underlying mechanisms of BCBMs that will help in designing an effective treatment regimen with multiple molecular targets.
Collapse
Affiliation(s)
- Mohammed Kaleem
- Department of Pharmacology, Faculty of Pharmacy, Dadasaheb Balpande College of Pharmacy, Nagpur 440037, India
| | - Mahmood Hassan Dalhat
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Lubna Azmi
- Department of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Turky Omar Asar
- Department of Biology, College of Science and Arts at Alkamil, University of Jeddah, Jeddah 23218, Saudi Arabia
| | - Wasim Ahmad
- Department of Kuliyate Tib, National Institute of Unani Medicine, Kottigepalya, Bengaluru 560091, India
| | - Maimonah Alghanmi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amal Almostadi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Torki A. Zughaibi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence:
| |
Collapse
|
277
|
Xiong J, Wu R, He A, Hou P, Wang J, Zhang R, Liao W, Wu L, Li E. Comprehensive analysis of the effects of KIF2C on prognosis, biological functions and immune infiltration in PAAD. Tissue Cell 2022; 78:101900. [DOI: 10.1016/j.tice.2022.101900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
|
278
|
Lian J, Zhang G, Zhang Y, Liu H, Zhang J, Nan P, Tian W. PD-L1 and HER2 expression in gastric adenocarcinoma and their prognostic significance. Dig Liver Dis 2022; 54:1419-1427. [PMID: 35123909 DOI: 10.1016/j.dld.2022.01.128] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 01/10/2022] [Accepted: 01/16/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND The upregulation of programmed death-ligand 1 (PD-L1) and epidermal growth factor receptor 2 (HER2) may play a role in gastric adenocarcinoma (GAC). AIM To study PD-L1 and HER-2 expression and prognosis in GAC. METHODS PD-L1 and HER2 expression was determined in tumor tissues of 75 patients with GAC. The correlations between PD-L1, HER2 expression, and clinicopathological factors were analyzed. RESULTS The positive expression rate for PD-L1 was 57.3% (43/75) and the HER2 over-expression rate was 17.3% (13/75). PD-L1 expression negatively correlated with the grade of GAC differentiation (r =-0.26, P<0.05). Approximately 85% of HER2-positive GACs were found to be PD-L1-positive and PD-L1 expression positively correlated with HER2 overexpression. The TNM stage and combined HER2 and PD-L1 expression were independent prognostic factors affecting the survival of patients with GAC. The median overall survival and recurrence-free survival of groups I (HER2 overexpression and PD-L1 positive), II (HER2 overexpression and PD-L1 negative), III (No HER2 overexpression and PD-L1 positive) and IV (No HER2 overexpression and PD-L1 negative) were (47 (17-77), 15 (0-44), 81 (62-101), and 78 (60-98) months, respectively. CONCLUSION PD-L1 expression is upregulated in more than half of patients with GAC. Anti-PD-L1 treatment combined with anti-HER2 therapy may benefit patients with locally advanced GAC with HER2 overexpression.
Collapse
Affiliation(s)
- Jie Lian
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Guanjun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China.
| | - Yun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Heng Liu
- Department of Radiology, Affiliated Hospital of Zunyi Medical University, Medical Imaging Center of Guizhou Province, Zunyi, Guizhou 563003, P.R. China
| | - Jiaojiao Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Pengfei Nan
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Wei Tian
- Department of Pathology, No. 215 Hospital of Shanxi Nuclear Industry, Xianyang 712000, China
| |
Collapse
|
279
|
Shafi S, Aung TN, Xirou V, Gavrielatou N, Vathiotis IA, Fernandez A, Moutafi M, Yaghoobi V, Herbst RS, Liu LN, Langermann S, Rimm DL. Quantitative assessment of Siglec-15 expression in lung, breast, head, and neck squamous cell carcinoma and bladder cancer. J Transl Med 2022; 102:1143-1149. [PMID: 35581307 PMCID: PMC10211373 DOI: 10.1038/s41374-022-00796-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/09/2022] Open
Abstract
Immune checkpoint blockade with programmed cell death (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors has resulted in significant progress in the treatment of various cancer types. However, not all patients respond to PD-1/PD-L1 blockade, underscoring the importance of identifying new potential targets for immunotherapy. One promising target is the immune system modulator Siglec-15. In this study, we assess Siglec-15 expression in solid tumors, with a focus on lung, breast, head and neck squamous and bladder cancers. Using quantitative immunofluorescence (QIF) with a previously validated antibody, we found increased Siglec-15 expression in both tumor and immune cells in all the four cancer types. Siglec-15 was seen to be predominantly expressed by the stromal immune cells (83% in lung, 70.1% in breast, 95.2% in head and neck squamous cell and 89% in bladder cancers). Considerable intra-tumoral heterogeneity was noted across cancer types. As previously described for non-small cell lung cancer (NSCLC), Siglec-15 expression was seen to be mutually exclusive to PD-L1 in all the four cancer types, although this differential expression was maintained but somewhat diminished in head and neck squamous cell carcinoma (HNSCC). Siglec-15 was not prognostic either for overall survival (OS) or progression-free survival (PFS). In summary, we show broad expression of this potential immune modulatory target in a wide range of cancer types. These data suggest potential future clinical trials in these tumor types.
Collapse
Affiliation(s)
- Saba Shafi
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Thazin Nwe Aung
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Vasiliki Xirou
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Niki Gavrielatou
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Ioannis A Vathiotis
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aileen Fernandez
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Myrto Moutafi
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Vesal Yaghoobi
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Roy S Herbst
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
280
|
Li RQ, Wang W, Yan L, Song LY, Guan X, Zhang W, Lian J. Identification of tumor antigens and immune subtypes in breast cancer for mRNA vaccine development. Front Oncol 2022; 12:973712. [PMID: 36226063 PMCID: PMC9548593 DOI: 10.3389/fonc.2022.973712] [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: 06/20/2022] [Accepted: 09/07/2022] [Indexed: 12/04/2022] Open
Abstract
Background Poor prognosis, resistance to chemotherapy, insensitivity to radiotherapy, and a high prevalence of adverse drug reactions remain urgent issues for breast cancer (BC) patients. Increased knowledge of tumor immunobiology and vaccine development suggests the possibility of cancer vaccination. Here, we investigated potential BC-associated antigens for the development of an anti-BC mRNA vaccine and populations suitable for mRNA vaccination. Methods Gene expression and clinical data were obtained from The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC). The single-cell sequencing data were obtained from the Single Cell Portal platform. cBioPortal was used to visualize and compare genetic alterations. Correlations between immune cell infiltration and antigen expression were visualized with the Tumor Immune Estimation Resource (TIMER). Immune subtypes were identified by consensus clustering and analysis of immune infiltration. Biomarkers for the assessment of mRNA vaccination suitability were investigated. Results Three tumor-associated antigens, CD74, IRF1, and PSME2, that showed overexpression, amplification, and mutation and were linked with prognosis and immune cell infiltration, were identified. Single-cell sequencing analysis showed the expression of the three tumor-associated antigens in different cells of BC. Three immune subtypes were identified among BC patients, with Cluster B patients having a tumor microenvironment conducive to immunotherapy. These subtypes also showed different expression patterns of immune checkpoints, immune cell death-promoting genes, and response to immune checkpoint inhibitor (ICI) therapy. Thus, we identified five biomarkers that could be applied for assessing vaccination suitability and predicted drugs that would be appropriate for patients unsuited for vaccination. Conclusions Our findings suggest new directions for the development of mRNA vaccines against breast cancer.
Collapse
Affiliation(s)
- Ruo Qi Li
- Department of Pathology, Cancer Hospital Affiliated to Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Wei Wang
- Department of Urologic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Lei Yan
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Li Ying Song
- Thyroid Surgery Department, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xin Guan
- Cardiovascular Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Wei Zhang
- Department of Pathology, Cancer Hospital Affiliated to Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jing Lian
- Department of Pathology, Cancer Hospital Affiliated to Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- *Correspondence: Jing Lian,
| |
Collapse
|
281
|
Gao Y, Liu M, Li L, Zhang J, Song F, Tian J. Immune checkpoint inhibitors and chemotherapy versus chemotherapy for early triple-negative breast cancer. Hippokratia 2022. [DOI: 10.1002/14651858.cd015072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Ya Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences; Lanzhou University; Lanzhou City China
| | - Ming Liu
- Evidence-Based Medicine Center, School of Basic Medical Sciences; Lanzhou University; Lanzhou City China
| | - Lun Li
- Department of Breast Cancer; The Second Xiangya Hospital, Central South University; Changsha China
| | - Junhua Zhang
- Evidence-Based Medicine Center; Tianjin University of Traditional Chinese Medicine; Tianjin China
| | - Fujian Song
- Norwich Medical School; University of East Anglia; Norwich UK
| | - JinHui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences; Lanzhou University; Lanzhou City China
| |
Collapse
|
282
|
MCT4/Lactate Promotes PD-L1 Glycosylation in Triple-Negative Breast Cancer Cells. JOURNAL OF ONCOLOGY 2022; 2022:3659714. [PMID: 36199799 PMCID: PMC9529401 DOI: 10.1155/2022/3659714] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/30/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022]
Abstract
Triple-negative breast cancer (TNBC) has the highest percentage of lymphocytic infiltration among breast cancer subtypes, and TNBC patients may benefit from anti-PD-1/PD-L1 immunotherapy. However, some cases whether the immune checkpoint blockade (ICB) shows low targeting efficiency have occurred and effective synergistic targets need to be found, which inspired our exploration of the co-expression analysis of MCT4 (SLC16A3) and PD-L1 (CD274) and their potential regulatory mechanisms. After bioinformatic analysis of the relationship between MCT4 and PD-L1, we validated their positive co-expression relationship in triple-negative breast cancer through multiple immunohistochemical staining (mIHC), CRISPR/Cas9, and lentiviral transduction for MCT4 knockout (sgMCT4/231 KO) or overexpression (pEGFP-N1-MCT4/231). We examined the effect of lactate treatment on PD-L1 expression in triple-negative breast cancer cells by qRT-PCR and Western blot. Combined with our results, we found that MCT4 positively regulated PD-L1 expression through discharging lactate and stabilized PD-L1 through promoting its glycosylation by the classic WNT pathway in MDA-MB-231 cells. More importantly, the high co-expression of MCT4 and PD-L1 appears to predict more effective targets for treating TNBC, which would improve immune checkpoint therapy for TNBC.
Collapse
|
283
|
Gadot M, Arad I, Atenafu EG, Levartovsky M, Portnoy O, Davidson T, Schor-Bardach R, Berger R, Leibowitz R. Response to Anti-PD1/L1 Antibodies in Advanced Urothelial Cancer in the 'Real-Life' Setting. Pharmaceuticals (Basel) 2022; 15:1154. [PMID: 36145376 PMCID: PMC9504505 DOI: 10.3390/ph15091154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/31/2022] [Accepted: 09/09/2022] [Indexed: 12/04/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are now the standard of care for metastatic urothelial carcinoma (mUC) patients. Our aim was to describe the activity of ICIs in mUC and find the clinical parameters associated with response. This is a retrospective, single-center chart review of mUC patients receiving ICIs. The overall survival (OS) was plotted using the Kaplan-Meier method and was compared using a log-rank test. Associations between the variables and responses were analyzed by univariate and multivariable analyses, using either logistic regression or a Chi-square/Fisher's exact test. Ninety-four patients received ICIs, 85% of which were in the second line or beyond; the median age was 71.8 years, and 82% were men. Six (6.4%), 11 (11.7%), 7 (7.4%) and 70 (74.5%) patients achieved a complete response (CR), partial response (PR), mixed response/stable disease (M/SD) or progressive disease (PD), respectively. The median overall survival was 3.2 months for the entire cohort and was significantly different according to the response pattern-not reached, 32.3, 6.4 and 2.0 months for CR, PR, M/SD and PD, respectively. The response was not significantly associated with the line of treatment. 'Site of metastasis' was associated with the response, and the absolute neutrophil count was borderline associated with the response. In summary, we found a substantial variance in the potential benefit from ICIs in mUC, emphasizing the need for predictive biomarkers and frequent monitoring of mUC patients receiving ICIs.
Collapse
Affiliation(s)
- Moran Gadot
- Sheba Medical Center, Oncology Institute, Tel-Hashomer 52621, Israel
| | - Ido Arad
- Sheba Medical Center, Oncology Institute, Tel-Hashomer 52621, Israel
| | - Eshetu G. Atenafu
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | | | - Orith Portnoy
- Sheba Medical Center, Diagnostic Imaging Department, Tel-Hashomer 52621, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 699781, Israel
| | - Tima Davidson
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 699781, Israel
- Department of Nuclear Medicine, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | | | - Raanan Berger
- Sheba Medical Center, Oncology Institute, Tel-Hashomer 52621, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 699781, Israel
| | - Raya Leibowitz
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 699781, Israel
- Shamir Medical Center, Oncology Institute, Zerifin 70300, Israel
| |
Collapse
|
284
|
Abstract
PURPOSE OF REVIEW Recently, immune checkpoint inhibitors (ICI) have demonstrated survival benefits in triple-negative breast cancer (TNBC) patients, treated in both the advanced and the early settings. RECENT FINDINGS As monotherapy, ICI failed to demonstrate a superiority over chemotherapy in pretreated advanced TNBC. In the first-line setting, ICI in combination with chemotherapy have shown consistent gains in progression-free survival in programmed death-ligand 1-positive TNBC, but only pembrolizumab indisputably demonstrated a significant overall survival benefit. In early-stage TNBC patients treated with neoadjuvant chemotherapy (NAC), ICI may improve the pathological complete response (pCR) rate. In the KEYNOTE-522 trial enrolling stage II to III TNBC patients, pembrolizumab, in combination with a NAC composed of carboplatin-paclitaxel followed by anthracyclines, and continued in the adjuvant phase led to significant increases in both pCR and disease-free survival, a practice-changing result in the field. Importantly, no unexpected safety signal was observed, but the possibility of definitive ICI-related toxicities may be challenging in curable early disease. SUMMARY Immunotherapy is now an important component in the therapeutic management of TNBC. Unresolved issues include the best chemotherapy partners, additional biomarkers to maximize the clinical benefit, and the possible extension of its use to other breast cancer subtypes.
Collapse
|
285
|
Bohm MS, Sipe LM, Pye ME, Davis MJ, Pierre JF, Makowski L. The role of obesity and bariatric surgery-induced weight loss in breast cancer. Cancer Metastasis Rev 2022; 41:673-695. [PMID: 35870055 PMCID: PMC9470652 DOI: 10.1007/s10555-022-10050-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023]
Abstract
Obesity is a complex metabolic condition considered a worldwide public health crisis, and a deeper mechanistic understanding of obesity-associated diseases is urgently needed. Obesity comorbidities include many associated cancers and are estimated to account for 20% of female cancer deaths in the USA. Breast cancer, in particular, is associated with obesity and is the focus of this review. The exact causal links between obesity and breast cancer remain unclear. Still, interactions have emerged between body mass index, tumor molecular subtype, genetic background, and environmental factors that strongly suggest obesity influences the risk and progression of certain breast cancers. Supportive preclinical research uses various diet-induced obesity models to demonstrate that weight loss, via dietary interventions or changes in energy expenditure, reduces the onset or progression of breast cancers. Ongoing and future studies are now aimed at elucidating the underpinning mechanisms behind weight-loss-driven observations to improve therapy and outcomes in patients with breast cancer and reduce risk. This review aims to summarize the rapidly emerging literature on obesity and weight loss strategies with a focused discussion of bariatric surgery in both clinical and preclinical studies detailing the complex interactions between metabolism, immune response, and immunotherapy in the setting of obesity and breast cancer.
Collapse
Affiliation(s)
- Margaret S Bohm
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Laura M Sipe
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Madeline E Pye
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Matthew J Davis
- Division of Bariatric Surgery, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Joseph F Pierre
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Department of Nutritional Sciences, College of Agriculture and Life Science, The University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Liza Makowski
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- College of Medicine, UTHSC Center for Cancer Research, The University of Tennessee Health Science Center, Cancer Research Building Room 322, 19 S Manassas Street, Memphis, TN, 38163, USA.
| |
Collapse
|
286
|
Li Y, Zhang H, Merkher Y, Chen L, Liu N, Leonov S, Chen Y. Recent advances in therapeutic strategies for triple-negative breast cancer. J Hematol Oncol 2022; 15:121. [PMID: 36038913 PMCID: PMC9422136 DOI: 10.1186/s13045-022-01341-0] [Citation(s) in RCA: 391] [Impact Index Per Article: 130.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 01/03/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer (BC) with a poor prognosis. Current treatment options are limited to surgery, adjuvant chemotherapy and radiotherapy; however, a proportion of patients have missed the surgical window at the time of diagnosis. TNBC is a highly heterogeneous cancer with specific mutations and aberrant activation of signaling pathways. Hence, targeted therapies, such as those targeting DNA repair pathways, androgen receptor signaling pathways, and kinases, represent promising treatment options against TNBC. In addition, immunotherapy has also been demonstrated to improve overall survival and response in TNBC. In this review, we summarize recent key advances in therapeutic strategies based on molecular subtypes in TNBC.
Collapse
Affiliation(s)
- Yun Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huajun Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yulia Merkher
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700
| | - Lin Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700. .,Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia, 142290.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
287
|
Liu J, Ma J, Zhang Q. Identification of the pyroptosis-related prognostic gene signature and characterization of tumor microenvironment infiltration in triple-negative breast cancer. Front Genet 2022; 13:929870. [PMID: 36092879 PMCID: PMC9453819 DOI: 10.3389/fgene.2022.929870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Triple-negative breast cancer remains a highly malignant disease due to the lack of specific targeted therapy and immunotherapy. A growing body of evidence supports the role of pyroptosis in tumorigenesis and prognosis, but further exploration is needed to improve our understanding of the tumor microenvironment in patients with triple-negative breast cancer.Methods: Consensus clustering analysis was performed to construct pattern clusters. A correlation analysis was conducted between the pattern clusters and the tumor microenvironment using GSVA, ESTIMATE, and CIBERSORT. Then, a risk score and a nomogram were constructed and verified to predict overall survival.Results: Two pyro-clusters and three pyro-gene clusters that differed significantly in terms of prognosis, biological processes, clinical features, and tumor microenvironment were identified. The different clusters corresponded to different immune expression profiles. The constructed risk score predicted patient prognosis and response to immunotherapy. Patients with low risk scores exhibited favorable outcomes with increased immune cell infiltration and expression of immune checkpoint molecules. Compared to other models, the nomogram was extremely effective in predicting prognosis.Conclusion: In the landscape of the immune microenvironment, pyroptosis-mediated pattern clusters differed markedly. Both the developed risk score and the nomogram were effective predictive models. These findings could help develop customized treatment for patients with triple-negative breast cancer.
Collapse
Affiliation(s)
- Ji Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianli Ma
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
- *Correspondence: Qingyuan Zhang,
| |
Collapse
|
288
|
Ribeiro R, Carvalho MJ, Goncalves J, Moreira JN. Immunotherapy in triple-negative breast cancer: Insights into tumor immune landscape and therapeutic opportunities. Front Mol Biosci 2022; 9:903065. [PMID: 36060249 PMCID: PMC9437219 DOI: 10.3389/fmolb.2022.903065] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/13/2022] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a clinically aggressive subtype of breast cancer that represents 15-20% of breast tumors and is more prevalent in young pre-menopausal women. It is the subtype of breast cancers with the highest metastatic potential and recurrence at the first 5 years after diagnosis. In addition, mortality increases when a complete pathological response is not achieved. As TNBC cells lack estrogen, progesterone, and HER2 receptors, patients do not respond well to hormone and anti-HER2 therapies, and conventional chemotherapy remains the standard treatment. Despite efforts to develop targeted therapies, this disease continues to have a high unmet medical need, and there is an urgent demand for customized diagnosis and therapeutics. As immunotherapy is changing the paradigm of anticancer treatment, it arises as an alternative treatment for TNBC patients. TNBC is classified as an immunogenic subtype of breast cancer due to its high levels of tumor mutational burden and presence of immune cell infiltrates. This review addresses the implications of these characteristics for the diagnosis, treatment, and prognosis of the disease. Herein, the role of immune gene signatures and tumor-infiltrating lymphocytes as biomarkers in TNBC is reviewed, identifying their application in patient diagnosis and stratification, as well as predictors of efficacy. The expression of PD-L1 expression is already considered to be predictive of response to checkpoint inhibitor therapy, but the challenges regarding its value as biomarker are described. Moreover, the rationales for different formats of immunotherapy against TNBC currently under clinical research are discussed, and major clinical trials are highlighted. Immune checkpoint inhibitors have demonstrated clinical benefit, particularly in early-stage tumors and when administered in combination with chemotherapy, with several regimens approved by the regulatory authorities. The success of antibody-drug conjugates and research on other emerging approaches, such as vaccines and cell therapies, will also be addressed. These advances give hope on the development of personalized, more effective, and safe treatments, which will improve the survival and quality of life of patients with TNBC.
Collapse
Affiliation(s)
- Rita Ribeiro
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Coimbra, Portugal
- iMed.ULisboa—Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Coimbra, Portugal
| | - Maria João Carvalho
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Coimbra, Portugal
- CHUC—Coimbra Hospital and University Centre, Department of Gynaecology, Coimbra, Portugal
- Univ Coimbra—University Clinic of Gynaecology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- iCBR—Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - João Goncalves
- iMed.ULisboa—Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - João Nuno Moreira
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Coimbra, Portugal
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Coimbra, Portugal
| |
Collapse
|
289
|
Wesolowski J, Tankiewicz-Kwedlo A, Pawlak D. Modern Immunotherapy in the Treatment of Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14163860. [PMID: 36010854 PMCID: PMC9406094 DOI: 10.3390/cancers14163860] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary This review summarizes reports from the latest clinical trials assessing the safety and clinical effectiveness of new biological drugs stimulating the immune system to fight cancer. The aim of this study is to show the enormous therapeutic potential of monoclonal antibodies in the treatment of cancer, in particular triple negative breast cancer (TNBC). Introduction of these innovative drugs to the standard clinical cancer therapies, including TNBC, allows for an increase in the response rate to the applied treatment, and consequently extending the lives of patients suffering from cancer. We hope to draw attention to the extremely difficult-to-treat TNBC, as well as the importance of the development of clinical trials evaluating drugs modulating the immune system in TNBC therapy. Abstract Triple-Negative Breast Cancer is a subtype of breast cancer characterized by the lack of expression of estrogen receptors, progesterone receptors, as well as human epidermal growth factor receptor 2. This cancer accounts for 15–20% of all breast cancers and is especially common in patients under 40 years of age, as well as with the occurring BRCA1 mutation. Its poor prognosis is reflected in the statistical life expectancy of 8–15 months after diagnosis of metastatic TNBC. So far, the lack of targeted therapy has narrowed therapeutic possibilities to classic chemotherapy. The idea behind the use of humanized monoclonal antibodies, as inhibitors of immunosuppressive checkpoints used by the tumor to escape from immune system control, is to reduce immunotolerance and direct an intensified anti-tumor immune response. An abundance of recent studies has provided numerous pieces of evidence about the safety and clinical benefits of immunotherapy using humanized monoclonal antibodies in the fight against many types of cancer, including TNBC. In particular, phase three clinical trials, such as the IMpassion 130, the KEYNOTE-355 and the KEYNOTE-522 resulted in the approval of immunotherapeutic agents, such as atezolizumab and pembrolizumab by the US Food and Drug Administration in TNBC therapy. This review aims to present the huge potential of immunotherapy using monoclonal antibodies directed against immunosuppressive checkpoints—such as atezolizumab, avelumab, durvalumab, pembrolizumab, nivolumab, cemiplimab, tremelimumab, ipilimumab—in the fight against difficult to treat TNBCs as monotherapy as well as in more advanced combination strategies.
Collapse
Affiliation(s)
- Jakub Wesolowski
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University in Bialystok, 15-089 Bialystok, Poland
- Correspondence:
| | - Anna Tankiewicz-Kwedlo
- Department of Monitored Pharmacotherapy, Faculty of Pharmacy, Medical University in Bialystok, 15-089 Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University in Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
290
|
Zhang C, Liu H, Tan Y, Xu Y, Li Y, Tong S, Qiu S, Chen Q, Su Z, Tian D, Zhou W, Zhong C. MS4A6A is a new prognostic biomarker produced by macrophages in glioma patients. Front Immunol 2022; 13:865020. [PMID: 36119086 PMCID: PMC9472524 DOI: 10.3389/fimmu.2022.865020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 07/04/2022] [Indexed: 12/12/2022] Open
Abstract
MS4A6A has been recognized as being associated with aging and the onset of neurodegenerative disease. However, the mechanisms of MS4A6A in glioma biology and prognosis are ill-defined. Here, we show that MS4A6A is upregulated in glioma tissues, resulting in unfavorable clinical outcomes and poor responses to adjuvant chemotherapy. Multivariate Cox regression analysis suggested that MS4A6A expression can act as a strong and independent predictor for glioma outcomes (CGGA1: HR: 1.765, p < 0.001; CGGA2: HR: 2.626, p < 0.001; TCGA: HR: 1.415, p < 0.001; Rembrandt: HR: 1.809, p < 0.001; Gravendeel: HR: 1.613, p < 0.001). A protein–protein interaction (PPI) network revealed that MS4A6A might be coexpressed with CD68, CD163, and macrophage-specific signatures. Enrichment analysis showed the innate immune response and inflammatory response to be markedly enriched in the high MS4A6A expression group. Additionally, single-cell RNA sequencing (scRNA-seq) analysis revealed distinctive expression features for MS4A6A in macrophages in the glioma immune microenvironment (GIME). Immunofluorescence staining confirmed colocalization of CD68/MS4A6A and CD163/MS4A6A in macrophages. Correlation analysis revealed that MS4A6A expression is positively related to the tumor mutation burden (TMB) of glioma, displaying the high potential of applying MS4A6A to evaluate responsiveness to immunotherapy. Altogether, our research indicates that MS4A6A upregulation may be used as a promising and effective indicator for adjuvant therapy and prognosis assessment.
Collapse
Affiliation(s)
- Chunyu Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou Normal University, Huzhou, China
| | - Haitao Liu
- Department of Cardiothoracic Surgery, Jiaxing University, The First Affiliated Hospital, Jiaxing, China
| | - Yinqiu Tan
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Xu
- Department of Neurosurgery, Wuhan University, Renmin Hospital, Wuhan, China
| | - Yuntao Li
- Department of Neurosurgery, Wuhan University, Renmin Hospital, Wuhan, China
| | - Shiao Tong
- Department of Neurosurgery, Wuhan University, Renmin Hospital, Wuhan, China
| | - Sheng Qiu
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou Normal University, Huzhou, China
| | - Qianxue Chen
- Department of Neurosurgery, Wuhan University, Renmin Hospital, Wuhan, China
| | - Zhongzhou Su
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou Normal University, Huzhou, China
| | - Daofeng Tian
- Department of Neurosurgery, Wuhan University, Renmin Hospital, Wuhan, China
- *Correspondence: Daofeng Tian, ; Chunlong Zhong, ; Wei Zhou,
| | - Wei Zhou
- Department of Anesthesia, Huzhou Central Hospital, Affiliated Central Hospital Huzhou Normal University, Huzhou, China
- *Correspondence: Daofeng Tian, ; Chunlong Zhong, ; Wei Zhou,
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Daofeng Tian, ; Chunlong Zhong, ; Wei Zhou,
| |
Collapse
|
291
|
Mulcahy CF, Gross ND. Advances in Surgical Therapy for HPV-Associated Squamous Cell Carcinoma. CURRENT OTORHINOLARYNGOLOGY REPORTS 2022. [DOI: 10.1007/s40136-022-00421-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
292
|
Lai HZ, Han JR, Fu X, Ren YF, Li ZH, You FM. Targeted Approaches to HER2-Low Breast Cancer: Current Practice and Future Directions. Cancers (Basel) 2022; 14:cancers14153774. [PMID: 35954438 PMCID: PMC9367369 DOI: 10.3390/cancers14153774] [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: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary HER2-low breast cancer (BC) accounts for more than half of breast cancer patients. Anti-HER2 therapy has been ineffective in HER2-low BC, for which palliative chemotherapy is the main treatment modality. The definitive efficacy of T-Dxd in HER2-low BC breaks previous treatment strategies, which will redefine HER2-low and thus reshape anti-HER2 therapy. This review summarizes detection technologies and novel agents for HER2-low BC, and explores their possible role in future clinics, to provide ideas for the diagnosis and treatment of HER2-low BC. Abstract HER2-low breast cancer (BC) has a poor prognosis, making the development of more suitable treatment an unmet clinical need. While chemotherapy is the main method of treatment for HER2-low BC, not all patients benefit from it. Antineoplastic therapy without chemotherapy has shown promise in clinical trials and is being explored further. As quantitative detection techniques become more advanced, they assist in better defining the expression level of HER2 and in guiding the development of targeted therapies, which include directly targeting HER2 receptors on the cell surface, targeting HER2-related intracellular signaling pathways and targeting the immune microenvironment. A new anti-HER2 antibody-drug conjugate called T-DM1 has been successfully tested and found to be highly effective in clinical trials. With this progress, it could eventually be transformed from a disease without a defined therapeutic target into a disease with a defined therapeutic molecular target. Furthermore, efforts are being made to compare the sequencing and combination of chemotherapy, endocrine therapy, and HER2-targeted therapy to improve prognosis to customize the subtype of HER2 low expression precision treatment regimens. In this review, we summarize the current and upcoming treatment strategies, to achieve accurate management of HER2-low BC.
Collapse
|
293
|
Cao J, Wang B, Zhang J, Tao Z, Wang L, Hu X. Phase 1b clinical trial of pucotenlimab (HX008), a novel anti-PD-1 monoclonal antibody, combined with gemcitabine and cisplatin in the first-line treatment of metastatic triple-negative breast cancer. Front Oncol 2022; 12:837963. [PMID: 35982961 PMCID: PMC9379318 DOI: 10.3389/fonc.2022.837963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundPucotenlimab, also called HX008, is a humanized anti-PD-1 antagonist IgG4 mAb. It blocks programmed cell death protein 1 (PD-1), programmed-death ligand 1 (PD-L1), and programmed death ligand-2 (PD-L2). In the CBCSG 006 trial, gemcitabine plus cisplatin (GP) has shown impressive antitumor activity as first-line therapy for metastatic triple-negative breast cancer (mTNBC). The phase 1b study was conducted to assess the safety and preliminary antitumor activity of pucotenlimab when combined with GP in patients with mTNBC in the first-line setting.MethodsEligible patients with mTNBC with ≥6 months of DFI (disease-free interval) who have never received antitumor therapy for metastatic disease were screened. Participants received pucotenlimab at 3 mg/kg (d1, q3w) plus gemcitabine at 1,250 mg/m2 (d1, 8, q3w) and cisplatin at 75 mg/m2 (d1, q3w). Eligible patients received up to six cycles of pucotenlimab along with GP chemotherapy, while pucotenlimab could be maintained until disease progression or unacceptable toxicity occurred or withdrawal of informed consent. This study was registered in China under registration number CTR20191353.ResultsBetween July 2019 and March 2020, 31 patients were enrolled in this study. The median age was 50 (range 28–68) years. Among 31 patients who were evaluated, 25 (80.6%) experienced objective response and the other six (19.4%) experienced stable disease (SD). As of 4 August, the median progression-free survival (PFS) was 9.0 months (95% CI, 6.2–9.2). The most common grade 3 or 4 treatment-related adverse events included neutropenia (74.1%), anemia (35.5%), thrombocytopenia (32.3%), hypocalcemia (9.7%), hypokalemia (9.7%), and alanine aminotransferase increased (6.5%). There were no treatment-related deaths.ConclusionPucotenlimab plus GP demonstrated promising activity and a manageable safety profile in patients with mTNBC in the first-line setting.
Collapse
Affiliation(s)
- Jun Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Leiping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Xichun Hu,
| |
Collapse
|
294
|
Howard FM, Pearson AT, Nanda R. Clinical trials of immunotherapy in triple-negative breast cancer. Breast Cancer Res Treat 2022; 195:1-15. [PMID: 35834065 PMCID: PMC9338129 DOI: 10.1007/s10549-022-06665-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/23/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE Immunotherapy has started to transform the treatment of triple-negative breast cancer (TNBC), in part due to the unique immunogenicity of this breast cancer subtype. This review summarizes clinical studies of immunotherapy in advanced and early-stage TNBC. FINDINGS Initial studies of checkpoint blockade monotherapy demonstrated occasional responses, especially in patients with untreated programmed death-ligand 1 (PD-L1) positive advanced TNBC, but failed to confirm a survival advantage over chemotherapy. Nonetheless, pembrolizumab monotherapy has tumor agnostic approval for microsatellite instability-high or high tumor mutational burden cancers, and thus can be considered for select patients with advanced TNBC. Combination chemoimmunotherapy approaches have been more successful, and pembrolizumab is approved for PD-L1 positive advanced TNBC in combination with chemotherapy. This success has been translated to the curative setting, where pembrolizumab is now approved in combination with neoadjuvant chemotherapy for high-risk early-stage TNBC. CONCLUSION Immunotherapy has been a welcome addition to the growing armamentarium for TNBC, but responses remain limited to a subset of patients. Innovative strategies are under investigation in an attempt to induce immune responses in resistant tumors-with regimens incorporating small-molecule inhibitors, novel immune checkpoint targets, and intratumoral injections that directly alter the tumor microenvironment. As the focus shifts toward the use of immunotherapy for early-stage TNBC, it will be critical to identify those who derive the most benefit from treatment, given the potential for irreversible autoimmune toxicity and the lack of predictive accuracy of PD-L1 expression in the early-stage setting.
Collapse
Affiliation(s)
- Frederick M Howard
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, 5841 S. Maryland Ave MC 2115, Chicago, IL, 60637, USA.
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, 5841 S. Maryland Ave MC 2115, Chicago, IL, 60637, USA
| | - Rita Nanda
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, 5841 S. Maryland Ave MC 2115, Chicago, IL, 60637, USA
| |
Collapse
|
295
|
Zhang X, Ge X, Jiang T, Yang R, Li S. Research progress on immunotherapy in triple‑negative breast cancer (Review). Int J Oncol 2022; 61:95. [PMID: 35762339 PMCID: PMC9256074 DOI: 10.3892/ijo.2022.5385] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
Triple‑negative breast cancer (TNBC) is a highly heterogeneous and aggressive malignancy. Due to the absence of estrogen receptors and progesterone receptors and the lack of overexpression of human epidermal growth factor receptor 2, TNBC responds poorly to endocrine and targeted therapies. As a neoadjuvant therapy, chemotherapy is usually the only option for TNBC; however, chemotherapy may induce tumor resistance. The emergence of immunotherapy as an adjuvant therapy is expected to make up for the deficiency of chemotherapy. Most of the research on immunotherapies has been performed on advanced metastatic TNBC, which has provided significant clinical benefits. In the present review, possible immunotherapy targets and ongoing immunotherapy strategies were discussed. In addition, progress in research on immune checkpoint inhibitors in early TNBC was outlined.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xueying Ge
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Tinghan Jiang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Ruming Yang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
296
|
Zheng D, Hou X, Yu J, He X. Combinatorial Strategies With PD-1/PD-L1 Immune Checkpoint Blockade for Breast Cancer Therapy: Mechanisms and Clinical Outcomes. Front Pharmacol 2022; 13:928369. [PMID: 35935874 PMCID: PMC9355550 DOI: 10.3389/fphar.2022.928369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
As an emerging antitumor strategy, immune checkpoint therapy is one of the most promising anticancer therapies due to its long response duration. Antibodies against the programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1) axis have been extensively applied to various cancers and have demonstrated unprecedented efficacy. Nevertheless, a poor response to monotherapy with anti-PD-1/PD-L1 has been observed in metastatic breast cancer. Combination therapy with other standard treatments is expected to overcome this limitation of PD-1/PD-L1 blockade in the treatment of breast cancer. In the present review, we first illustrate the biological functions of PD-1/PD-L1 and their role in maintaining immune homeostasis as well as protecting against immune-mediated tissue damage in a variety of microenvironments. Several combination therapy strategies for the combination of PD-1/PD-L1 blockade with standard treatment modalities have been proposed to solve the limitations of anti-PD-1/PD-L1 treatment, including chemotherapy, radiotherapy, targeted therapy, antiangiogenic therapy, and other immunotherapies. The corresponding clinical trials provide valuable estimates of treatment effects. Notably, several combination options significantly improve the response and efficacy of PD-1/PD-L1 blockade. This review provides a PD-1/PD-L1 clinical trial landscape survey in breast cancer to guide the development of more effective and less toxic combination therapies.
Collapse
Affiliation(s)
- Dan Zheng
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Xiaolin Hou
- Department of Neurosurgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Yu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Xiujing He
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
- *Correspondence: Xiujing He,
| |
Collapse
|
297
|
Bhura N, Gupta P, Gupta J. Target-based in-silico screening of basil polysaccharides against different epigenetic targets responsible for breast cancer. J Recept Signal Transduct Res 2022; 42:521-530. [PMID: 35862239 DOI: 10.1080/10799893.2022.2058016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE Breast cancer (BC) is one of the leading types of cancer found in women. One of the causes reported for BC is improper regulation of epigenetic modifications. Various epigenetic targets such as histone deacetylases (HDAC) and histone acetyltransferases (HAT) regulate many types of cancer, including BC. Basil is known to possess anti-cancer properties; however, the role of its polysaccharides against different epigenetic targets is still not very clear. Therefore, the molecular docking method is used to find out the binding potential of the BPSs against different epigenetic targets responsible for BC. METHODS All the basil polysaccharides (BPSs) were screened against the diverse epigenetic targets reported for BC (HDAC1-2, 4-8, and HAT) using molecular docking studies alongwith swissADME studies to check the drug likeliness of the BPSs. RESULTS It was found that glucosamine ring, glucosamine linear, glucuronic acid linear, rhamnose linear, glucuronic acid ring, galactose ring, mannose, glucose, and xylose were exhibited consistent binding potential against the epigenetic targets (HDAC1, HDAC2, HDAC4, HDAC5, HDAC6, HDAC7, HDAC8, and HAT,) responsible for BC. CONCLUSION This is the first report where BPSs were reported against these epigenetic targets. These studies can help to understand the underlying mechanism of BPSs used against epigenetic targets for BC. These results can be further validated experimentally to confirm their potential as a promising inhibitor against the epigenetic targets (HDAC1-2, 4-8, and HAT) having a role in BC.
Collapse
Affiliation(s)
- Nancy Bhura
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Pawan Gupta
- Department of Research and Development, Lovely Professional University, Phagwara, Punjab, India.,Department of Pharmacology, Shree SK Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, Gujarat, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
298
|
Cortes J, Rugo HS, Cescon DW, Im SA, Yusof MM, Gallardo C, Lipatov O, Barrios CH, Perez-Garcia J, Iwata H, Masuda N, Torregroza Otero M, Gokmen E, Loi S, Guo Z, Zhou X, Karantza V, Pan W, Schmid P. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N Engl J Med 2022; 387:217-226. [PMID: 35857659 DOI: 10.1056/nejmoa2202809] [Citation(s) in RCA: 521] [Impact Index Per Article: 173.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND In an interim analysis of this phase 3 trial, the addition of pembrolizumab to chemotherapy resulted in longer progression-free survival than chemotherapy alone among patients with advanced triple-negative breast cancer whose tumors expressed programmed death ligand 1 (PD-L1) with a combined positive score (CPS; the number of PD-L1-staining tumor cells, lymphocytes, and macrophages, divided by the total number of viable tumor cells, multiplied by 100) of 10 or more. The results of the final analysis of overall survival have not been reported. METHODS We randomly assigned patients with previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer in a 2:1 ratio to receive pembrolizumab (200 mg) every 3 weeks plus the investigator's choice of chemotherapy (nanoparticle albumin-bound paclitaxel, paclitaxel, or gemcitabine-carboplatin) or placebo plus chemotherapy. The primary end points were progression-free survival (reported previously) and overall survival among patients whose tumors expressed PD-L1 with a CPS of 10 or more (the CPS-10 subgroup), among patients whose tumors expressed PD-L1 with a CPS of 1 or more (the CPS-1 subgroup), and in the intention-to-treat population. Safety was also assessed. RESULTS A total of 847 patients underwent randomization: 566 were assigned to the pembrolizumab-chemotherapy group, and 281 to the placebo-chemotherapy group. The median follow-up was 44.1 months. In the CPS-10 subgroup, the median overall survival was 23.0 months in the pembrolizumab-chemotherapy group and 16.1 months in the placebo-chemotherapy group (hazard ratio for death, 0.73; 95% confidence interval [CI], 0.55 to 0.95; two-sided P = 0.0185 [criterion for significance met]); in the CPS-1 subgroup, the median overall survival was 17.6 and 16.0 months in the two groups, respectively (hazard ratio, 0.86; 95% CI, 0.72 to 1.04; two-sided P = 0.1125 [not significant]); and in the intention-to-treat population, the median overall survival was 17.2 and 15.5 months, respectively (hazard ratio, 0.89; 95% CI, 0.76 to 1.05 [significance not tested]). Adverse events of grade 3, 4, or 5 that were related to the trial regimen occurred in 68.1% of the patients in the pembrolizumab-chemotherapy group and in 66.9% in the placebo-chemotherapy group, including death in 0.4% of the patients in the pembrolizumab-chemotherapy group and in no patients in the placebo-chemotherapy group. CONCLUSIONS Among patients with advanced triple-negative breast cancer whose tumors expressed PD-L1 with a CPS of 10 or more, the addition of pembrolizumab to chemotherapy resulted in significantly longer overall survival than chemotherapy alone. (Funded by Merck Sharp and Dohme; KEYNOTE-355 ClinicalTrials.gov number, NCT02819518.).
Collapse
Affiliation(s)
- Javier Cortes
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Hope S Rugo
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - David W Cescon
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Seock-Ah Im
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Mastura M Yusof
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Carlos Gallardo
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Oleg Lipatov
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Carlos H Barrios
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Jose Perez-Garcia
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Hiroji Iwata
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Norikazu Masuda
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Marco Torregroza Otero
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Erhan Gokmen
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Sherene Loi
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Zifang Guo
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Xuan Zhou
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Vassiliki Karantza
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Wilbur Pan
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| | - Peter Schmid
- From the International Breast Cancer Center, Pangaea Oncology, Quirónsalud Group, Barcelona (J.C., J.P.-G.), and the Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid (J.C.) - both in Spain; the Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco (H.S.R.); Princess Margaret Cancer Centre, Toronto (D.W.C.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul (S.-A.I.); Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia (M.M.Y.); the Oncology Institute, Arturo Lopez Perez Foundation, Santiago, Chile (C.G.); the Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia (O.L.); the Oncology Research Unit, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil (C.H.B.); the Department of Breast Oncology, Aichi Cancer Center Hospital (H.I.), and the Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine (N.M.) - both in Nagoya, Japan; the Department of Hematology and Oncology, Oncomedica, Montería, Colombia (M.T.O.); Ege University Medical Faculty, Izmir, Turkey (E.G.); the Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville - both in Australia (S.L.); Merck, Rahway, NJ (Z.G., X.Z., V.K., W.P.); and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London (P.S.)
| |
Collapse
|
299
|
Ma S, Zhao Y, Lee WC, Ong LT, Lee PL, Jiang Z, Oguz G, Niu Z, Liu M, Goh JY, Wang W, Bustos MA, Ehmsen S, Ramasamy A, Hoon DSB, Ditzel HJ, Tan EY, Chen Q, Yu Q. Hypoxia induces HIF1α-dependent epigenetic vulnerability in triple negative breast cancer to confer immune effector dysfunction and resistance to anti-PD-1 immunotherapy. Nat Commun 2022; 13:4118. [PMID: 35840558 PMCID: PMC9287350 DOI: 10.1038/s41467-022-31764-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
The hypoxic tumor microenvironment has been implicated in immune escape, but the underlying mechanism remains elusive. Using an in vitro culture system modeling human T cell dysfunction and exhaustion in triple-negative breast cancer (TNBC), we find that hypoxia suppresses immune effector gene expression, including in T and NK cells, resulting in immune effector cell dysfunction and resistance to immunotherapy. We demonstrate that hypoxia-induced factor 1α (HIF1α) interaction with HDAC1 and concurrent PRC2 dependency causes chromatin remolding resulting in epigenetic suppression of effector genes and subsequent immune dysfunction. Targeting HIF1α and the associated epigenetic machinery can reverse the immune effector dysfunction and overcome resistance to PD-1 blockade, as demonstrated both in vitro and in vivo using syngeneic and humanized mice models. These findings identify a HIF1α-mediated epigenetic mechanism in immune dysfunction and provide a potential strategy to overcome immune resistance in TNBC. Hypoxia can promote tumor escape from immune surveillance and immunotherapy. Here, the authors show that hypoxia induces T and NK cell dysfunction through HIF1α-mediated epigenetic suppression of effector gene expression, conferring resistance to anti-PD1 blockade in triple negative breast cancer models.
Collapse
Affiliation(s)
- Shijun Ma
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Yue Zhao
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Wee Chyan Lee
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Li-Teng Ong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Puay Leng Lee
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Zemin Jiang
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Gokce Oguz
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Zhitong Niu
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Min Liu
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Jian Yuan Goh
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Wenyu Wang
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Matias A Bustos
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Providence Health System, Santa Monica, CA, 90404, USA
| | - Sidse Ehmsen
- Department of Oncology, Odense University Hospital, Odense, 5230, Denmark
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Dave S B Hoon
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Providence Health System, Santa Monica, CA, 90404, USA
| | - Henrik J Ditzel
- Department of Oncology, Odense University Hospital, Odense, 5230, Denmark.,Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, 5230, Denmark
| | - Ern Yu Tan
- Department of General Surgery, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
| | - Qiang Yu
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore. .,Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
300
|
Zhou J, Feng C, Huang K. LCOR Reverses Immune-Checkpoint Inhibitors Therapy Resistance Out of IFN Constraint in Triple-Negative Breast Cancer. Front Oncol 2022; 12:911572. [PMID: 35912168 PMCID: PMC9326162 DOI: 10.3389/fonc.2022.911572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jialin Zhou
- School of Medicine, Shandong University, Jinan, China
| | - Chun Feng
- The Center of Rehabilitation Therapy, The First Rehabilitation Hospital of Shanghai, Rehabilitation Hospital Affiliated to Tongji University, Shanghai, China
| | - Kai Huang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Kai Huang,
| |
Collapse
|