1
|
Garg S, Rai G, Singh S, Gauba P, Ali J, Dang S. An insight into the role of innate immune cells in breast tumor microenvironment. Breast Cancer 2024:10.1007/s12282-024-01645-8. [PMID: 39460874 DOI: 10.1007/s12282-024-01645-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
The immune background of breast cancer is highly heterogeneous and the immune system of the human body plays a dual role by both promoting and suppressing its progression. Innate immune cells are the first line of defense in the immune system and impart protection by identifying and interacting with foreign pathogens and cancer cells. Different innate immune cells like natural killer cells, macrophages, dendritic cells, and myeloid suppressor cells take part in hosting the cancer cells. Autophagy is another key component inside the tumor microenvironment and is linked to the disintegration and recycling of cellular components. Within the tumor microenvironment autophagy is involved with Pattern Recognition Receptors and inflammation. Various clinical studies have shown prominent results where innate immune cells and autophagy in combination are used for pathogen as well as cancer cell clearance. However, it is necessary to comprehend the complex tumor microenvironment so that different therapeutic approaches can be developed to enhance the suppressive actions of the cells toward breast cancer cells. In this review article, the complex interaction between immune cells and breast cancer cells and their role in developing effective immunotherapies to improve patient outcomes are discussed in detail.
Collapse
Affiliation(s)
- Sandini Garg
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India
| | - Garima Rai
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India
| | - Sakshi Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India
| | - Pammi Gauba
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India.
| |
Collapse
|
2
|
Saastad SA, Skjervold AH, Ytterhus B, Engstrøm MJ, Bofin AM. PD-L1 protein expression in breast cancer. J Clin Pathol 2024; 77:730-736. [PMID: 37553245 DOI: 10.1136/jcp-2023-208942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023]
Abstract
AIMS The immune checkpoint marker, Programmed cell death-ligand 1 (PD-L1), is expressed by both cancer epithelial cells and tumour-infiltrating immune cells (TICs) thus constituting a potential target for immunotherapy. This is of particular interest in triple negative breast cancer. In this study, we assessed the prognostic value of PD-L1 expression in tumour epithelial cells and TICs in a series of patients with breast cancer with long-term follow-up, and associations between PD-L1 expression and histopathological type and grade, proliferation and molecular subtype. METHODS Using immunohistochemistry for PD-L1 in tissue microarrays, we assessed PD-L1 expression in 821 tumours. Expression of PD-L1 was assessed separately in the epithelial and stromal compartments and classified as <1%, ≥1% to <10% or ≥10% positive staining cells. We correlated PD-L1 expression in tumour epithelial cells and TICs with tumour characteristics using Pearson's χ2 test, and prognosis by cumulative incidence of death from breast cancer and Cox regression analyses. RESULTS We found membranous staining in ≥1% of tumour epithelial cells in 53/821 cases (6.5%). Of these, 21 (2.6%) were ≥10%. Among TICs, staining (≥1%) was seen in 144/821 cases (17.6%). Of these, 62 were ≥10% (7.6%). PD-L1 was associated with high histopathological grade and proliferation, and the medullary and metaplastic patterns. In TICs, PD-L1 ≥1% found in 22/34 (34.4%) human epidermal growth factor receptor 2 type and 29/58 (50%) basal phenotype. An independent association between PD-L1 expression and prognosis was not observed. CONCLUSIONS PD-L1 is expressed more frequently in TICs than tumour epithelial cells. Expression in TICs is associated with aggressive tumour characteristics and non-luminal tumours but not with prognosis.
Collapse
Affiliation(s)
- Sigurd A Saastad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anette H Skjervold
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Monica Jernberg Engstrøm
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Sugrery, St. Olav's Hospital Trondheim University Hospital, Trondheim, Norway
| | - Anna M Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
3
|
Xiong X, Zhang Y, Huang X, Zhang S, Li Q. Generating Immunological Memory Against Cancer by Camouflaging Gold-Based Photothermal Nanoparticles in NIR-II Biowindow for Mimicking T-Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407038. [PMID: 39394989 DOI: 10.1002/smll.202407038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Indexed: 10/14/2024]
Abstract
Photothermal therapy (PTT) against cancer not only directly ablates tumors but also induces tumor immunogenic cell death (ICD). However, the antitumor immune response elicited by ICD is insufficient to prevent relapse and metastasis because of the immunosuppressive tumor microenvironment (TME). A biomimetic nanoplatform (bmNP) mimicking cytotoxic lymphocytes (CTLs) for combinational photothermal-immunotherapy to effectively regulate the immunosuppressive TME is reported here. The bmNP is constructed by wrapping the T-cell membrane onto a new type of photothermal agents, spherical Au-based PNCs (sAuPNCs). Similar to T-cells, the bmNP enhanced accumulation at the tumor site by targeting the tumor via adhesion proteins on T-cell membrane. The obtained sAuPNCs have a wide absorption band in the second near-infrared (NIR-II) region with a high photothermal conversion efficiency (PCE) up to about 75% and excellent photostability. The bmNP with a smaller size is more superior compete with T-cells to bond with tumor cells via PD-1/PD-L1 interaction to effectively block the PD-1 checkpoint of T-cells for preventing T-cell exhaustion. Furthermore, in vivo studies reveal the immunological memory effect is significantly elicited in mice received bmNPs therapy. Collectively, bmNPs show great potential in photothermal-enhanced immunotherapy.
Collapse
Affiliation(s)
- Xuefan Xiong
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, P. R. China
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi, 276005, P. R. China
| | - Ying Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, P. R. China
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi, 276005, P. R. China
| | - Xinqi Huang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, P. R. China
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi, 276005, P. R. China
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, P. R. China
| | - Qiong Li
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, P. R. China
| |
Collapse
|
4
|
Li G, Ma X, Sui S, Chen Y, Li H, Liu L, Zhang X, Zhang L, Hao Y, Yang Z, Yang S, He X, Wang Q, Tao W, Xu S. NAT10/ac4C/JunB facilitates TNBC malignant progression and immunosuppression by driving glycolysis addiction. J Exp Clin Cancer Res 2024; 43:278. [PMID: 39363363 PMCID: PMC11451012 DOI: 10.1186/s13046-024-03200-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND N4-Acetylcytidine (ac4C), a highly conserved post-transcriptional mechanism, plays a pivotal role in RNA modification and tumor progression. However, the molecular mechanism by which ac4C modification mediates tumor immunosuppression remains elusive in triple-negative breast cancer (TNBC). METHODS NAT10 expression was analyzed in TNBC samples in the level of mRNA and protein, and compared with the corresponding normal tissues. ac4C modification levels also measured in the TNBC samples. The effects of NAT10 on immune microenvironment and tumor metabolism were investigated. NAT10-mediated ac4C and its downstream regulatory mechanisms were determined in vitro and in vivo. The combination therapy of targeting NAT10 in TNBC was further explored. RESULTS The results revealed that the loss of NAT10 inhibited TNBC development and promoted T cell activation. Mechanistically, NAT10 upregulated JunB expression by increasing ac4C modification levels on its mRNA. Moreover, JunB further up-regulated LDHA expression and facilitated glycolysis. By deeply digging, remodelin, a NAT10 inhibitor, elevated the surface expression of CTLA-4 on T cells. The combination of remodelin and CTLA-4 mAb can further activate T cells and inhibite tumor progression. CONCLUSION Taken together, our study demonstrated that the NAT10-ac4C-JunB-LDHA pathway increases glycolysis levels and creates an immunosuppressive tumor microenvironment (TME). Consequently, targeting this pathway may assist in the identification of novel therapeutic strategies to improve the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Guozheng Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xin Ma
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Shiyao Sui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Yihai Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
- National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Hui Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Lei Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xin Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Lei Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
- National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zihan Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Shuai Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xu He
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China
- Weihan Yu Academy, Harbin Medical University, Harbin, 150086, China
| | - Qin Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China.
- Key Laboratory of Tumor Biotherapy of Heilongjiang Province, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Weiyang Tao
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150040, China.
- Key Laboratory of Tumor Biotherapy of Heilongjiang Province, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
5
|
Monteran L, Ershaid N, Scharff Y, Zoabi Y, Sanalla T, Ding Y, Pavlovsky A, Zait Y, Langer M, Caller T, Eldar-Boock A, Avivi C, Sonnenblick A, Satchi-Fainaro R, Barshack I, Shomron N, Zhang XHF, Erez N. Combining TIGIT Blockade with MDSC Inhibition Hinders Breast Cancer Bone Metastasis by Activating Antitumor Immunity. Cancer Discov 2024; 14:1252-1275. [PMID: 38427556 DOI: 10.1158/2159-8290.cd-23-0762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 01/17/2024] [Accepted: 02/28/2024] [Indexed: 03/03/2024]
Abstract
Bone is the most common site of breast cancer metastasis. Bone metastasis is incurable and is associated with severe morbidity. Utilizing an immunocompetent mouse model of spontaneous breast cancer bone metastasis, we profiled the immune transcriptome of bone metastatic lesions and peripheral bone marrow at distinct metastatic stages, revealing dynamic changes during the metastatic process. We show that cross-talk between granulocytes and T cells is central to shaping an immunosuppressive microenvironment. Specifically, we identified the PD-1 and TIGIT signaling axes and the proinflammatory cytokine IL1β as central players in the interactions between granulocytes and T cells. Targeting these pathways in vivo resulted in attenuated bone metastasis and improved survival, by reactivating antitumor immunity. Analysis of patient samples revealed that TIGIT and IL1β are prominent in human bone metastasis. Our findings suggest that cotargeting immunosuppressive granulocytes and dysfunctional T cells may be a promising novel therapeutic strategy to inhibit bone metastasis. Significance: Temporal transcriptome profiling of the immune microenvironment in breast cancer bone metastasis revealed key communication pathways between dysfunctional T cells and myeloid derived suppressor cells. Cotargeting of TIGIT and IL1β inhibited bone metastasis and improved survival. Validation in patient data implicated these targets as a novel promising approach to treat human bone metastasis.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nour Ershaid
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ye'ela Scharff
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yazeed Zoabi
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamer Sanalla
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Anna Pavlovsky
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Zait
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marva Langer
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Caller
- Tamman Cardiovascular Research Institute, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Camila Avivi
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Sonnenblick
- Oncology Division, Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Neta Erez
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
6
|
Wu M, Wang S, Yuan K, Xiong B, Li Y, Lyu S. Alteration of the immune microenvironment in the axillary metastatic lymph nodes of luminal A breast cancer patients. World J Surg Oncol 2024; 22:172. [PMID: 38937736 PMCID: PMC11210032 DOI: 10.1186/s12957-024-03454-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/16/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND The alteration of the immune microenvironment in the axillary metastatic lymph nodes of luminal A breast cancer patients is still unclear. METHODS Postsurgical tissues from the enrolled luminal A BCs were divided into five categories: primary BC lesion at stage N0 (PL1), primary BC lesion at stage N1 (PL2), negative axillary lymph node at stage N0 BC (LN1), negative axillary lymph node at stage N1 BC (LN2), and positive axillary lymph node at stage N1 BC (LN3). The frequencies of positive immune markers (CD4, CD8, PD1, PD-L1, T-cell immunoglobulin and mucin domain 3 (TIM3), and forkhead box protein 3 (Foxp3)) in the above tissues were quantified by AKOYA Opal Polaris 7 Color Manual IHC Detection Kit. RESULTS A total of 50 female patients with luminal A BC were enrolled in this study. Among these patients, 23 had stage N1 disease, and 27 had stage N0 disease. Compared with that in the PL2 subgroup, the frequency of PD-1-positive cells was significantly greater in the PL1 subgroup, whether at the stromal or intratumoral level (P value < 0.05). Both the frequency of CD8 + T cells in LN1 and that in LN2 were significantly greater than that in LN3 (P value < 0.05). The frequency of TIM3 + T cells in LN1 was significantly greater than that in PL1 (P value < 0.05). The frequency of CD8 + TIM3 + T cells was significantly greater in both the LN2 and LN3 groups than in the PL2 group (P value < 0.05). The frequency of CD4 + Foxp3 + T cells was significantly greater in LN1 than in PL1 (P value < 0.05), which was the same for both LN3 and PL2 (P value < 0.05). CONCLUSION Increased frequencies of CD8 + PD1+, CD8 + TIM3 + and CD4 + Foxp3 + T cells might inhibit the immune microenvironment of axillary metastatic lymph nodes in luminal A breast cancer patients and subsequently promote lymph node metastasis.
Collapse
Affiliation(s)
- Min Wu
- Department of Breast Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyi Road 10, Haidian District, Beijing, 100038, China
| | - Shuo Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing, 100038, China
| | - Keyu Yuan
- Department of Breast Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyi Road 10, Haidian District, Beijing, 100038, China
| | - Bingjun Xiong
- Department of Breast Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyi Road 10, Haidian District, Beijing, 100038, China
| | - Yanping Li
- Department of Breast Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyi Road 10, Haidian District, Beijing, 100038, China
| | - Shuzhen Lyu
- Department of Breast Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyi Road 10, Haidian District, Beijing, 100038, China.
| |
Collapse
|
7
|
Ridnour LA, Cheng RY, Kedei N, Somasundaram V, Bhattacharyya DD, Basudhar D, Wink AL, Walke AJ, Kim C, Heinz WF, Edmondson EF, Butcher DO, Warner AC, Dorsey TH, Pore M, Kinders RJ, Lipkowitz S, Bryant RJ, Rittscher J, Wong ST, Hewitt SM, Chang JC, Shalaby A, Callagy GM, Glynn SA, Ambs S, Anderson SK, McVicar DW, Lockett SJ, Wink DA. Adjuvant COX inhibition augments STING signaling and cytolytic T cell infiltration in irradiated 4T1 tumors. JCI Insight 2024; 9:e165356. [PMID: 38912586 PMCID: PMC11383366 DOI: 10.1172/jci.insight.165356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 05/08/2024] [Indexed: 06/25/2024] Open
Abstract
Immune therapy is the new frontier of cancer treatment. Therapeutic radiation is a known inducer of immune response and can be limited by immunosuppressive mediators including cyclooxygenase-2 (COX2) that is highly expressed in aggressive triple negative breast cancer (TNBC). A clinical cohort of TNBC tumors revealed poor radiation therapeutic efficacy in tumors expressing high COX2. Herein, we show that radiation combined with adjuvant NSAID (indomethacin) treatment provides a powerful combination to reduce both primary tumor growth and lung metastasis in aggressive 4T1 TNBC tumors, which occurs in part through increased antitumor immune response. Spatial immunological changes including augmented lymphoid infiltration into the tumor epithelium and locally increased cGAS/STING1 and type I IFN gene expression were observed in radiation-indomethacin-treated 4T1 tumors. Thus, radiation and adjuvant NSAID treatment shifts "immune desert phenotypes" toward antitumor M1/TH1 immune mediators in these immunologically challenging tumors. Importantly, radiation-indomethacin combination treatment improved local control of the primary lesion, reduced metastatic burden, and increased median survival when compared with radiation treatment alone. These results show that clinically available NSAIDs can improve radiation therapeutic efficacy through increased antitumor immune response and augmented local generation of cGAS/STING1 and type I IFNs.
Collapse
Affiliation(s)
- Lisa A Ridnour
- Cancer Innovation Laboratory, CCR, NCI, NIH, Frederick, Maryland, USA
| | - Robert Ys Cheng
- Cancer Innovation Laboratory, CCR, NCI, NIH, Frederick, Maryland, USA
| | - Noemi Kedei
- Collaborative Protein Technology Resource (CPTR) Nanoscale Protein Analysis, OSTR, CCR, NCI, NIH, Bethesda, Maryland, USA
| | | | | | | | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, and
| | - Abigail J Walke
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, and
| | - Caleb Kim
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, and
| | - William F Heinz
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, and
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, Maryland, USA
| | - Donna O Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, Maryland, USA
| | - Andrew C Warner
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, Maryland, USA
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, Maryland, USA
| | - Milind Pore
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research, and
| | - Robert J Kinders
- Office of the Director, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, Maryland, USA
| | | | - Richard J Bryant
- Department of Urology, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Jens Rittscher
- Institute of Biomedical Engineering, Big Data Institute, Ludwig Oxford Branch, University of Oxford, Oxford, United Kingdom
| | - Stephen Tc Wong
- Houston Methodist Neal Cancer Center, Weill Cornell Medical College, Houston Methodist Hospital, Houston, Texas, USA
| | | | - Jenny C Chang
- Houston Methodist Neal Cancer Center, Weill Cornell Medical College, Houston Methodist Hospital, Houston, Texas, USA
| | - Aliaa Shalaby
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | - Grace M Callagy
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, Maryland, USA
| | - Stephen K Anderson
- Cancer Innovation Laboratory, CCR, NCI, NIH, Frederick, Maryland, USA
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Daniel W McVicar
- Cancer Innovation Laboratory, CCR, NCI, NIH, Frederick, Maryland, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, and
| | - David A Wink
- Cancer Innovation Laboratory, CCR, NCI, NIH, Frederick, Maryland, USA
| |
Collapse
|
8
|
Buisseret L, Bareche Y, Venet D, Girard E, Gombos A, Emonts P, Majjaj S, Rouas G, Serra M, Debien V, Agostinetto E, Garaud S, Willard-Gallo K, Larsimont D, Stagg J, Rothé F, Sotiriou C. The long and winding road to biomarkers for immunotherapy: a retrospective analysis of samples from patients with triple-negative breast cancer treated with pembrolizumab. ESMO Open 2024; 9:102964. [PMID: 38703428 PMCID: PMC11087916 DOI: 10.1016/j.esmoop.2024.102964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) in combination with chemotherapy improves outcome of patients with triple-negative breast cancer (TNBC) in metastatic and early settings. The identification of predictive biomarkers able to guide treatment decisions is challenging and currently limited to programmed death-ligand 1 (PD-L1) expression and high tumor mutational burden (TMB) in the advanced setting, with several limitations. MATERIALS AND METHODS We carried out a retrospective analysis of clinical-pathological and molecular characteristics of tumor samples from 11 patients with advanced TNBC treated with single-agent pembrolizumab participating in two early-phase clinical trials: KEYNOTE-012 and KEYNOTE-086. Clinical, imaging, pathological [i.e. tumor-infiltrating lymphocytes (TILs), PD-L1 status], RNA sequencing, and whole-exome sequencing data were analyzed. We compared our results with publicly available transcriptomic data from TNBC cohorts from TCGA and METABRIC. RESULTS Response to pembrolizumab was heterogeneous: two patients experienced exceptional long-lasting responses, six rapid progressions, and three relatively slower disease progression. Neither PD-L1 nor stromal TILs were significantly associated with response to treatment. Increased TMB values were observed in tumor samples from exceptional responders compared to the rest of the cohort (P = 3.4 × 10-4). Tumors from exceptional responders were enriched in adaptive and innate immune cell signatures. Expression of regulatory T-cell markers (FOXP3, CCR4, CCR8, TIGIT) was mainly observed in tumors from responders except for glycoprotein-A repetitions predominant (GARP), which was overexpressed in tumors from rapid progressors. GARP RNA expression in primary breast tumors from the public dataset was significantly associated with a worse prognosis. CONCLUSIONS The wide spectrum of clinical responses to ICB supports that TNBC is a heterogeneous disease. Tumors with high TMB respond better to ICB. However, the optimal cut-off of 10 mutations (mut)/megabase (Mb) may not reflect the complexity of all tumor subtypes, despite its approval as a tumor-agnostic biomarker. Further studies are required to better elucidate the relevance of the tumor microenvironment and its components as potential predictive biomarkers in the context of ICB.
Collapse
Affiliation(s)
- L Buisseret
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels; Medical Oncology Department, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
| | - Y Bareche
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal; Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - D Venet
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - E Girard
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels; Centre Oscar Lambret, Lille, France
| | - A Gombos
- Medical Oncology Department, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - P Emonts
- Radiology Department, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - S Majjaj
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - G Rouas
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - M Serra
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - V Debien
- Academic Trials Promoting Team, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - E Agostinetto
- Academic Trials Promoting Team, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - S Garaud
- Molecular Immunology Unit, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - K Willard-Gallo
- Molecular Immunology Unit, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - D Larsimont
- Pathology Department, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - J Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal; Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - F Rothé
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| | - C Sotiriou
- Breast Cancer Translational Research Laboratory J-C Heuson, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels
| |
Collapse
|
9
|
Taghavi BA, Salehi M, Mokhtarzadeh A, Baradaran B. Suppression of B7-H7 Enhanced MCF-7 Cancer Cell Line's Chemosensitivity to Paclitaxel. Mol Biotechnol 2024:10.1007/s12033-024-01145-2. [PMID: 38662256 DOI: 10.1007/s12033-024-01145-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/06/2024] [Indexed: 04/26/2024]
Abstract
The B7-H7 is the newest addition to the B7 family of proteins that is present in the majority of malignancies. In this respect, the goal of the work was to investigate the impact of B7-H7 inhibition on breast cancer cells when paclitaxel and small interference RNA (siRNA) were combined. B7-H7 siRNA was used with Paclitaxel to treat MCF-7 cells. The IC50 of Paclitaxel and the cell survival was then assessed through using MTT assay. Investigation was conducted using flow cytometry to both the induction of apoptosis and the cell cycle. In addition, the clonogenic capacity of MCF-7 cells was investigated. Furthermore, qRT-PCR, was used to evaluate expression of genes. Our results demonstrated that suppressing B7-H7 sensitizes MCF-7 cells to Paclitaxel by triggering apoptosis and altering the expression of critical apoptosis mediator genes. In addition, the cell cycle was stopped in the sub-G1 and also G2-M phases as a result of the combination therapy leading prevention of developing colonies by MCF-7 cells. B7-H7 silencing improved the chemosensitivity of MCF-7 cells to Paclitaxel and demonstrated antiproliferative effects. After the adequate study has been conducted, this strategy may be regarded as a possible alternative treatment option for this cancer.
Collapse
Affiliation(s)
- Bita Amir Taghavi
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mitra Salehi
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St., Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Golgasht St., Tabriz, Iran.
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Sun P, Mo C, Bai L, Wang M, Chen Z, Zhang M, Han Y, Liang H, Tang G. Synthesis and preclinical evaluation of a novel molecular probe [ 18F]AlF-NOTA-PEG 2-Asp 2-PDL1P for PET imaging of PD-L1 positive tumor. Bioorg Chem 2024; 145:107193. [PMID: 38442611 DOI: 10.1016/j.bioorg.2024.107193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 03/07/2024]
Abstract
Immunotherapy has brought great benefits to cancer patients, but only some patients benefit from it. Noninvasive, real-time and dynamic monitoring of the effectiveness of immunotherapy through PET imaging may provide assistance for the treatment plan of immunotherapy. In this study, we designed and synthesized a new targeted PD-L1 peptide NOTA-PEG2-Asp2-PDL1P, which was labeled with nuclide 18F to obtain a new imaging agent [18F]AlF-NOTA-PEG2-Asp2-PDL1P. The total radiochemical yield of [18F]AlF-NOTA-PEG2-Asp2-PDL1P was 13.7 % (Uncorrected radiochemical yield, n > 5). [18F]AlF-NOTA-PEG2-Asp2-PDL1P achieved high radiochemical purity (>95 %) with a molar activity more than 51.2 GBq/μmol. [18F]AlF-NOTA-PEG2-Asp2-PDL1P exhibited good hydrophilicity and had good stability both in vivo and in vitro, it can specifically targets B16F10 tumor with PD-L1 expression, and had a relatively high retention in tumor, a relatively fast clearance in vivo and a higher tumor-to-non-target ratio, all of which could make [18F]AlF-NOTA-PEG2-Asp2-PDL1P a potential tracer for PD-L1 prediction before clinical immunotherapy.
Collapse
Affiliation(s)
- Penghui Sun
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Chunwei Mo
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Lu Bai
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Meng Wang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Zihao Chen
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Meilian Zhang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Yanjiang Han
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Haoran Liang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China
| | - Ganghua Tang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangdong Province, China; Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
11
|
Zhang Y, Deshane JS, Yang ES, Larimer B. A Novel Translational PET Imaging Approach to Quantifying Distal Tumor Immune Activation After Targeted Radiation Therapy and Checkpoint Blockade. Int J Radiat Oncol Biol Phys 2024; 118:1217-1227. [PMID: 38199384 DOI: 10.1016/j.ijrobp.2023.12.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
PURPOSE This study aimed to provide a novel noninvasive method to quantify abscopal immune activation and predict combinational treatment response using [68Ga]-NOTA-GZP positron emission tomography (PET) imaging. METHODS AND MATERIALS 4T1 breast cancer cells were implanted bilaterally in the mammary fat pad of Balb/c mice and Lewis's lung cancer cells (LLC) were implanted bilaterally on the shoulders of C57/Bl6 mice. One of the tumors received a single fraction of 12 Gy irradiation followed by combination of concurrent PD-1 and CTLA-4 inhibitors or controls. Tumor growth of the irradiated and nonirradiated tumors was measured and compared with 12 Gy irradiation only, checkpoint inhibitor only, and no treatment control group. Changes in granzyme B activity were assessed with [68Ga]-NOTA-GZP PET imaging from baseline and every 3 days until day 9. RESULTS In the 4T1 model, concurrent treatment with dual checkpoint inhibitors and radiation resulted in reduction of the irradiated tumor volume at day 30. At this same time point, the nonirradiated tumor volume for combination treatment decreased significantly, consistent with abscopal immune activation. Similarly, in the LLC model, concurrent treatment inhibited tumor growth on the nonirradiated tumor at day 15. On day 9, granzyme B PET signal in both 4T1 and LLC models was significantly higher in the nonirradiated tumors that responded to concurrent treatment compared with subsequent nonresponding tumors. A similar lack of granzyme B signal was observed in the nonirradiated tumors from mice that received radiation or checkpoint inhibitors only and control tumors. Receiver operating characteristic analysis identified a PET threshold of 1.505 and 1.233 on day 9 that predicted treatment response in 4T1 and LLC models, respectively. CONCLUSIONS [68Ga]-NOTA-GZP PET imaging was able to noninvasively predict abscopal immune activation before subsequent tumor volume changes after combination treatment. It provides a potential translational paradigm for investigating distal immune activation postradiation in a clinical setting.
Collapse
Affiliation(s)
- Yujun Zhang
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Jessy S Deshane
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Eddy S Yang
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Benjamin Larimer
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
12
|
Yin J, Gu T, Chaudhry N, Davidson NE, Huang Y. Epigenetic modulation of antitumor immunity and immunotherapy response in breast cancer: biological mechanisms and clinical implications. Front Immunol 2024; 14:1325615. [PMID: 38268926 PMCID: PMC10806158 DOI: 10.3389/fimmu.2023.1325615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024] Open
Abstract
Breast cancer (BC) is the most common non-skin cancer and the second leading cause of cancer death in American women. The initiation and progression of BC can proceed through the accumulation of genetic and epigenetic changes that allow transformed cells to escape the normal cell cycle checkpoint control. Unlike nucleotide mutations, epigenetic changes such as DNA methylation, histone posttranslational modifications (PTMs), nucleosome remodeling and non-coding RNAs are generally reversible and therefore potentially responsive to pharmacological intervention. Epigenetic dysregulations are critical mechanisms for impaired antitumor immunity, evasion of immune surveillance, and resistance to immunotherapy. Compared to highly immunogenic tumor types, such as melanoma or lung cancer, breast cancer has been viewed as an immunologically quiescent tumor which displays a relatively low population of tumor-infiltrating lymphocytes (TIL), low tumor mutational burden (TMB) and modest response rates to immune checkpoint inhibitors (ICI). Emerging evidence suggests that agents targeting aberrant epigenetic modifiers may augment host antitumor immunity in BC via several interrelated mechanisms such as enhancing tumor antigen presentation, activation of cytotoxic T cells, inhibition of immunosuppressive cells, boosting response to ICI, and induction of immunogenic cell death (ICD). These discoveries have established a highly promising basis for using combinatorial approaches of epigenetic drugs with immunotherapy as an innovative paradigm to improve outcomes of BC patients. In this review, we summarize the current understanding of how epigenetic processes regulate immune cell function and antitumor immunogenicity in the context of the breast tumor microenvironment. Moreover, we discuss the therapeutic potential and latest clinical trials of the combination of immune checkpoint blockers with epigenetic agents in breast cancer.
Collapse
Affiliation(s)
- Jun Yin
- The University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tiezheng Gu
- The University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Norin Chaudhry
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Nancy E. Davidson
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, United States
| | - Yi Huang
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
13
|
Liu CC, Wolf M, Ortego R, Grencewicz D, Sadler T, Eng C. Characterization of immunomodulating agents from Staphylococcus aureus for priming immunotherapy in triple-negative breast cancers. Sci Rep 2024; 14:756. [PMID: 38191648 PMCID: PMC10774339 DOI: 10.1038/s41598-024-51361-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024] Open
Abstract
Immunotherapy, specifically immune checkpoint blockade (ICB), has revolutionized the treatment paradigm of triple-negative breast cancers (TNBCs). However, a subset of TNBCs devoid of tumor-infiltrating T cells (TILs) or PD-L1 expression generally has a poor response to immunotherapy. In this study, we aimed to sensitize TNBCs to ICB by harnessing the immunomodulating potential of S. aureus, a breast-resident bacterium. We show that intratumoral injection of spent culture media from S. aureus recruits TILs and suppresses tumor growth in a preclinical TNBC model. We further demonstrate that α-hemolysin (HLA), an S. aureus-produced molecule, increases the levels of CD8+ T cells and PD-L1 expression in tumors, delays tumor growth, and triggers tumor necrosis. Mechanistically, while tumor cells treated with HLA display Gasdermin E (GSDME) cleavage and a cellular phenotype resembling pyroptosis, splenic T cells incubated with HLA lead to selective expansion of CD8+ T cells. Notably, intratumoral HLA injection prior to ICB augments the therapeutic efficacy compared to ICB alone. This study uncovers novel immunomodulatory properties of HLA and suggests that intratumoral administration of HLA could be a potential priming strategy to expand the population of TNBC patients who may respond to ICB.
Collapse
Affiliation(s)
- Chin-Chih Liu
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Matthew Wolf
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Ruth Ortego
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Dennis Grencewicz
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Tammy Sadler
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA
| | - Charis Eng
- Cleveland Clinic, Genomic Medicine Institute, Lerner Research Institute, 9500 Euclid Avenue NE50, Cleveland, OH, 44195, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
- Cleveland Clinic, Center for Personalized Genetic Healthcare, Medical Specialties Institute, Cleveland, OH, 44195, USA.
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, 44195, USA.
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Germline High Risk Cancer Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
14
|
Lama GIG, Noél G, Márquez FCL, Galarza FFG, Hinojosa AIP, Rivera LEN, Willard-Gallo K, Astorga JRA. Concomitant Expression of CD39, CD69, and CD103 Identifies Antitumor CD8 + T Cells in Breast Cancer Implications for Adoptive Cell Therapy. Curr Pharm Biotechnol 2024; 25:1747-1757. [PMID: 37680154 DOI: 10.2174/1389201025666230901094219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/22/2023] [Accepted: 07/20/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND In cancer, an effective immune response involves the action of several different cell types, among which CD8 T cells play a major role as they can specifically recognize and kill cancer cells via the release of cytotoxic molecules and cytokines, being of major importance for adoptive cell transfer (ACT) of ex vivo expanded tumor-infiltrating lymphocytes (TILs). The inflammation resulting from the tumor growth attracts both activated and bystander T cells. For an effective antitumor response, the T cell must express a specific group of chemokine receptors and integrins which include CD103, CD39, CD69, and CD25. These markers had already been analyzed in various cancers, not including breast cancer and their subsequent subtypes, until now. To analyze, the key receptors on ex vivo expanded tumor-infiltrating lymphocytes in luminal A and luminal B breast cancer (BC) subtypes. MATERIALS AND METHODS We were successful in expanding TILs ex vivo using a standard TIL culture condition from a cohort study of 15 primary luminal A and luminal B breast cancer patients. Furthermore, we examined the expression of CD103, CD39, CD69, and CD25 biomarkers after the expansion by flow cytometry. RESULTS We found that the information about the percentage of TILs obtainable after the ex vivo expansion is not associated to nor it is dependent on the heterogeneity of the TIL population before the expansion and does not differ by the molecular subtype (p>0.05). We also found that there is a major population of memory-resident antitumor CD8+CD103+CD39+ and CD8+CD103+ CD69+ TILs present in the stroma after the expansion when compared to CD4 immunosubtypes (p<0.0001). Only the CD8+CD103+CD39+ subpopulation was related to BC subtype (0.0009). CONCLUSION Evidence from our study suggests that CD8 TILs present in the stroma of luminal A and luminal B breast cancer patients can be quantified and phenotyped by flow cytometry and be further expanded ex vivo. The immuno-phenotyping of these markers may be targeted to improve the success of immunotherapeutic approaches, such as adoptive cellular therapy (ACT) in patients with BC.
Collapse
MESH Headings
- Breast Neoplasms/immunology
- Breast Neoplasms/therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Humans
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Integrin alpha Chains/metabolism
- Integrin alpha Chains/immunology
- Female
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Apyrase/metabolism
- Apyrase/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Immunotherapy, Adoptive/methods
- Lectins, C-Type/metabolism
- Lectins, C-Type/immunology
- Middle Aged
- Aged
Collapse
Affiliation(s)
- Grace Ivonne Gattas Lama
- Universidad Iberoamericana de Torreón, Coahuila, México
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | | | - Francisco Carlos López Márquez
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | | | - Adria Imelda Prieto Hinojosa
- Universidad Iberoamericana de Torreón, Coahuila, México
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | - Lydia Enith Nava Rivera
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | - Karen Willard-Gallo
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | - Jesús Rafael Arguëllo Astorga
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
- Instituto de Ciencias y Medicina Genómica, Torreón, Coahuila, México
| |
Collapse
|
15
|
Taifour T, Attalla SS, Zuo D, Gu Y, Sanguin-Gendreau V, Proud H, Solymoss E, Bui T, Kuasne H, Papavasiliou V, Lee CG, Kamle S, Siegel PM, Elias JA, Park M, Muller WJ. The tumor-derived cytokine Chi3l1 induces neutrophil extracellular traps that promote T cell exclusion in triple-negative breast cancer. Immunity 2023; 56:2755-2772.e8. [PMID: 38039967 DOI: 10.1016/j.immuni.2023.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/22/2023] [Accepted: 11/05/2023] [Indexed: 12/03/2023]
Abstract
In triple-negative breast cancer (TNBC), stromal restriction of CD8+ T cells associates with poor clinical outcomes and lack of responsiveness to immune-checkpoint blockade (ICB). To identify mediators of T cell stromal restriction, we profiled murine breast tumors lacking the transcription factor Stat3, which is commonly hyperactive in breast cancers and promotes an immunosuppressive tumor microenvironment. Expression of the cytokine Chi3l1 was decreased in Stat3-/- tumors. CHI3L1 expression was elevated in human TNBCs and other solid tumors exhibiting T cell stromal restriction. Chi3l1 ablation in the polyoma virus middle T (PyMT) breast cancer model generated an anti-tumor immune response and delayed mammary tumor onset. These effects were associated with increased T cell tumor infiltration and improved response to ICB. Mechanistically, Chi3l1 promoted neutrophil recruitment and neutrophil extracellular trap formation, which blocked T cell infiltration. Our findings provide insight into the mechanism underlying stromal restriction of CD8+ T cells and suggest that targeting Chi3l1 may promote anti-tumor immunity in various tumor types.
Collapse
Affiliation(s)
- Tarek Taifour
- McGill University, Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Montreal, QC H4A 3J1, Canada; Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada
| | - Sherif Samer Attalla
- Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada; McGill University, Department of Biochemistry, Faculty of Medicine, Montreal, QC H3A 1A3, Canada
| | - Dongmei Zuo
- Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada
| | - Yu Gu
- Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada; McGill University, Department of Biochemistry, Faculty of Medicine, Montreal, QC H3A 1A3, Canada
| | | | - Hailey Proud
- Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada; McGill University, Department of Biochemistry, Faculty of Medicine, Montreal, QC H3A 1A3, Canada
| | - Emilie Solymoss
- McGill University, Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Montreal, QC H4A 3J1, Canada; Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada
| | - Tung Bui
- Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada
| | - Hellen Kuasne
- Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada
| | | | - Chun Geun Lee
- Brown University, Molecular Biology and Immunology, Faculty of Medicine, Providence, RI 02903, USA
| | - Suchitra Kamle
- Brown University, Molecular Biology and Immunology, Faculty of Medicine, Providence, RI 02903, USA
| | - Peter M Siegel
- McGill University, Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Montreal, QC H4A 3J1, Canada; Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada; McGill University, Department of Biochemistry, Faculty of Medicine, Montreal, QC H3A 1A3, Canada
| | - Jack A Elias
- Brown University, Molecular Biology and Immunology, Faculty of Medicine, Providence, RI 02903, USA
| | - Morag Park
- McGill University, Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Montreal, QC H4A 3J1, Canada; Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada; McGill University, Department of Biochemistry, Faculty of Medicine, Montreal, QC H3A 1A3, Canada
| | - William J Muller
- McGill University, Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Montreal, QC H4A 3J1, Canada; Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada; McGill University, Department of Biochemistry, Faculty of Medicine, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
16
|
Buisseret L, Loirat D, Aftimos P, Maurer C, Punie K, Debien V, Kristanto P, Eiger D, Goncalves A, Ghiringhelli F, Taylor D, Clatot F, Van den Mooter T, Ferrero JM, Bonnefoi H, Canon JL, Duhoux FP, Mansi L, Poncin R, Barthélémy P, Isambert N, Denis Z, Catteau X, Salgado R, Agostinetto E, de Azambuja E, Rothé F, Craciun L, Venet D, Romano E, Stagg J, Paesmans M, Larsimont D, Sotiriou C, Ignatiadis M, Piccart-Gebhart M. Paclitaxel plus carboplatin and durvalumab with or without oleclumab for women with previously untreated locally advanced or metastatic triple-negative breast cancer: the randomized SYNERGY phase I/II trial. Nat Commun 2023; 14:7018. [PMID: 37919269 PMCID: PMC10622534 DOI: 10.1038/s41467-023-42744-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023] Open
Abstract
Chemo-immunotherapy is the first-line standard of care for patients with PD-L1 positive metastatic triple-negative breast cancer (mTNBC). SYNERGY (NCT03616886) is a dose-finding phase I and a randomized phase II, open-label trial evaluating if targeting the immunosuppressive adenosine pathway can enhance the antitumor activity of chemo-immunotherapy. The phase I part included 6 patients with untreated locally-advanced or mTNBC to determine the safety and recommended phase II dose of the anti-CD73 antibody oleclumab in combination with the anti-PD-L1 durvalumab and 12 cycles of weekly carboplatin and paclitaxel. In the phase II part, 127 women were randomized 1:1 to receive chemo-immunotherapy, with (arm A) or without (arm B) oleclumab. The primary endpoint was the clinical benefit rate at week 24, defined as stable disease, partial or complete response per RECIST v1.1. Secondary endpoints included objective response rate, duration of response, survival outcomes (progression-free survival and overall survival), and safety. The trial did not meet its primary endpoint, as the 24-week clinical benefit rate was not significantly improved by adding oleclumab (43% vs. 44%, p = 0.61). Exploratory median progression-free survival was 5.9 months in arm A as compared to 7.0 months in arm B (p = 0.90). The safety profile was manageable in both arms.
Collapse
Affiliation(s)
- Laurence Buisseret
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium.
| | - Delphine Loirat
- Medical Oncology Department, Institut Curie, 75005, Paris, France
| | - Philippe Aftimos
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Christian Maurer
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, 52074, Cologne, Germany
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Unit, Leuven Cancer Institute, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Véronique Debien
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Paulus Kristanto
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Daniel Eiger
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Anthony Goncalves
- Medical Oncology Department, Institut Paoli-Calmettes, 13274, Marseille, France
| | | | - Donatienne Taylor
- Department of Oncology, CHU-UCL-Namur - Site Sainte-Elisabeth, 5000, Namur, Belgium
| | - Florent Clatot
- Medical Oncology Department, Centre Henri Becquerel, 76038, Rouen, France
| | - Tom Van den Mooter
- Department of Oncology, GZA Ziekenhuizen Campus Sint-Augustinus, 2610, Antwerp, Belgium
| | - Jean-Marc Ferrero
- Department of Oncology, Centre Antoine Lacassagne, 06189, Nice, France
| | - Hervé Bonnefoi
- Medical Oncology Department, Institut Bergonié, 33000, Bordeaux, France
| | - Jean-Luc Canon
- Department of Oncology-Hematology, Grand Hôpital de Charleroi - Site Notre Dame, 6000, Charleroi, Belgium
| | - Francois P Duhoux
- Medical Oncology Department, Cliniques Universitaires Saint-Luc (UCLouvain), 1200, Brussels, Belgium
| | - Laura Mansi
- Department of Oncology, CHU Besançon - Hôpital Jean Minjoz, 25030, Besancon, France
| | - Renaud Poncin
- Medical Oncology Department, Clinique Saint-Pierre, 1340, Ottignies-Louvain-la-Neuve, Belgium
| | - Philippe Barthélémy
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe (ICANS), 67000, Strasbourg, France
| | - Nicolas Isambert
- Medical Oncology Department, CHU Poitiers, 86000, Poitiers, France
| | - Zoë Denis
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Xavier Catteau
- CurePath Laboratory (CHU Tivoli, CHIREC), 6040, Jumet, Belgium
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, 2610, Antwerp, Belgium
| | - Elisa Agostinetto
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Evandro de Azambuja
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Françoise Rothé
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Ligia Craciun
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - David Venet
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Emanuela Romano
- Centre for Cancer Immunotherapy, Medical Oncology Department, INSERM U932, Institut Curie, PSL Research University, 75005, Paris, France
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Pharmacie et Institut du Cancer de Montréal, Montréal, QC, 11290, Canada
| | - Marianne Paesmans
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Denis Larsimont
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Christos Sotiriou
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Michail Ignatiadis
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| | - Martine Piccart-Gebhart
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, 1070, Brussels, Belgium
| |
Collapse
|
17
|
Chen H, Wang S, Zhang Y, Gao X, Guan Y, Wu N, Wang X, Zhou T, Zhang Y, Cui D, Wang M, Zhang D, Wang J. A prognostic mathematical model based on tumor microenvironment-related genes expression for breast cancer patients. Front Oncol 2023; 13:1209707. [PMID: 37860187 PMCID: PMC10583559 DOI: 10.3389/fonc.2023.1209707] [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: 04/21/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Background Tumor microenvironment (TME) status is closely related to breast cancer (BC) prognosis and systemic therapeutic effects. However, to date studies have not considered the interactions of immune and stromal cells at the gene expression level in BC as a whole. Herein, we constructed a predictive model, for adjuvant decision-making, by mining TME molecular expression information related to BC patient prognosis and drug treatment sensitivity. Methods Clinical information and gene expression profiles were extracted from The Cancer Genome Atlas (TCGA), with patients divided into high- and low-score groups according to immune/stromal scores. TME-related prognostic genes were identified using Kaplan-Meier analysis, functional enrichment analysis, and protein-protein interaction (PPI) networks, and validated in the Gene Expression Omnibus (GEO) database. Least absolute shrinkage and selection operator (LASSO) Cox regression analysis was used to construct and verify a prognostic model based on TME-related genes. In addition, the patients' response to chemotherapy and immunotherapy was assessed by survival outcome and immunohistochemistry (IPS). Immunohistochemistry (IHC) staining laid a solid foundation for exploring the value of novel therapeutic target genes. Results By dividing patients into low- and high-risk groups, a significant distinction in overall survival was found (p < 0.05). The risk model was independent of multiple clinicopathological parameters and accurately predicted prognosis in BC patients (p < 0.05). The nomogram-integrated risk score had high prediction accuracy and applicability, when compared with simple clinicopathological features. As predicted by the risk model, regardless of the chemotherapy regimen, the survival advantage of the low-risk group was evident in those patients receiving chemotherapy (p < 0.05). However, in patients receiving anthracycline (A) therapy, outcomes were not significantly different when compared with those receiving no-A therapy (p = 0.24), suggesting these patients may omit from A-containing adjuvant chemotherapy. Our risk model also effectively predicted tumor mutation burden (TMB) and immunotherapy efficacy in BC patients (p < 0.05). Conclusion The prognostic score model based on TME-related genes effectively predicted prognosis and chemotherapy effects in BC patients. The model provides a theoretical basis for novel driver-gene discover in BC and guides the decision-making for the adjuvant treatment of early breast cancer (eBC).
Collapse
Affiliation(s)
- Hong Chen
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shan Wang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuting Zhang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xue Gao
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yufu Guan
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Nan Wu
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xinyi Wang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianyang Zhou
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Zhang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Di Cui
- Information Center, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mijia Wang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dianlong Zhang
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jia Wang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
18
|
Abdel-Rahman SA, Świderek K, Gabr MT. First-in-class small molecule inhibitors of ICOS/ICOSL interaction as a novel class of immunomodulators. RSC Med Chem 2023; 14:1767-1777. [PMID: 37731692 PMCID: PMC10507805 DOI: 10.1039/d3md00150d] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/28/2023] [Indexed: 09/22/2023] Open
Abstract
The interaction of the inducible co-stimulator (ICOS) with its ligand (ICOSL) plays key roles in T-cell differentiation and activation of T-cell to B-cell functions. The ICOS/ICOSL pathway is a validated target for T-cell lymphomas induced by the proliferation of T-follicular helper (Tfh) cells. Moreover, the inhibition of ICOS/ICOSL interaction can decrease the enhancement of immunosuppressive regulatory T cells (Tregs) in both hematologic malignancies and solid tumors. However, targeting ICOS/ICOSL interaction is currently restricted to monoclonal antibodies (mAbs) and there are no small molecules in existence that can block ICOS/ICOSL. To fill this gap, we report herein the first time-resolved fluorescence resonance energy transfer (TR-FRET) assay to evaluate the ability of small molecules to inhibit ICOS/ICOSL interaction. Implementation of the developed TR-FRET assay in high-throughput screening (HTS) of a focused chemical library resulted in the identification of AG-120 as a first-in-class inhibitor of ICOS/ICOSL interaction. We further employed docking studies and molecular dynamics (MD) simulations to identify the plausible mechanism of blocking ICOS/ICOSL complex formation by AG-120. Using the structure-activity relationship (SAR) by catalog approach, we identified AG-120-X with an IC50 value of 4.68 ± 0.47 μM in the ICOS/ICOSL TR-FRET assay. Remarkably, AG-120-X revealed a dose-dependent ability to block ICOS/ICOSL interaction in a bioluminescent cellular assay based on co-culturing Jurkat T cells expressing ICOS and CHO-K1 cells expressing ICOSL. This work will pave the way for future drug discovery efforts aiming at the development of small molecule inhibitors of ICOS/ICOSL interaction as potential therapeutics for cancer as well as other diseases.
Collapse
Affiliation(s)
- Somaya A Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine New York NY 10065 USA
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Katarzyna Świderek
- BioComp Group, Institute of Advanced Materials (INAM), Universitat Jaume I 12071 Castellon Spain
| | - Moustafa T Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine New York NY 10065 USA
| |
Collapse
|
19
|
Qian D, Xu Y, Wu Y, Qiu J, Hong W, Meng X. Assessment of the safety and efficacy of combination chemotherapy and PD-1/PD-L1 inhibitor treatment of breast cancer: A meta-analysis. Chin Med J (Engl) 2023; 136:1663-1670. [PMID: 37279392 PMCID: PMC10344516 DOI: 10.1097/cm9.0000000000002631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND As the efficacy of programmed cell death-1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors combined with chemotherapy in curing breast cancer is still controversial, this meta-analysis compares the efficacy and safety of PD-1/PD-L1 inhibitors combined with chemotherapy and chemotherapy alone in the treatment of breast cancer, which provides guidance for the clinical treatment. METHODS Relevant studies published as of April 2022 in the various databases including EMBASE, PubMed, and Cochrane Library were selected. Randomized controlled trials (RCTs) in which control patients underwent chemotherapy alone and experimental group patients underwent combination chemotherapy and PD-1/PD-L1 inhibitor treatment were included in this investigation. Investigations without complete information, researches from which information could not be extracted, duplicate articles, animal studies, review articles, and systematic reviews were excluded. STATA 15.1 was employed for all statistical analyses. RESULTS In total, eight eligible studies were identified, revealing that combination chemotherapy and PD-1/PD-L1 inhibitor treatment was linked to significant increases in progression-free survival (PFS) relative to chemotherapy alone (hazard ratio [HR] = 0.83, 95% confidence interval [CI]: 0.70-0.99, P = 0.032) but not overall survival (HR = 0.92, 95% CI: 0.80-1.06, P = 0.273). Pooled adverse event rates were also increased within the group of combination treatment relative to the chemotherapy group (risk ratio [RR] = 1.08, 95% CI: 1.03-1.14, P = 0.002). Specifically, nausea rates were lesser within the group of combination treatment relative to the group of chemotherapy (RR = 0.48, 95% CI: 0.25-0.92, P = 0.026). Subgroup analyses indicated that the PFS of patients who underwent combination atezolizumab or pembrolizumab and chemotherapy treatment were substantially longer than those of patients who underwent chemotherapy alone (HR = 0.79, 95% CI: 0.69-0.89, P ≤0.001; HR = 0.79, 95% CI: 0.67-0.92, P = 0.002). CONCLUSIONS The pooled results suggest that combination chemotherapy and PD-1/PD-L1 inhibitor treatment approaches help prolong PFS in breast cancer patients, but have no statistically significant effect on overall survival (OS). Additionally, combination therapy can significantly improve complete response rate (CRR) compared with chemotherapy alone. However, combination therapy was associated with greater rates of adverse events.
Collapse
Affiliation(s)
- Da Qian
- Department of Burn and Plastic Surgery-Hand Surgery, The Changshu Hospital Affiliated to Soochow University, Changshu, Soochow, Jiangsu 215500, China
| | - Yuhao Xu
- Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yihao Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310000, China
| | - Jie Qiu
- Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Weimin Hong
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310000, China
| | - Xuli Meng
- Department of Breast Surgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
20
|
Ensenyat-Mendez M, Orozco JIJ, Llinàs-Arias P, Íñiguez-Muñoz S, Baker JL, Salomon MP, Martí M, DiNome ML, Cortés J, Marzese DM. Construction and validation of a gene expression classifier to predict immunotherapy response in primary triple-negative breast cancer. COMMUNICATIONS MEDICINE 2023; 3:93. [PMID: 37430006 DOI: 10.1038/s43856-023-00311-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/31/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) improve clinical outcomes in triple-negative breast cancer (TNBC) patients. However, a subset of patients does not respond to treatment. Biomarkers that show ICI predictive potential in other solid tumors, such as levels of PD-L1 and the tumor mutational burden, among others, show a modest predictive performance in patients with TNBC. METHODS We built machine learning models based on pre-ICI treatment gene expression profiles to construct gene expression classifiers to identify primary TNBC ICI-responder patients. This study involved 188 ICI-naïve and 721 specimens treated with ICI plus chemotherapy, including TNBC tumors, HR+/HER2- breast tumors, and other solid non-breast tumors. RESULTS The 37-gene TNBC ICI predictive (TNBC-ICI) classifier performs well in predicting pathological complete response (pCR) to ICI plus chemotherapy on an independent TNBC validation cohort (AUC = 0.86). The TNBC-ICI classifier shows better performance than other molecular signatures, including PD-1 (PDCD1) and PD-L1 (CD274) gene expression (AUC = 0.67). Integrating TNBC-ICI with molecular signatures does not improve the efficiency of the classifier (AUC = 0.75). TNBC-ICI displays a modest accuracy in predicting ICI response in two different cohorts of patients with HR + /HER2- breast cancer (AUC = 0.72 to pembrolizumab and AUC = 0.75 to durvalumab). Evaluation of six cohorts of patients with non-breast solid tumors treated with ICI plus chemotherapy shows overall poor performance (median AUC = 0.67). CONCLUSION TNBC-ICI predicts pCR to ICI plus chemotherapy in patients with primary TNBC. The study provides a guide to implementing the TNBC-ICI classifier in clinical studies. Further validations will consolidate a novel predictive panel to improve the treatment decision-making for patients with TNBC.
Collapse
Affiliation(s)
- Miquel Ensenyat-Mendez
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Javier I J Orozco
- Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Pere Llinàs-Arias
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Sandra Íñiguez-Muñoz
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jennifer L Baker
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew P Salomon
- Department of Medicine, University of Southern California (USC), Los Angeles, CA, USA
| | - Mercè Martí
- Immunology Unit, Department of Cell Biology, Physiology, and Immunology, Institut de Biotecnologia I Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
- Biosensing and Bioanalysis Group, Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Maggie L DiNome
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona, Spain
- Medical Scientia Innovation Research (MedSIR), Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - Diego M Marzese
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
| |
Collapse
|
21
|
Ren X, Song Y, Pang J, Chen L, Zhou L, Liang Z, Wu H. Prognostic value of various immune cells and Immunoscore in triple-negative breast cancer. Front Immunol 2023; 14:1137561. [PMID: 37090736 PMCID: PMC10117828 DOI: 10.3389/fimmu.2023.1137561] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
BackgroundThis study aimed to evaluate the expression status and prognostic role of various immunoregulatory cells and test in triple-negative breast cancer (TNBC).MethodsThe expression of five markers (CD3/CD4/CD8/CD19/CD163) of tumor immune cells was evaluated retrospectively in tumor sections from 68 consecutive cases of TNBC by immunohistochemistry. Computational image analysis was used to quantify the density and distribution of each immune marker within the tumor region, tumor invasive margin, and expression hotspots. Immunoscores were calculated using an automated approach. Other clinical characteristics were also analyzed.ResultsFor all patients, Kaplan–Meier survival analysis showed that high CD3+ signals in the tumor region (disease-free survival (DFS), P=0.0014; overall survival (OS), P=0.0031) and total region (DFS, P=0.0014; OS, P=0.0031) were significantly associated with better survival. High CD4+ levels in the tumor region and total regions were significantly associated with better survival (P<0.05). For Hotspot analysis, CD3+ was associated with significantly better survival for all Top1, Top2, and Top3 densities (DFS and OS, P<0.05). High CD4+ levels were significantly associated with better prognosis for Top1 and Top3 densities (DFS and OS, P<0.05). For stage IIB and IIIC patients, CD3+ in the tumor region and all Top hotspots was found to be significantly correlated with survival (DFS and OS, P<0.05). CD4+ cells were significantly associated with survival in the tumor region, total region, and Top3 density (DFS, P=0.0213; OS, P=0.0728). CD8+ cells were significantly associated with survival in the invasive margin, Top2 density, and Top3 density. Spatial parameter analysis showed that high colocalization of tumor cells and immune cells (CD3+, CD4+, or CD8+) was significantly associated with patient survival.ConclusionComputational image analysis is a reliable tool for evaluating the density and distribution of immune regulatory cells and for calculating the Immunoscore in TNBC. The Immunoscore retains its prognostic significance in TNBC later than IIB stage breast cancer. Future studies are required to confirm its potential to predict tumor responses to chemotherapy and immune therapy.
Collapse
Affiliation(s)
- Xinyu Ren
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Song
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Junyi Pang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Longyun Chen
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liangrui Zhou
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyong Liang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Zhiyong Liang, ; Huanwen Wu,
| | - Huanwen Wu
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Zhiyong Liang, ; Huanwen Wu,
| |
Collapse
|
22
|
Little A, Tangney M, Tunney MM, Buckley NE. Fusobacterium nucleatum: a novel immune modulator in breast cancer? Expert Rev Mol Med 2023; 25:e15. [PMID: 37009688 PMCID: PMC10407221 DOI: 10.1017/erm.2023.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 04/04/2023]
Abstract
Breast cancer was the most commonly diagnosed cancer worldwide in 2020. Greater understanding of the factors which promote tumour progression, metastatic development and therapeutic resistance is needed. In recent years, a distinct microbiome has been detected in the breast, a site previously thought to be sterile. Here, we review the clinical and molecular relevance of the oral anaerobic bacterium Fusobacterium nucleatum in breast cancer. F. nucleatum is enriched in breast tumour tissue compared with matched healthy tissue and has been shown to promote mammary tumour growth and metastatic progression in mouse models. Current literature suggests that F. nucleatum modulates immune escape and inflammation within the tissue microenvironment, two well-defined hallmarks of cancer. Furthermore, the microbiome, and F. nucleatum specifically, has been shown to affect patient response to therapy including immune checkpoint inhibitors. These findings highlight areas of future research needed to better understand the influence of F. nucleatum in the development and treatment of breast cancer.
Collapse
Affiliation(s)
- Alexa Little
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Mark Tangney
- Cancer Research, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Michael M. Tunney
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Niamh E. Buckley
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| |
Collapse
|
23
|
Rapoport BL, Steel HC, Benn CA, Nayler S, Smit T, Heyman L, Theron AJ, Hlatshwayo N, Kwofie LL, Meyer PW, Anderson R. Dysregulation of systemic soluble immune checkpoints in early breast cancer is attenuated following administration of neoadjuvant chemotherapy and is associated with recovery of CD27, CD28, CD40, CD80, ICOS and GITR and substantially increased levels of PD-L1, LAG-3 and TIM-3. Front Oncol 2023; 13:1097309. [PMID: 37064132 PMCID: PMC10098332 DOI: 10.3389/fonc.2023.1097309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/09/2023] [Indexed: 04/03/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) may alter the immune landscape of patients with early breast cancer (BC), potentially setting the scene for more effective implementation of checkpoint-targeted immunotherapy. This issue has been investigated in the current study in which alterations in the plasma concentrations of 16 soluble co-stimulatory and co-inhibitory, immune checkpoints were measured sequentially in a cohort of newly diagnosed, early BC patients (n=72), pre-treatment, post-NAC and post-surgery using a Multiplex® bead array platform. Relative to a group of healthy control subjects (n=45), the median pre-treatment levels of five co-stimulatory (CD27, CD40, GITRL, ICOS, GITR) and three co-inhibitory (TIM-3, CTLA-4, PD-L1) soluble checkpoints were significantly lower in the BC patients vs. controls (p<0.021-p<0.0001; and p<0.008-p<0.00001, respectively). Following NAC, the plasma levels of six soluble co-stimulatory checkpoints (CD28, CD40, ICOS, CD27, CD80, GITR), all involved in activation of CD8+ cytotoxic T cells, were significantly increased (p<0.04-p<0.00001), comparable with control values and remained at these levels post-surgery. Of the soluble co-inhibitory checkpoints, three (LAG-3, PD-L1, TIM-3) increased significantly post-NAC, reaching levels significantly greater than those of the control group. PD-1 remained unchanged, while BTLA and CTLA-4 decreased significantly (p<0.03 and p<0.00001, respectively). Normalization of soluble co-stimulatory immune checkpoints is seemingly indicative of reversal of systemic immune dysregulation following administration of NAC in early BC, while recovery of immune homeostasis may explain the increased levels of several negative checkpoint proteins, albeit with the exceptions of CTLA-4 and PD-1. Although a pathological complete response (pCR) was documented in 61% of patients (mostly triple-negative BC), surprisingly, none of the soluble immune checkpoints correlated with the pCR, either pre-treatment or post-NAC. Nevertheless, in the case of the co-stimulatory ICMs, these novel findings are indicative of the immune-restorative potential of NAC in early BC, while in the case of the co-inhibitory ICMs, elevated levels of soluble PD-L1, LAG-3 and TIM-3 post-NAC underscore the augmentative immunotherapeutic promise of targeting these molecules, either individually or in combination, as a strategy, which may contribute to the improved management of early BC.
Collapse
Affiliation(s)
- Bernardo L. Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Medical Oncology Centre of Rosebank, Johannesburg, South Africa
- Netcare Breast Care Centre, Johannesburg, South Africa
- *Correspondence: Bernardo L. Rapoport,
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Carol A. Benn
- Netcare Breast Care Centre, Johannesburg, South Africa
- Department of Surgery, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Simon Nayler
- Netcare Breast Care Centre, Johannesburg, South Africa
- Drs Gritzman & Thatcher Inc. Laboratories, University of the Witwatersrand Donald Gordon Medical Centre, Johannesburg, South Africa
| | - Teresa Smit
- Medical Oncology Centre of Rosebank, Johannesburg, South Africa
| | - Liezl Heyman
- Medical Oncology Centre of Rosebank, Johannesburg, South Africa
| | - Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Nomsa Hlatshwayo
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa
| | - Luyanda L.I. Kwofie
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa
| | - Pieter W.A. Meyer
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
24
|
Su W, Qiu W, Li SJ, Wang S, Xie J, Yang QC, Xu J, Zhang J, Xu Z, Sun ZJ. A Dual-Responsive STAT3 Inhibitor Nanoprodrug Combined with Oncolytic Virus Elicits Synergistic Antitumor Immune Responses by Igniting Pyroptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209379. [PMID: 36545949 DOI: 10.1002/adma.202209379] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Immune checkpoint blockade (ICB) therapy shows excellent efficacy against malignancies; however, insufficient tumor immunogenicity and the immunosuppressive tumor microenvironment (TME) are considered as the two major stumbling blocks to a broad ICB response. Here, a combinational therapeutic strategy is reported, wherein TME-reactive oxygen species/pH dual-responsive signal transducers and activators of transcription 3 inhibitor nanoprodrugs MPNPs are combined with oncolytic herpes simplex virus 1 virotherapy to synergistically ignite pyroptosis for enhancing immunotherapy. MPNPs exhibit a certain level of tumor accumulation, reduce tumor cell stemness, and enhance antitumor immune responses. Furthermore, the simultaneous application of oncolytic viruses (OVs) confers MPNPs with higher tumor penetration capacity and remarkable gasdermin-E-mediated pyroptosis, thereby reshaping the TME and transforming "cold" tumors into "hot" ones. This "fire of immunity" strategy successfully activates robust T-cell-dependent antitumor responses, potentiating ICB effects against local recurrence and pulmonary metastasis in preclinical "cold" murine triple-negative breast cancer and syngeneic oral cancer models. Collectively, this work may pave a new way and offer an unprecedented opportunity for the combination of OVs with nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Wen Su
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Wei Qiu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Shu-Jin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Shuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jun Xie
- State Key Laboratory of Virology, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, P. R. China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Qi-Chao Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jiming Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Junjie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
- State Key Laboratory of Virology, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, P. R. China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| |
Collapse
|
25
|
Immunotherapy in breast cancer: an overview of current strategies and perspectives. NPJ Breast Cancer 2023; 9:7. [PMID: 36781869 PMCID: PMC9925769 DOI: 10.1038/s41523-023-00508-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 01/21/2023] [Indexed: 02/15/2023] Open
Abstract
Recent progress in immunobiology has led the way to successful host immunity enhancement against breast cancer. In triple-negative breast cancer, the combination of cancer immunotherapy based on PD-1/PD-L1 immune checkpoint inhibitors with chemotherapy was effective both in advanced and early setting phase 3 clinical trials. These encouraging results lead to the first approvals of immune checkpoint inhibitors in triple-negative breast cancer and thus offer new therapeutic possibilities in aggressive tumors and hard-to-treat populations. Furthermore, several ongoing trials are investigating combining immunotherapies involving immune checkpoint inhibitors with conventional therapies and as well as with other immunotherapeutic strategies such as cancer vaccines, CAR-T cells, bispecific antibodies, and oncolytic viruses in all breast cancer subtypes. This review provides an overview of immunotherapies currently under clinical development and updated key results from clinical trials. Finally, we discuss the challenges to the successful implementation of immune treatment in managing breast cancer and their implications for the design of future clinical trials.
Collapse
|
26
|
[Triple-negative breast cancer : Classification, current concepts, and therapy-related factors]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:32-38. [PMID: 36595080 DOI: 10.1007/s00292-022-01177-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/04/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for about 10% of all breast cancer cases and is defined by the lack of expression of estrogen and progesterone receptors and the lack of overexpression or amplification of HER2. It differs with regard to the younger age of the patients, an increased association with a mutation of BRCA1 and a mostly low differentiation from hormone receptor-positive breast cancer. The spectrum of triple-negative breast cancer shows considerable heterogeneity both at the morphological and at the molecular level. It includes most commonly TNBC of no special type, with and without basal phenotype, triple-negative metaplastic breast carcinomas, triple-negative breast carcinomas with apocrine differentiation and rare triple-negative tumor types. At the gene-expression level, TNBC most commonly is associated with a basal phenotype, with rarer molecular variants of TNBC involving the Claudin-low, molecular apocrine types, and other rarer subtypes. Therefore, a critical use of the term TNBC, considering the histopathological tumor differentiation, is recommended.
Collapse
|
27
|
Hajibabaei S, Sotoodehnejadnematalahi F, Nafissi N, Zeinali S, Azizi M. Aberrant promoter hypermethylation of miR-335 and miR-145 is involved in breast cancer PD-L1 overexpression. Sci Rep 2023; 13:1003. [PMID: 36653507 PMCID: PMC9849328 DOI: 10.1038/s41598-023-27415-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
PD-L1 is one of the most important immune checkpoint molecules in breast cancer that plays an important role in suppressing the immune system when confronted with tumor cells and is regulated by various microRNAs. Among them, microRNA-335-3p and microRNA-145-5p, regulated by DNA methylation, have tumor suppressor activities. We studied the role of miR-335 and -145 on PD-L1 suppression in breast cancer. The expression of miR-355 and miR-145 was significantly downregulated in BC tissues and cell lines compared to their controls, and their downregulation was negatively correlated with PD-L1 overexpression. In-silico and luciferase reporter systems confirmed that miR-335 and -145 target PD-L1. In BC tissues and cell lines, cancer-specific methylation was found in CpG-rich areas upstream of miR-335 and-145, and up-regulation of PD-L1 expression was connected with hypermethylation (r = 0.4089, P = 0.0147, and r = 0.3373, P = 0.0475, respectively). The higher levels of miR-355 and -145 in BC cells induced apoptosis, arrested the cell cycle, and reduced proliferation significantly. In summary, we found that miR-335 and -145 are novel tumor suppressors inactivated in BC, and these miRs may serve as potential therapeutic targets for breast cancer treatment.
Collapse
Affiliation(s)
- Sara Hajibabaei
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Nahid Nafissi
- Breast Surgery Department, Iran University of Medical Sciences, Tehran, Iran
| | - Sirous Zeinali
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, 69th Pasteur Street, Kargar Avenue, Tehran, Iran
| | - Masoumeh Azizi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, 69th Pasteur Street, Kargar Avenue, Tehran, Iran.
| |
Collapse
|
28
|
Shojaei S, Aznab M, Rahimi A, Ahmadi SM, Eslampia K, Golpazir A, Iravani M. Evaluation of the Role of Tumor-Infiltrating Lymphocytes and CD8 + Cytotoxic Lymphocytes in the Survival of Patients with Breast Cancer. Int J Hematol Oncol Stem Cell Res 2023; 17:9-17. [PMID: 37638284 PMCID: PMC10448926 DOI: 10.18502/ijhoscr.v17i1.11708] [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: 09/23/2020] [Accepted: 05/09/2021] [Indexed: 08/29/2023] Open
Abstract
Background: This study aimed to evaluate the significance of tumor-infiltrating lymphocyte (TIL) and the number of CD8+ T cells in breast cancer and their relationship with the other clinicopathological factors, and overall survival (OS) was investigated. Materials and Methods: The studied samples were breast cancer patients (2005-2017) referring to the medical oncology departments for treatment. Pathologic samples of breast cancer patients were evaluated in terms of TIL and positive immunohistochemical staining for CD8 cytotoxic cells. Results: 299 patients were entered into the study, three were male and 296 female. Their mean follow-up period was 61 months. Statistical findings indicated that lymph involvement is more accompanied by low TIL within the tumor (0.011). Correlations were observed between the estrogen, progesterone receptors, P53 state, and TIL, which were significant by P-value<0.049, P-value=0.024, P-value =0.002, respectively. With any Ki67 value, the number of patients with less than 30% TIL was more considerable than the other two groups with lymphocyte cut-off of 30-50% and more than 50%. Comparison of the OS of patients with positive and negative CD8 cytotoxic lymphocytes in 45 patients with lymphocyte infiltration of equal or more than 40% showed that the OS results were in favor of patients with CD8+ cytotoxic lymphocyte (0.022). Out of 299 patients, 17 died. Conclusion: Our findings showed that in cases of CD8+ cytotoxic lymphocytes in tumors, the OS of the patients will be enhanced which can act as an independent factor.
Collapse
Affiliation(s)
| | - Mozaffar Aznab
- Department of Internal Medicine, Talaghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Seyed Mojtaba Ahmadi
- Department of Clinical Psychology School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | | | - Arash Golpazir
- Department of Surgery, Cancer Surgery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
29
|
Wang D. Targeting the stage-specific embryonic antigen (SSEA)-0 tumor neoantigen. CURRENT TRENDS IN IMMUNOLOGY 2023; 24:1-7. [PMID: 38699667 PMCID: PMC11064955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Recognition of abnormal glycosylation in virtually any cancer type has raised a great interest in the glycan-based tumor biomarkers. Our team explored carbohydrate microarrays as a broad-spectrum immunoassay to probe the immunologically potent tumor glycan targets. This effort has led to the identification of a blood group precursor antigen SSEA-0 as a conserved breast cancer (BCA) marker. Since this immunogenic O-core glycan is normally hidden as a cryptic antigen but becomes overexpressed and surface-exposed by metastatic breast cancer cells (MBCA), its potential as a novel immunological target for precision immunotherapy against tumor metastasis warrants a focused investigation.
Collapse
Affiliation(s)
- Denong Wang
- Tumor Glycomics Laboratory, SRI International Biosciences Division, 333 Ravenswood Avenue, Menlo Park, CA 94025-3493, USA
| |
Collapse
|
30
|
Somasundaram V, Ridnour LA, Cheng RY, Walke AJ, Kedei N, Bhattacharyya DD, Wink AL, Edmondson EF, Butcher D, Warner AC, Dorsey TH, Scheiblin DA, Heinz W, Bryant RJ, Kinders RJ, Lipkowitz S, Wong ST, Pore M, Hewitt SM, McVicar DW, Anderson SK, Chang J, Glynn SA, Ambs S, Lockett SJ, Wink DA. Systemic Nos2 Depletion and Cox inhibition limits TNBC disease progression and alters lymphoid cell spatial orientation and density. Redox Biol 2022; 58:102529. [PMID: 36375380 PMCID: PMC9661390 DOI: 10.1016/j.redox.2022.102529] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/10/2022] Open
Abstract
Antitumor immune polarization is a key predictor of clinical outcomes to cancer therapy. An emerging concept influencing clinical outcome involves the spatial location of CD8+ T cells, within the tumor. Our earlier work demonstrated immunosuppressive effects of NOS2 and COX2 tumor expression. Here, we show that NOS2/COX2 levels influence both the polarization and spatial location of lymphoid cells including CD8+ T cells. Importantly, elevated tumor NOS2/COX2 correlated with exclusion of CD8+ T cells from the tumor epithelium. In contrast, tumors expressing low NOS2/COX2 had increased CD8+ T cell penetration into the tumor epithelium. Consistent with a causative relationship between these observations, pharmacological inhibition of COX2 with indomethacin dramatically reduced tumor growth of the 4T1 model of TNBC in both WT and Nos2- mice. This regimen led to complete tumor regression in ∼20-25% of tumor-bearing Nos2- mice, and these animals were resistant to tumor rechallenge. Th1 cytokines were elevated in the blood of treated mice and intratumoral CD4+ and CD8+ T cells were higher in mice that received indomethacin when compared to control untreated mice. Multiplex immunofluorescence imaging confirmed our phenotyping results and demonstrated that targeted Nos2/Cox2 blockade improved CD8+ T cell penetration into the 4T1 tumor core. These findings are consistent with our observations in low NOS2/COX2 expressing breast tumors proving that COX2 activity is responsible for limiting the spatial distribution of effector T cells in TNBC. Together these results suggest that clinically available NSAID's may provide a cost-effective, novel immunotherapeutic approach for treatment of aggressive tumors including triple negative breast cancer.
Collapse
Affiliation(s)
- Veena Somasundaram
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Lisa A Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Robert Ys Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Abigail J Walke
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Noemi Kedei
- Collaborative Protein Technology Resource Nanoscale Protein Analysis, Office of Science Technology Resources, CCR, NCI, NIH, Bethesda, MD, USA
| | - Dibyangana D Bhattacharyya
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Andrew C Warner
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD, USA
| | - David A Scheiblin
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - William Heinz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Richard J Bryant
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Robert J Kinders
- Office of the Director, Division of Cancer Treatment and Diagnosis, NCI, Frederick, MD, USA
| | | | - Stephen Tc Wong
- Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center and Weill Cornell Medical College, Houston, TX, USA
| | - Milind Pore
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research, USA
| | | | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Stephen K Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jenny Chang
- Mary and Ron Neal Cancer Center, Houston Methodist Weill Cornell Medical College, Houston, TX, USA
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, H91 TK33, Ireland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA.
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
31
|
Pan YR, Wu CE, Huang WK, Chen MH, Lan KH, Yeh CN. Chimeric immune checkpoint protein vaccines inhibit the tumorigenesis and growth of rat cholangiocarcinoma. Front Immunol 2022; 13:982196. [PMID: 36341387 PMCID: PMC9631822 DOI: 10.3389/fimmu.2022.982196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022] Open
Abstract
Cholangiocarcinoma (CCA) is the second most common primary liver malignancy and carries a dismal prognosis due to difficulties in achieving an optimal resection, and poor response to current standard-of-care systemic therapies. We previously devised a CTLA4-PD-L1 DNA cancer vaccine (DNA vaccine) and demonstrated its therapeutic effects on reducing tumor growth in a thioacetamide (TAA)-induced rat intrahepatic CCA (iCCA) model. Here, we developed a CTLA4-PD-L1 chimeric protein vaccine (Protein vaccine), and examined its effects in the rat iCCA model. In a therapeutic setting, iCCA-bearing rats received either DNA plus Protein vaccines or Protein vaccine alone, resulting in increased PD-L1 and CTLA-4 antibody titers, and reduced iCCA tumor burden as verified by animal positron emission tomography (PET) scans. Treating iCCA-bearing rats with Protein vaccine alone led to the increase of CTAL4 antibody titers that correlated with the decrease of tumor SUV ratio, indicating regressed tumor burden, along with increased CD8 and granzyme A (GZMA) expression, and decreased PD-L1 expression on tumor cells. In a preventive setting, DNA or Protein vaccines were injected in rats before the induction of iCCA by TAA. Protein vaccines induced a more sustained PD-L1 and CTLA-4 antibody titers compared with DNA vaccines, and was more potent in preventing iCCA tumorigenesis. Correspondingly, Protein vaccines, but not DNA vaccines, downregulated PD-L1 gene expression and hindered the carcinogenesis of iCCA. Taken together, the CTLA4-PD-L1 chimeric protein vaccine may function both as a therapeutic cancer vaccine and as a preventive cancer vaccine in the TAA-induced iCCA rat model.
Collapse
Affiliation(s)
- Yi-Ru Pan
- Department of Surgery and Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Chiao-En Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ming-Huang Chen
- Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Keng-Hsueh Lan
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- *Correspondence: Keng-Hsueh Lan, ; Chun-Nan Yeh,
| | - Chun-Nan Yeh
- Department of Surgery and Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
- *Correspondence: Keng-Hsueh Lan, ; Chun-Nan Yeh,
| |
Collapse
|
32
|
Wang T, Gao Z, Zhang Y, Hong Y, Tang Y, Shan K, Kong X, Wang Z, Shi Y, Ding D. A supramolecular self-assembled nanomaterial for synergistic therapy of immunosuppressive tumor. J Control Release 2022; 351:272-283. [PMID: 36116581 DOI: 10.1016/j.jconrel.2022.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/30/2022] [Accepted: 09/11/2022] [Indexed: 11/25/2022]
Abstract
Triple negative breast cancer (TNBC) is an immunosuppressive "cold" tumor that lacks immune cell infiltration and activation, resulting in a poor response to immune checkpoint blockade (ICB) therapies. In addition, TNBC is poorly responsive to targeted therapies due to the absence of efficient molecular targets. A strategy that can block molecular signal transduction, stimulate immunogenicity, and activate the immune response is a promising approach to achieve ideal clinical benefit. Herein, we designed and synthesized an aggregation-induced emission luminogen (AIEgen)-conjugated self-assembling peptide that targets epidermal growth factor receptor (EGFR), named TPA-FFG-LA. TPA-FFG-LA peptides form nanoassemblies on the surface of EGFR-positive TNBC cells and are internalized into cells through endocytosis, which inhibit EGFR signaling transduction and provoke lysosomal membrane permeabilization (LMP). Upon light irradiation, the aggregated AIEgens produce massive reactive oxygen species (ROS) to exacerbate LMP and trigger immunogenic cell death (ICD), resulting in elimination of residual EGFR-negative tumor cells and exerting long-term antitumor effects. The in vitro and in vivo experiments verified that TPA-FFG-LA nanoassemblies suppress tumor growth, provoke immune cell activation and infiltration, and cause EGFR degradation and LMP. These results suggest that the combination of supramolecular assembly induced molecular targeting effects and lysosome dysfunction with ICD-stimulated immune activation is a plausible strategy for the efficient therapy of immunosuppressive TNBC.
Collapse
Affiliation(s)
- Tianjiao Wang
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhiyuan Gao
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yufan Zhang
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuning Hong
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Youhong Tang
- Australia-China Joint Centre for Personal Health Technologies, Medical Device Research Institute, Flinders University, South Australia 5042, Australia
| | - Ke Shan
- Shandong Artificial intelligence Institute and Shandong Computer Science Center, Qilu University of Technology, Jinan 250353, China
| | - Xianglong Kong
- Shandong Artificial intelligence Institute and Shandong Computer Science Center, Qilu University of Technology, Jinan 250353, China
| | - Zhiming Wang
- AIE Institute, Center for Aggregation-Induced Emission, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Yang Shi
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Dan Ding
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
33
|
Bojko M, Węgrzyn K, Sikorska E, Kocikowski M, Parys M, Battin C, Steinberger P, Kogut MM, Winnicki M, Sieradzan AK, Spodzieja M, Rodziewicz-Motowidło S. Design, synthesis and biological evaluation of PD-1 derived peptides as inhibitors of PD-1/PD-L1 complex formation for cancer therapy. Bioorg Chem 2022; 128:106047. [DOI: 10.1016/j.bioorg.2022.106047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/04/2022] [Accepted: 07/19/2022] [Indexed: 12/11/2022]
|
34
|
TNFRSF9 Suppressed the Progression of Breast Cancer via the p38MAPK/PAX6 Signaling Pathway. JOURNAL OF ONCOLOGY 2022; 2022:8549781. [PMID: 35799609 PMCID: PMC9256432 DOI: 10.1155/2022/8549781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022]
Abstract
Worldwide, breast cancer is the most common cancer in females. Endocrine therapy can effectively treat 85% of breast cancer patients, but 15% of patients could only be treated with chemotherapy and surgery, and the prognosis is much worse. Immunotherapy is the novel treatment for breast cancer, where PD-1 and CTLA-4 antibodies have shown evidence of immune modulation in breast cancer drug trials. In this study, we report that TNFRSF9 regulates the cell proliferation, invasion, and apoptosis of breast cancer cells through regulating the phosphorylation of p38, thus further regulating the expression of PAX6. In both breast cancer tissues and cell lines, the levels of TNFRSF9 are significantly decreased, and breast cancer cell development will be promoted with knockdown of TNFRSF9. Moreover, we identify that downregulation of TNFRSF9 can upregulate the phosphorylated p38 (p-p38) and PAX6. We further elucidate that p-p38 is essential for PAX6 expression as p38 phosphorylation inhibitor can reverse the upregulation of PAX6 and suppress cell proliferation and invasion and promote apoptosis in breast cancer cells. In summary, this study proposed a novel TNFRSF9/p38/PAX6 axis that contributes to tumor suppression, which suggests a potential immunotherapy target for breast cancer.
Collapse
|
35
|
Čeprnja T, Mrklić I, Perić Balja M, Marušić Z, Blažićević V, Spagnoli GC, Juretić A, Čapkun V, Tečić Vuger A, Vrdoljak E, Tomić S. Prognostic Significance of Lymphocyte Infiltrate Localization in Triple-Negative Breast Cancer. J Pers Med 2022; 12:941. [PMID: 35743725 PMCID: PMC9224650 DOI: 10.3390/jpm12060941] [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: 05/10/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
High infiltration by tumor-infiltrating lymphocytes (TILs) is associated with favorable prognosis in different tumor types, but the clinical significance of their spatial localization within the tumor microenvironment is debated. To address this issue, we evaluated the accumulation of intratumoral TILs (itTILs) and stromal TILs (sTILs) in samples from 97 patients with early triple-negative breast cancer (TNBC) in the center (sTIL central) and periphery (sTIL peripheral) of tumor tissues. Moreover, the presence of primary and secondary lymphoid aggregates (LAs) and the expression levels of the cancer testis antigen (CTA), NY-ESO-1, and PD-L1 were explored. High infiltration by itTILs was observed in 12/97 samples (12.3%), unrelated to age, Ki67 expression, tumor size, histologic type and grade, and LA presence. NY-ESO-1 was expressed in tumor cells in 37 samples (38%), with a trend suggesting a correlation with itTIL infiltration (p = 0.0531). PD-L1 expression was detected in immune cells in 47 samples (49%) and was correlated with histologic grade, sTILs, and LA formation. The presence of primary LAs was significantly correlated with better disease-free survival (DFS) (p = 0.027). Moreover, no tumor progression was observed during >40 months of clinical follow up in the 12 patients with high itTILs or in the 14 patients with secondary LAs. Thus, careful evaluation of lymphoid infiltrate intratumoral localization might provide important prognostic information.
Collapse
Affiliation(s)
- Toni Čeprnja
- Department of Pathology, Forensic Medicine and Cytology, University Hospital Center Split, School of Medicine, University of Split, 21000 Split, Croatia; (I.M.); (S.T.)
| | - Ivana Mrklić
- Department of Pathology, Forensic Medicine and Cytology, University Hospital Center Split, School of Medicine, University of Split, 21000 Split, Croatia; (I.M.); (S.T.)
| | - Melita Perić Balja
- Department of Pathology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Zlatko Marušić
- Department of Pathology, Zagreb University Hospital Center, 10000 Zagreb, Croatia;
| | | | | | - Antonio Juretić
- Department of Oncology, Clinical Hospital “Sveti Duh”, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Vesna Čapkun
- Department of Nuclear Medicine, University Hospital Centre Split, School of Medicine, University of Split, 21000 Split, Croatia;
| | - Ana Tečić Vuger
- Department of Oncology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Eduard Vrdoljak
- Department of Oncology, University Hospital Center Split, University of Split, 21000 Split, Croatia;
| | - Snježana Tomić
- Department of Pathology, Forensic Medicine and Cytology, University Hospital Center Split, School of Medicine, University of Split, 21000 Split, Croatia; (I.M.); (S.T.)
| |
Collapse
|
36
|
Mao C, Yeh S, Fu J, Porosnicu M, Thomas A, Kucera GL, Votanopoulos KI, Tian S, Ming X. Delivery of an ectonucleotidase inhibitor with ROS-responsive nanoparticles overcomes adenosine-mediated cancer immunosuppression. Sci Transl Med 2022; 14:eabh1261. [PMID: 35675434 DOI: 10.1126/scitranslmed.abh1261] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Tumor evasion of immune destruction is associated with the production of immunosuppressive adenosine in the tumor microenvironment (TME). Anticancer therapies can trigger adenosine triphosphate (ATP) release from tumor cells, causing rapid formation of adenosine by the ectonucleotidases CD39 and CD73, thereafter exacerbating immunosuppression in the TME. The goal of this study was to develop an approach to facilitate cancer therapy-induced immunogenic cell death including ATP release and to limit ATP degradation into adenosine, in order to achieve durable antitumor immune response. Our approach was to construct reactive oxygen species (ROS)-producing nanoparticles that carry an ectonucleotidase inhibitor ARL67156 by electronic interaction and phenylboronic ester. Upon near-infrared irradiation, nanoparticle-produced ROS induced ATP release from MOC1 cancer cells in vitro and triggered the cleavage of phenylboronic ester, facilitating the release of ARL67156 from the nanoparticles. ARL67156 prevented conversion of ATP to adenosine and enhanced anticancer immunity in an MOC1-based coculture model. We tested this approach in mouse tumor models. Nanoparticle-based ROS-responsive drug delivery reprogramed the immunogenic landscape in tumors, eliciting tumor-specific T cell responses and tumor regression, conferring long-term survival in mouse models. We demonstrated that TME reprograming sets the stage for response to anti-programmed cell death protein 1 (PD1) immunotherapy, and the combination resulted in tumor regression in a 4T1 breast cancer mouse model that was resistant to PD1 blockade. Furthermore, our approach also induced immunological effects in patient-derived organotypic tumor spheroid model, suggesting potential translation of our nanoparticle approach for treating human cancers.
Collapse
Affiliation(s)
- Chengqiong Mao
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Stacy Yeh
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Juan Fu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Mercedes Porosnicu
- Depatment of Internal Medicine - Section of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Alexandra Thomas
- Depatment of Internal Medicine - Section of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Gregory L Kucera
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Konstantinos I Votanopoulos
- Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA.,Department of Surgery - Section of Surgical Oncology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Shaomin Tian
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xin Ming
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| |
Collapse
|
37
|
Valencia GA, Rioja P, Morante Z, Ruiz R, Fuentes H, Castaneda CA, Vidaurre T, Neciosup S, Gomez HL. Immunotherapy in triple-negative breast cancer: A literature review and new advances. World J Clin Oncol 2022; 13:219-236. [PMID: 35433291 PMCID: PMC8966508 DOI: 10.5306/wjco.v13.i3.219] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/23/2021] [Accepted: 02/19/2022] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly complex, heterogeneous disease and historically has limited treatment options. It has a high probability of disease recurrence and rapid disease progression despite adequate systemic treatment. Immunotherapy has emerged as an important alternative in the management of this malignancy, showing an impact on progression-free survival and overall survival in selected populations. In this review we focused on immunotherapy and its current relevance in the management of TNBC, including various scenarios (metastatic and early -neoadjuvant, adjuvant-), new advances in this subtype and the research of potential predictive biomarkers of response to treatment.
Collapse
Affiliation(s)
| | - Patricia Rioja
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Zaida Morante
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Rossana Ruiz
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Hugo Fuentes
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Carlos A Castaneda
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Tatiana Vidaurre
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Silvia Neciosup
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| | - Henry L Gomez
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15036, Peru
| |
Collapse
|
38
|
Zhu L, Chen M, Huang B, Zhang T, Chen K, Lian H, Liu M, Zhao K, Pang Y, Zhang J, Li Q, Zhong C. Genomic Analysis Uncovers Immune Microenvironment Characteristics and Drug Sensitivity of Ferroptosis in Breast Cancer Brain Metastasis. Front Genet 2022; 12:819632. [PMID: 35154262 PMCID: PMC8830939 DOI: 10.3389/fgene.2021.819632] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/24/2021] [Indexed: 01/31/2023] Open
Abstract
Background: The role of ferroptosis in breast cancer brain metastasis (BCBM) is unclear. This study aimed to explore the ferroptosis-related genes (FRG) relations with the tumor microenvironment, as well as evaluate their values in predicting survival and drug sensitivity in patients with BCBM. Materials and Methods: Genes expression and clinical data were downloaded from Gene Expression Omnibus (GEO). Univariate and multivariate Cox regression analyses were performed to explore the independent prognostic factors. Consensus cluster principal component analysis (PCA) was used to establish the ferroptosis score. Immunological signatures were analyzed by the single-sample gene set enrichment analysis (ssGSEA). Drug sensitivity was evaluated through the estimated half-maximal inhibitory concentration (IC50). Finally, results were validated in external cohorts. Results: Fourteen significantly different FRG were identified between breast cancer (BC) and BCBM tissues. Survival analysis demonstrated HMOX1, PEBP1, KEAP1, and LPCAT3 were significantly associated with overall survival (OS) and relapse-free survival (RFS) (all p < 0.05). High ferroptosis score was correlated with iron ion homeostasis, iron metabolism, higher stromal cells and immune cells scores. Patients with high- and low-ferroptosis scores were characterized by different drug sensitivities. Following external validations, the ferroptosis had distinct expression profiles between the BC and BCBM, and could serve as biomarkers for OS and drug response. Conclusion: Our findings suggested that ferroptosis may be involved in the process of BCBM, and ferroptosis could serve as prognostic biomarkers. Evaluation of ferroptosis may deepen our understanding about the tumor microenvironment, and could help clinicians to make individualized therapy.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Thoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mu Chen
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingsong Huang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kui Chen
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Lian
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Min Liu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Pang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Ying Pang, ; Jing Zhang, ; Qinchuan Li, ; Chunlong Zhong,
| | - Jing Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Advanced Study, Tongji University, Shanghai, China
- *Correspondence: Ying Pang, ; Jing Zhang, ; Qinchuan Li, ; Chunlong Zhong,
| | - Qinchuan Li
- Department of Thoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Ying Pang, ; Jing Zhang, ; Qinchuan Li, ; Chunlong Zhong,
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Ying Pang, ; Jing Zhang, ; Qinchuan Li, ; Chunlong Zhong,
| |
Collapse
|
39
|
Pharmacological disruption of the MTDH-SND1 complex enhances tumor antigen presentation and synergizes with anti-PD-1 therapy in metastatic breast cancer. NATURE CANCER 2022; 3:60-74. [PMID: 35121988 PMCID: PMC8818088 DOI: 10.1038/s43018-021-00280-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 09/23/2021] [Indexed: 01/08/2023]
Abstract
Despite increased overall survival rates, curative options for metastatic breast cancer remain limited. We have previously shown that metadherin (MTDH) is frequently overexpressed in poor prognosis breast cancer, where it promotes metastasis and therapy resistance through its interaction with staphylococcal nuclease domain-containing 1 (SND1). Through genetic and pharmacological targeting of the MTDH-SND1 interaction, we reveal a key role for this complex in suppressing antitumor T cell responses in breast cancer. The MTDH-SND1 complex reduces tumor antigen presentation and inhibits T cell infiltration and activation by binding to and destabilizing Tap1/2 messenger RNAs, which encode key components of the antigen-presentation machinery. Following small-molecule compound C26-A6 treatment to disrupt the MTDH-SND1 complex, we showed enhanced immune surveillance and sensitivity to anti-programmed cell death protein 1 therapy in preclinical models of metastatic breast cancer, in support of this combination therapy as a viable approach to increase immune-checkpoint blockade therapy responses in metastatic breast cancer.
Collapse
|
40
|
Crosstalk between non-coding RNAs expression profile, drug resistance and immune response in breast cancer. Pharmacol Res 2021; 176:106041. [PMID: 34952200 DOI: 10.1016/j.phrs.2021.106041] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/15/2021] [Accepted: 12/19/2021] [Indexed: 12/18/2022]
Abstract
Drug resistance is one of the most critical challenges facing researchers in treating breast cancer. Despite numerous treatments for breast cancer, including conventional chemical drugs, monoclonal antibodies, and immunotherapeutic drugs known as immune checkpoint inhibitors (ICI), many patients resist various approaches. In recent years, the relationship between gene expression profiles and drug resistance phenotypes has attracted much attention. Non-coding RNAs (ncRNAs) are regulatory molecules that have been shown to regulate gene expression and cell transcriptome. Two categories, microRNAs and long non-coding RNAs have been more considered and studied among these ncRNAs. Studying the role of different ncRNAs in chemical drug resistance and ICI resistance together can be beneficial in selecting more effective treatments for breast cancer. Changing the expression and action mechanism of these regulatory molecules on drug resistance phenotypes is the main topic of this review article.
Collapse
|
41
|
Ledys F, Kalfeist L, Galland L, Limagne E, Ladoire S. Therapeutic Associations Comprising Anti-PD-1/PD-L1 in Breast Cancer: Clinical Challenges and Perspectives. Cancers (Basel) 2021; 13:5999. [PMID: 34885109 PMCID: PMC8656936 DOI: 10.3390/cancers13235999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
Despite a few cases of long-responder patients, immunotherapy with anti-PD-(L)1 has so far proved rather disappointing in monotherapy in metastatic breast cancer, prompting the use of synergistic therapeutic combinations incorporating immunotherapy by immune-checkpoint inhibitors. In addition, a better understanding of both the mechanisms of sensitivity and resistance to immunotherapy, as well as the immunological effects of the usual treatments for breast cancer, make it possible to rationally consider this type of therapeutic combination. For several years, certain treatments, commonly used to treat patients with breast cancer, have shown that in addition to their direct cytotoxic effects, they may have an impact on the tumor immune microenvironment, by increasing the antigenicity and/or immunogenicity of a "cold" tumor, targeting the immunosuppressive microenvironment or counteracting the immune-exclusion profile. This review focuses on preclinical immunologic synergic mechanisms of various standard therapeutic approaches with anti-PD-(L)1, and discusses the potential clinical use of anti-PD-1/L1 combinations in metastatic or early breast cancer.
Collapse
Affiliation(s)
- Fanny Ledys
- Platform of Transfer in Cancer Biology, Georges-François Leclerc Center, 21000 Dijon, France; (F.L.); (L.K.); (L.G.); (E.L.)
- School of Medicine and Pharmacy, University of Burgundy Franche-Comté, 21000 Dijon, France
- UMR INSERM 1231, Lipides Nutrition Cancer, 21000 Dijon, France
| | - Laura Kalfeist
- Platform of Transfer in Cancer Biology, Georges-François Leclerc Center, 21000 Dijon, France; (F.L.); (L.K.); (L.G.); (E.L.)
- School of Medicine and Pharmacy, University of Burgundy Franche-Comté, 21000 Dijon, France
- UMR INSERM 1231, Lipides Nutrition Cancer, 21000 Dijon, France
| | - Loick Galland
- Platform of Transfer in Cancer Biology, Georges-François Leclerc Center, 21000 Dijon, France; (F.L.); (L.K.); (L.G.); (E.L.)
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
| | - Emeric Limagne
- Platform of Transfer in Cancer Biology, Georges-François Leclerc Center, 21000 Dijon, France; (F.L.); (L.K.); (L.G.); (E.L.)
- School of Medicine and Pharmacy, University of Burgundy Franche-Comté, 21000 Dijon, France
- UMR INSERM 1231, Lipides Nutrition Cancer, 21000 Dijon, France
| | - Sylvain Ladoire
- Platform of Transfer in Cancer Biology, Georges-François Leclerc Center, 21000 Dijon, France; (F.L.); (L.K.); (L.G.); (E.L.)
- School of Medicine and Pharmacy, University of Burgundy Franche-Comté, 21000 Dijon, France
- UMR INSERM 1231, Lipides Nutrition Cancer, 21000 Dijon, France
- Department of Medical Oncology, Georges-François Leclerc Center, 21000 Dijon, France
| |
Collapse
|
42
|
Spacek J, Vocka M, Zavadova E, Konopasek B, Petruzelka L. Immunomodulation with β-glucan from Pleurotus ostreatus in patients with endocrine-dependent breast cancer. Immunotherapy 2021; 14:31-40. [PMID: 34784798 DOI: 10.2217/imt-2021-0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To evaluate the effect of pleuran (β-glucan from Pleurotus ostreatus) administration on the immune profile of patients with endocrine-dependent breast cancer (clinical stages I-II) in clinical and imaging remission. Methodology: Antitumor cellular immunity (CD19+, CD3+, CD4+ and CD8+ T lymphocytes, and natural killer cells) of 195 patients (49 in the pleuran group and 146 in the control group) was measured by flow cytometry. Results: We observed a significant increase in the absolute number of CD3+, CD19+, CD4+ and CD8+ T lymphocytes in the pleuran group compared with the control group. Conclusion: Our results suggest potential benefit of continuous pleuran administration on immune rehabilitation of cellular antitumor immunity and better prognosis in breast cancer patients in remission.
Collapse
Affiliation(s)
- Jan Spacek
- Department of Oncology, First Faculty of Medicine, Charles University & General University Hospital in Prague, U Nemocnice 2, Praha 2, 128 08, Czech Republic
| | - Michal Vocka
- Department of Oncology, First Faculty of Medicine, Charles University & General University Hospital in Prague, U Nemocnice 2, Praha 2, 128 08, Czech Republic
| | - Eva Zavadova
- Department of Oncology, First Faculty of Medicine, Charles University & General University Hospital in Prague, U Nemocnice 2, Praha 2, 128 08, Czech Republic
| | - Bohuslav Konopasek
- Department of Oncology, First Faculty of Medicine, Charles University & General University Hospital in Prague, U Nemocnice 2, Praha 2, 128 08, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, First Faculty of Medicine, Charles University & General University Hospital in Prague, U Nemocnice 2, Praha 2, 128 08, Czech Republic
| |
Collapse
|
43
|
Wang J, Browne L, Slapetova I, Shang F, Lee K, Lynch J, Beretov J, Whan R, Graham PH, Millar EKA. Multiplexed immunofluorescence identifies high stromal CD68 +PD-L1 + macrophages as a predictor of improved survival in triple negative breast cancer. Sci Rep 2021; 11:21608. [PMID: 34732817 PMCID: PMC8566595 DOI: 10.1038/s41598-021-01116-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancer (TNBC) comprises 10-15% of all breast cancers and has a poor prognosis with a high risk of recurrence within 5 years. PD-L1 is an important biomarker for patient selection for immunotherapy but its cellular expression and co-localization within the tumour immune microenvironment and associated prognostic value is not well defined. We aimed to characterise the phenotypes of immune cells expressing PD-L1 and determine their association with overall survival (OS) and breast cancer-specific survival (BCSS). Using tissue microarrays from a retrospective cohort of TNBC patients from St George Hospital, Sydney (n = 244), multiplexed immunofluorescence (mIF) was used to assess staining for CD3, CD8, CD20, CD68, PD-1, PD-L1, FOXP3 and pan-cytokeratin on the Vectra Polaris™ platform and analysed using QuPath. Cox multivariate analyses showed high CD68+PD-L1+ stromal cell counts were associated with improved prognosis for OS (HR 0.56, 95% CI 0.33-0.95, p = 0.030) and BCSS (HR 0.47, 95% CI 0.25-0.88, p = 0.018) in the whole cohort and in patients receiving chemotherapy, improving incrementally upon the predictive value of PD-L1+ alone for BCSS. These data suggest that CD68+PD-L1+ status can provide clinically useful prognostic information to identify sub-groups of patients with good or poor prognosis and guide treatment decisions in TNBC.
Collapse
Affiliation(s)
- James Wang
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
| | - Lois Browne
- Cancer Care Centre, St George Hospital, Kogarah, Australia
| | - Iveta Slapetova
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Kensington, Australia
| | - Fei Shang
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Kensington, Australia
| | - Kirsty Lee
- Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jodi Lynch
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
- Cancer Care Centre, St George Hospital, Kogarah, Australia
| | - Julia Beretov
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- Department of Anatomical Pathology, New South Wales Health Pathology, St George Hospital, Kogarah, Australia
| | - Renee Whan
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Kensington, Australia
| | - Peter H Graham
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia
- Cancer Care Centre, St George Hospital, Kogarah, Australia
| | - Ewan K A Millar
- St George and Sutherland Clinical School, University of New South Wales Sydney, Kensington, Australia.
- Department of Anatomical Pathology, New South Wales Health Pathology, St George Hospital, Kogarah, Australia.
- Faculty of Medicine and Health Sciences, Western Sydney University, Campbelltown, Australia.
- University of Technology, Sydney, Australia.
| |
Collapse
|
44
|
Xu Z, Xiang L, Wang R, Xiong Y, Zhou H, Gu H, Wang J, Peng L. Bioinformatic Analysis of Immune Significance of RYR2 Mutation in Breast Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8072796. [PMID: 34888385 PMCID: PMC8651385 DOI: 10.1155/2021/8072796] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Currently, immunotherapy is widely used for breast cancer (BC) patients, and tumor mutation burden (TMB) is regarded as a valuable independent predictor of response to immunotherapy. However, specific gene mutations and their relationship with TMB and tumor-infiltrating immune cells in BC are not fully understood. METHODS Comprehensive bioinformatic analyses were performed using data from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) datasets. Survival curves were analyzed via Kaplan-Meier analysis. Univariate and multivariate Cox regression analyses were used for prognosis analysis. Gene set enrichment analysis (GSEA) was performed to explore regulatory mechanisms and functions. The CIBERSORT algorithm was used to calculate the tumor-infiltrating immune cell fractions. RESULTS We analyzed somatic mutation data of BC from TCGA and ICGC datasets and found that 19 frequently mutated genes were reported in both cohorts, namely, SPTA1, TTN, MUC17, MAP3K1, CDH1, FAT3, SYNE1, FLG, HMCN1, RYR2 (ryanodine receptor 2), GATA3, MUC4, PIK3CA, KMT2C, TP53, PTEN, ZFHX4, MUC16, and USH2A. Among them, we observed that RYR2 mutation was significantly associated with higher TMB and better clinical prognosis. Moreover, GSEA revealed that RYR2 mutation-enriched signaling pathways were related to immune-associated pathways. Furthermore, based on the CIBERSORT algorithm, we found that RYR2 mutation enhanced the antitumor immune response by enriching CD8+ T cells, activated memory CD4+ T cells, and M1 macrophages. CONCLUSION RYR2 is frequently mutated in BC, and its mutation is related to increased TMB and promotes antitumor immunity; thus, RYR2 may serve as a valuable biomarker to predict the immune response.
Collapse
Affiliation(s)
- Zhiquan Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ling Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Rong Wang
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Yongfu Xiong
- Department of Hepatobiliary Surgery, The Affiliated Hospital of North Sichuan Medical College, Sichuan 637000, China
| | - He Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Sichuan 637000, China
| | - Haitao Gu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jijian Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linglong Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
45
|
Roussos Torres ET, Rafie C, Wang C, Lim D, Brufsky A, LoRusso P, Eder JP, Chung V, Downs M, Geare M, Piekarz R, Streicher H, Anforth L, Rudek MA, Zhu Q, Besharati S, Cimino-Mathews A, Anders RA, Stearns V, Jaffee EM, Connolly RM. Phase I Study of Entinostat and Nivolumab with or without Ipilimumab in Advanced Solid Tumors (ETCTN-9844). Clin Cancer Res 2021; 27:5828-5837. [PMID: 34135021 PMCID: PMC8563383 DOI: 10.1158/1078-0432.ccr-20-5017] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/28/2021] [Accepted: 06/11/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Epigenetic modulators improve immune checkpoint inhibitor (ICI) efficacy and increase CD8+ effector:FoxP3+ regulatory T cell ratios in preclinical models. We conducted a multicenter phase I clinical trial combining the histone deacetylase inhibitor entinostat with nivolumab ± ipilimumab in advanced solid tumors. PATIENTS AND METHODS Patients received an entinostat run-in (5 mg, weekly × 2) prior to the addition of ICIs. Dose escalation followed a modified 3+3 design [dose level (DL)1/2: entinostat + nivolumab; DL 3/4: entinostat + nivolumab + ipilimumab]. Blood and tissue samples were collected at baseline, after entinostat run-in, and after 8 weeks of combination therapy. Primary endpoints included safety and tolerability, and the recommended phase II dose (RP2D). Secondary endpoints included antitumor activity and change in tumor CD8/FoxP3 ratio pre- and post-therapy. RESULTS Thirty-three patients were treated across four dose levels. Treatment-related adverse events (AE) included fatigue (65%), nausea (41%), anemia (38%), diarrhea (26%), and anorexia (26%). Grade 3/4 AEs included fatigue (n = 7, 21%), anemia (n = 9, 27%), and neutropenia (n = 4, 12%). The RP2D was 3 mg entinostat weekly, 3 mg/kg every 2 weeks nivolumab, and 1 mg/kg every 6 weeks ipilimumab (max four doses). The objective response rate by RECIST 1.1 was 16%, including a complete response in triple-negative breast cancer. A statistically significant increase in CD8/FoxP3 ratio was seen following the addition of ICIs to entinostat, but not post-entinostat alone. CONCLUSIONS The combination of entinostat with nivolumab ± ipilimumab was safe and tolerable with expected rates of immune-related AEs. Preliminary evidence of both clinical efficacy and immune modulation supports further investigation.
Collapse
Affiliation(s)
- Evanthia T Roussos Torres
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Christine Rafie
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- University of Miami Miller School of Medicine, Miami, Florida
| | - Chenguang Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - David Lim
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Adam Brufsky
- University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, Pennsylvania
| | | | | | | | - Melinda Downs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Molly Geare
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Richard Piekarz
- Cancer Therapy Evaluation Program (CTEP), NCI, Bethesda, Maryland
| | - Howard Streicher
- Cancer Therapy Evaluation Program (CTEP), NCI, Bethesda, Maryland
| | - Leslie Anforth
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- NIH Clinical Center, Bethesda, Maryland
| | - Michelle A Rudek
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Qingfeng Zhu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sepideh Besharati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ashley Cimino-Mathews
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Robert A Anders
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Roisin M Connolly
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland.
- Cancer Research @ UCC, College of Medicine and Health, University College Cork, Ireland
| |
Collapse
|
46
|
Du J, Dong Y, Li Y. Identification and Prognostic Value Exploration of Cyclophosphamide (Cytoxan)-Centered Chemotherapy Response-Associated Genes in Breast Cancer. DNA Cell Biol 2021; 40:1356-1368. [PMID: 34704810 DOI: 10.1089/dna.2021.0077] [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: 12/13/2022] Open
Abstract
In this study, we aimed to explore cyclophosphamide (Cytoxan) response-associated genes and constructed a model to predict the prognosis of breast cancer (BRCA) patients. Samples obtained from TCGA and GEO databases were subjected to Weighted Gene Coexpression Network Analysis (WGCNA) and univariate Cox and LASSO Cox regression analysis to identify and validate the Cytoxan response-related prognostic signature. Moreover, multivariate Cox regression analysis was performed to analyze the independence of factors, and the nomogram model was constructed by including all the independent factors. WGCNA revealed that 159 genes are significantly correlated with Cytoxan response in BRCA samples, and the samples with a different prognosis could be effectively distinguished based on the expression of those 159 genes. Ten genes were further selected to be related to the prognosis of BRCA patients, including PCDHB2, GRIK2, FRMD7, CCSER1, PCDHGA1, PCDHA1, LRRC37A6P, PCDHGA12, ZNF486, and PCDHGB5, based on the Risk Score model. Among them, PCDHA1 expression was validated in cells and patient samples. Multivariate Cox regression analysis confirmed that the Risk Score is an independent factor. Furthermore, the nomogram model showed that the predicted survival probability is closely related to the actual survival probability. In conclusion, we identified 159 genes potentially correlated with the Cytoxan response of BRCA patients, which had prognostic value in BRCA.
Collapse
Affiliation(s)
- Jiawei Du
- Department of Medicine, Soochow University, Suzhou, China.,Department of Ultrasonography, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yanru Dong
- Department of Clinical Laboratory, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yuhong Li
- Department of Ultrasonography, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
47
|
Das A, Agarwal P, Jain GK, Aggarwal G, Lather V, Pandita D. Repurposing drugs as novel triple negative breast cancer therapeutics. Anticancer Agents Med Chem 2021; 22:515-550. [PMID: 34674627 DOI: 10.2174/1871520621666211021143255] [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] [Received: 01/10/2021] [Revised: 05/23/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Among all the types of breast cancer (BC), triple negative breast cancer (TNBC) is the most aggressive form having high metastasis and recurrence rate with limited treatment options. Conventional treatments such as chemotherapy and radiotherapy have lots of toxic side effects and also no FDA approved therapies are available till now. Repurposing of old clinically approved drugs towards various targets of TNBC is the new approach with lesser side effects and also leads to successful inexpensive drug development with less time consuming. Medicinal plants containg various phytoconstituents (flavonoids, alkaloids, phenols, essential oils, tanins, glycosides, lactones) plays very crucial role in combating various types of diseases and used in drug development process because of having lesser side effects. OBJECTIVE The present review focuses in summarization of various categories of repurposed drugs against multitarget of TNBC and also summarizes the phytochemical categories that targets TNBC singly or in combination with synthetic old drugs. METHODS Literature information was collected from various databases such as Pubmed, Web of Science, Scopus and Medline to understand and clarify the role and mechanism of repurposed synthetic drugs and phytoconstituents aginst TNBC by using keywords like "breast cancer", "repurposed drugs", "TNBC" and "phytoconstituents". RESULTS Various repurposed drugs and phytochemicals targeting different signaling pathways that exerts their cytotoxic activities on TNBC cells ultimately leads to apoptosis of cells and also lowers the recurrence rate and stops the metastasis process. CONCLUSION Inhibitory effects seen in different levels, which provides information and evidences to researchers towards drug developments process and thus further more investigations and researches need to be taken to get the better therapeutic treatment options against TNBC.
Collapse
Affiliation(s)
- Amiya Das
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, 201313. India
| | - Pallavi Agarwal
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, 201313. India
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017. India
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017. India
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector-125, Noida, 201313. India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017. India
| |
Collapse
|
48
|
Bräutigam K, Kabore-Wolff E, Hussain AF, Polack S, Rody A, Hanker L, Köster F. Inhibitors of PD-1/PD-L1 and ERK1/2 impede the proliferation of receptor positive and triple-negative breast cancer cell lines. J Cancer Res Clin Oncol 2021; 147:2923-2933. [PMID: 34185141 PMCID: PMC8397671 DOI: 10.1007/s00432-021-03694-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/10/2021] [Indexed: 12/31/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is characterized by an unfavorable prognosis and missing systemic therapeutic approaches beside chemotherapy. Targeting the immune checkpoint PD-1/PD-L1 showed promising results in breast cancer and especially in TNBC. The extracellular signal-regulated kinase 1/2 (ERK1/2) is an important driver of carcinogenesis. Here, the effect of combined PD-1/PD-L1 and ERK1/2 inhibitor treatment is investigated of cell growth and intracellular impact of breast cancer cell lines. METHODS The IC50 values of each inhibitor and the effect of combined treatment were determined in three TNBC cell lines of different subtypes and one non-TNBC cell line. Phospho-specific antibodies were used in western blot analyses to investigate an effect on ERK1/2 activation. Expressions of immune modulatory and cell cycle-associated genes were examined by quantitative reverse transcription PCR. RESULTS Both inhibitors PD-1/PD-L1 and ERK1/2 impeded the proliferation of TNBC to a higher extent than of non-TNBC. By combined treatment, cell lines were inhibited either synergistically or additively. ERK1/2 and S6 phosphorylation were reduced and expressions of c-Fos and FosL were diminished after ERK1/2 inhibitor as single and combined treatment. Between genes involved in immune modulation, IL-8 was upregulated in TNBC cells after combined treatment. CONCLUSION In conclusion, combination of PD-1/PD-L1 and ERK1/2 inhibitors showed favorable effects for a new therapy strategy, with better results in TNBC cell lines than in non-TNBC cells. The effects have to be validated in models that can reflect the interaction between immune and tumor cells like the situation in the tumor micro-environment.
Collapse
Affiliation(s)
- Karen Bräutigam
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| | - Elodie Kabore-Wolff
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Ahmad Fawzi Hussain
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Giessen, Germany
| | - Stephan Polack
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Achim Rody
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Lars Hanker
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Frank Köster
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| |
Collapse
|
49
|
Chumsri S, Sokol ES, Soyano-Muller AE, Parrondo RD, Reynolds GA, Nassar A, Thompson EA. Durable Complete Response With Immune Checkpoint Inhibitor in Breast Cancer With High Tumor Mutational Burden and APOBEC Signature. J Natl Compr Canc Netw 2021; 18:517-521. [PMID: 32380464 DOI: 10.6004/jnccn.2020.7543] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/30/2020] [Indexed: 11/17/2022]
Abstract
Increasing data support the importance of preexisting host immune response and neoantigen burden for determining response to immune checkpoint inhibitors (ICIs). In lung cancer and melanoma, tumor mutational burden (TMB) has emerged as an independent biomarker for ICI response. However, the significance of TMB in breast cancer, particularly in the context of PD-L1 negativity, remains unclear. This report describes a patient with HER2-negative breast cancer with high TMB and an apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC) trinucleotide signature; her disease was refractory to multiple lines of treatments but achieved durable complete response using ICIs and capecitabine. Additional analysis of the tumor revealed a low amount of stromal tumor-infiltrating lymphocytes (sTILs) and PD-L1 negativity, reflecting a poor preexisting host immune response. In collaboration with Foundation Medicine, comprehensive genomic profiling from 14,867 patients with breast cancer with the FoundationOne test was evaluated. Using the cutoff of ≥10 mutations/megabase (mut/Mb) for high TMB, PD-L1 positivity and TMB-high populations were not significantly overlapping (odds ratio, 1.02; P=.87). Up to 79% of TMB-high tumors with >20 mut/Mb were PD-L1-negative. Our study highlights that despite having low TILs and PD-L1 negativity, some patients may still experience response to ICIs.
Collapse
Affiliation(s)
- Saranya Chumsri
- Robert and Monica Jacoby Center for Breast Health, Mayo Clinic, Jacksonville, Florida
| | | | - Aixa E Soyano-Muller
- Robert and Monica Jacoby Center for Breast Health, Mayo Clinic, Jacksonville, Florida
| | - Ricardo D Parrondo
- Robert and Monica Jacoby Center for Breast Health, Mayo Clinic, Jacksonville, Florida
| | - Gina A Reynolds
- Robert and Monica Jacoby Center for Breast Health, Mayo Clinic, Jacksonville, Florida
| | | | | |
Collapse
|
50
|
Crosstalk between Tumor-Infiltrating Immune Cells and Cancer-Associated Fibroblasts in Tumor Growth and Immunosuppression of Breast Cancer. J Immunol Res 2021; 2021:8840066. [PMID: 34337083 PMCID: PMC8294979 DOI: 10.1155/2021/8840066] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/04/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023] Open
Abstract
Signals from the tumor microenvironment (TME) have a profound influence on the maintenance and progression of cancers. Chronic inflammation and the infiltration of immune cells in breast cancer (BC) have been strongly associated with early carcinogenic events and a switch to a more immunosuppressive response. Cancer-associated fibroblasts (CAFs) are the most abundant stromal component and can modulate tumor progression according to their secretomes. The immune cells including tumor-infiltrating lymphocytes (TILs) (cytotoxic T cells (CTLs), regulatory T cells (Tregs), and helper T cell (Th)), monocyte-infiltrating cells (MICs), myeloid-derived suppressor cells (MDSCs), mast cells (MCs), and natural killer cells (NKs) play an important part in the immunological balance, fluctuating TME between protumoral and antitumoral responses. In this review article, we have summarized the impact of these immunological players together with CAF secreted substances in driving BC progression. We explain the crosstalk of CAFs and tumor-infiltrating immune cells suppressing antitumor response in BC, proposing these cellular entities as predictive markers of poor prognosis. CAF-tumor-infiltrating immune cell interaction is suggested as an alternative therapeutic strategy to regulate the immunosuppressive microenvironment in BC.
Collapse
|