1
|
Foldi J, Blenman KRM, Marczyk M, Gunasekharan V, Polanska A, Gee R, Davis M, Kahn AM, Silber A, Pusztai L. Peripheral blood immune parameters, response, and adverse events after neoadjuvant chemotherapy plus durvalumab in early-stage triple-negative breast cancer. Breast Cancer Res Treat 2024; 208:369-377. [PMID: 39002068 DOI: 10.1007/s10549-024-07426-3] [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: 02/28/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE We evaluated T- and B-cell receptor (TCR and BCR) repertoire diversity and 38 serum cytokines in pre- and post-treatment peripheral blood of 66 patients with triple-negative breast cancer (TNBC) who received neoadjuvant chemotherapy plus durvalumab and assessed associations with pathologic response and immune-related adverse events (irAEs) during treatment. METHODS Genomic DNA was isolated from buffy coat for TCR and BCR clonotype profiling using the Immunoseq platform and diversity was quantified with Pielou's evenness index. MILLIPLEX MAP Human Cytokine/Chemokine Magnetic Bead Panel was used to measure serum cytokine levels, which were compared between groups using moderated t-statistic with Benjamini-Hochberg correction for multiple testing. RESULTS TCR and BCR diversity was high (Pielou's index > 0.75) in all samples. Baseline receptor diversities and change in diversity pre- and post-treatment were not associated with pathologic response or irAE status, except for BCR diversity that was significantly lower post-treatment in patients who developed irAE (unadjusted p = 0.0321). Five cytokines increased after treatment in patients with pathologic complete response (pCR) but decreased in patients with RD, most prominently IL-8. IFNγ, IL-7, and GM-CSF levels were higher in pre-treatment than in post-treatment samples of patients who developed irAEs but were lower in those without irAEs. CONCLUSION Baseline peripheral blood cytokine levels may predict irAEs in patients treated with immune checkpoint inhibitors and chemotherapy, and increased post-treatment B-cell clonal expansion might mediate irAEs.
Collapse
Affiliation(s)
- Julia Foldi
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA.
- Division of Hematology and Oncology, Department of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- University of Pittsburgh School of Medicine, 300 Halket Street, Room 3524, Pittsburgh, PA, USA.
| | - Kim R M Blenman
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Michal Marczyk
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Vignesh Gunasekharan
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Alicja Polanska
- Mullard Space Science Laboratory, University College London, London, UK
| | - Renelle Gee
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Mya Davis
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Adriana M Kahn
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Andrea Silber
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Lajos Pusztai
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
2
|
Kok M, Gielen RJ, Adams S, Lennerz JK, Sharma P, Loibl S, Reardon E, Sonke G, Linn S, Delaloge S, Lacombe D, Robinson T, Badve S, Martin M, Balko JM, Ignatiadis M, Curigliano G, Wolff AC, Mittendorf EA, Loi S, Pusztai L, Tolaney SM, Salgado R. Academic Uphill Battle to Personalize Treatment for Patients With Stage II/III Triple-Negative Breast Cancer. J Clin Oncol 2024; 42:3523-3529. [PMID: 39038259 DOI: 10.1200/jco.24.00372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/17/2024] [Accepted: 05/08/2024] [Indexed: 07/24/2024] Open
Affiliation(s)
- Marleen Kok
- Departments of Medical Oncology and Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Robbert-Jan Gielen
- Departments of Medical Oncology and Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sylvia Adams
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, Manhattan, NY
| | | | | | - Sibylle Loibl
- GBG Forschungs GmbH, Neu-Isenburg, Germany
- Centre for Haematology and Oncology, Bethanien, and Goethe University, Frankfurt, Germany
| | | | - Gabe Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sabine Linn
- Departments of Medical Oncology and Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Suzette Delaloge
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Denis Lacombe
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Tim Robinson
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Winship Cancer Institute, Atlanta, GA
| | - Miguel Martin
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, CIBERONC, GEICAM, Madrid, Spain
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Michail Ignatiadis
- Breast Medical Oncology Clinic, Institut Jules Bordet, Universite Libre de Bruxelles, Bruxelles, Belgium
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - Antonio C Wolff
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- The Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Lajos Pusztai
- Department of Medicine, Yale Cancer Center, Yale University, New Haven, CT
| | | | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, PeterMacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Mittendorf EA, Tolaney SM. Neoadjuvant Immunotherapy-From Trials to Practice. JAMA Oncol 2024; 10:1319-1321. [PMID: 39207783 DOI: 10.1001/jamaoncol.2024.2924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Affiliation(s)
- Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
4
|
Sun X, Kennedy LC, Gonzalez-Ericsson PI, Sanchez V, Sanders M, Perou CM, Troester MA, Balko JM, Reid SA. Associations of Immune Checkpoint Predictive Biomarkers (MHC-I and MHC-II) with Clinical and Molecular Features in a Diverse Breast Cancer Cohort. Clin Cancer Res 2024; 30:4077-4081. [PMID: 39007881 PMCID: PMC11398978 DOI: 10.1158/1078-0432.ccr-24-1286] [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: 04/23/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
PURPOSE Immunotherapy (IO) in triple-negative breast cancer (TNBC) has improved survival outcomes, with promising improvements in pCR rates among early high-risk hormone receptor (HR)+/HER2- breast cancers. However, biomarkers are needed to select patients likely to benefit from IO. MHC-I and tumor-specific MHC-II (tsMHC-II) expression are candidate biomarkers for PD-(L)1 checkpoint inhibition but existing data from clinical trials included limited racial/ethnic diversity. EXPERIMENTAL DESIGN We performed multiplexed immunofluorescence assays in the Carolina Breast Cancer Study (CBCS; n = 1,628, 48% Black, 52% non-Black). Intrinsic subtype and P53 mutant-like status were identified using RNA-based multigene assays. We ranked participants based on tumoral MHC-I intensity (top 33% categorized as "MHC-Ihigh") and MHC-II+ (≥5% of tumor cells as tsMHC-II+). MHC-I/II were evaluated in association with clinicopathological features by race. RESULTS Black participants had higher frequency of TNBC (25% vs. 12.5%, P ≤ 0.001) and basal-like (30% vs. 14%, P ≤ 0.001) tumors overall, and higher frequency of basal-like (11% vs. 5.5%, P = 0.002) and TP53 mutant tumors (26% vs. 17%, P = 0.002) among HR+/HER2-. The frequency of tsMHC-II+ was higher in HR+/HER2- Black participants (7.9% vs. 4.9%, P = 0.04). Black participants also had higher frequency of MHC-Ihigh (38.7% vs. 28.2%, P < 0.001), which was significant among HR+/HER2- (28.2% vs. 22.1%, P = 0.02). CONCLUSIONS In this diverse study population, MHC-I and MHC-II tumor cell expression were more highly expressed in HR+/HER2- tumors from Black women, underscoring the importance of diverse and equitable enrollment in future IO trials.
Collapse
Affiliation(s)
- Xiaopeng Sun
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Laura C Kennedy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Paula I Gonzalez-Ericsson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Violeta Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Melinda Sanders
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | | | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sonya A Reid
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|
5
|
Beutgen VM, Shinkevich V, Pörschke J, Meena C, Steitz AM, Pogge von Strandmann E, Graumann J, Gómez-Serrano M. Secretome Analysis Using Affinity Proteomics and Immunoassays: A Focus on Tumor Biology. Mol Cell Proteomics 2024; 23:100830. [PMID: 39147028 PMCID: PMC11417252 DOI: 10.1016/j.mcpro.2024.100830] [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: 02/29/2024] [Revised: 07/20/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
The study of the cellular secretome using proteomic techniques continues to capture the attention of the research community across a broad range of topics in biomedical research. Due to their untargeted nature, independence from the model system used, historically superior depth of analysis, as well as comparative affordability, mass spectrometry-based approaches traditionally dominate such analyses. More recently, however, affinity-based proteomic assays have massively gained in analytical depth, which together with their high sensitivity, dynamic range coverage as well as high throughput capabilities render them exquisitely suited to secretome analysis. In this review, we revisit the analytical challenges implied by secretomics and provide an overview of affinity-based proteomic platforms currently available for such analyses, using the study of the tumor secretome as an example for basic and translational research.
Collapse
Affiliation(s)
- Vanessa M Beutgen
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany; Core Facility Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
| | - Veronika Shinkevich
- Institute of Pharmacology, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
| | - Johanna Pörschke
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Celina Meena
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Anna M Steitz
- Translational Oncology Group, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany; Core Facility Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany.
| | - María Gómez-Serrano
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.
| |
Collapse
|
6
|
Feng HR, Shen XN, Zhu XM, Zhong WT, Zhu DX, Zhao J, Chen YJ, Shen F, Liu K, Liang L. Unveiling major histocompatibility complex-mediated pan-cancer immune features by integrated single-cell and bulk RNA sequencing. Cancer Lett 2024; 597:217062. [PMID: 38878852 DOI: 10.1016/j.canlet.2024.217062] [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: 03/30/2024] [Revised: 05/22/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed cancer therapy, yet persistent challenges such as low response rate and significant heterogeneity necessitate attention. The pivotal role of the major histocompatibility complex (MHC) in ICI efficacy, its intricate impacts and potentials as a prognostic marker, warrants comprehensive exploration. This study integrates single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, and spatial transcriptomic analyses to unveil pan-cancer immune characteristics governed by the MHC transcriptional feature (MHC.sig). Developed through scRNA-seq analysis of 663,760 cells across diverse cohorts and validated in 30 solid cancer types, the MHC.sig demonstrates a robust correlation between immune-related genes and infiltrating immune cells, highlighting its potential as a universal pan-cancer marker for anti-tumor immunity. Screening the MHC.sig for therapeutic targets using CRISPR data identifies potential genes for immune therapy synergy and validates its predictive efficacy for ICIs responsiveness across diverse datasets and cancer types. Finally, analysis of cellular communication patterns reveals interactions between C1QC+macrophages and malignant cells, providing insights into potential therapeutic agents and their sensitivity characteristics. This comprehensive analysis positions the MHC.sig as a promising marker for predicting immune therapy outcomes and guiding combinatorial therapeutic strategies.
Collapse
Affiliation(s)
- Hao-Ran Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Xiao-Nan Shen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Xiao-Ming Zhu
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200082, People's Republic of China
| | - Wen-Tao Zhong
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, People's Republic of China
| | - De-Xiang Zhu
- Department of Colorectal Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Ji Zhao
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, People's Republic of China
| | - Yan-Jie Chen
- Department of Gastroenterology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, People's Republic of China; Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Feng Shen
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, People's Republic of China.
| | - Kun Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| | - Li Liang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
7
|
Burdett NL, Willis MO, Pandey A, Twomey L, Alaei S, Bowtell DDL, Christie EL. Timing of whole genome duplication is associated with tumor-specific MHC-II depletion in serous ovarian cancer. Nat Commun 2024; 15:6069. [PMID: 39025846 PMCID: PMC11258338 DOI: 10.1038/s41467-024-50137-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: 11/08/2023] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
Whole genome duplication is frequently observed in cancer, and its prevalence in our prior analysis of end-stage, homologous recombination deficient high grade serous ovarian cancer (almost 80% of samples) supports the notion that whole genome duplication provides a fitness advantage under the selection pressure of therapy. Here, we therefore aim to identify potential therapeutic vulnerabilities in primary high grade serous ovarian cancer with whole genome duplication by assessing differentially expressed genes and pathways in 79 samples. We observe that MHC-II expression is lowest in tumors which have acquired whole genome duplication early in tumor evolution, and further demonstrate that reduced MHC-II expression occurs in subsets of tumor cells rather than in canonical antigen-presenting cells. Early whole genome duplication is also associated with worse patient survival outcomes. Our results suggest an association between the timing of whole genome duplication, MHC-II expression and clinical outcome in high grade serous ovarian cancer that warrants further investigation for therapeutic targeting.
Collapse
Affiliation(s)
- Nikki L Burdett
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Box Hill Hospital, Eastern Health, Box Hill, VIC, 3128, Australia
| | | | - Ahwan Pandey
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Laura Twomey
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sara Alaei
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3168, Australia
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
8
|
Rayson VC, Harris MA, Savas P, Hun ML, Virassamy B, Salgado R, Loi S. The anti-cancer immune response in breast cancer: current and emerging biomarkers and treatments. Trends Cancer 2024; 10:490-506. [PMID: 38521654 DOI: 10.1016/j.trecan.2024.02.008] [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: 08/25/2023] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
Triple-negative breast cancers (TNBCs) exhibit heightened T cell infiltration, contributing to an enhanced response to immune checkpoint blockade (ICB) compared with other subtypes. An immune-rich immune microenvironment correlates with improved prognosis in early and advanced TNBC. Combination chemotherapy and ICB is now the standard of care in early- and late-stage TNBC. Although programmed death ligand-1 (PD-L1) positivity predicts ICB response in advanced stages, its role in early-stage disease remains uncertain. Despite neoadjuvant ICB becoming common in early-stage TNBC, the necessity of adjuvant ICB after surgery remains unclear. Understanding the molecular basis of the immune response in breast cancer is vital for precise biomarkers for ICB and effective combination therapy strategies.
Collapse
Affiliation(s)
- Victoria C Rayson
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael A Harris
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter Savas
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael L Hun
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Balaji Virassamy
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Roberto Salgado
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
| | - Sherene Loi
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| |
Collapse
|
9
|
Li L, Fei Y, Dong T, Song Y, Chen X, Zhang H, Zhou H, Liang M, Tang J. IFI30 as a key regulator of PDL1 immunotherapy prognosis in breast cancer. Int Immunopharmacol 2024; 133:112093. [PMID: 38669947 DOI: 10.1016/j.intimp.2024.112093] [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: 01/08/2024] [Revised: 03/30/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND IFI30 is a lysosomal thiol reductase involved in antigen presentation and immune regulation in various cancers, including breast cancer. Despite its known involvement, the precise mechanism, function, and relationship with the PD-L1 axis and immune response remain unclear. METHODS We conducted an extensive investigation into IFI30 mRNA expression in breast cancer utilizing data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. Furthermore, we characterized IFI30 mRNA expression across various cell types using publicly available single-cell RNA sequencing datasets, and assessed protein expression through immunohistochemistry using an in-house breast cancer tissue microarray. Functional experiments were performed to elucidate the effects of IFI30 overexpression on PD-L1 expression and inhibitory efficacy in both macrophages and breast tumor cells. RESULTS Our study unveiled a marked upregulation of IFI30 expression in breast cancer tissues compared to their normal counterparts, with notable associations identified with tumor stage and prognosis. Additionally, IFI30 expression demonstrated significant correlations with various immune-related signaling pathways, encompassing peptide antigen binding, cytokine binding, and MHC class II presentation. Notably, breast cancer samples exhibiting high IFI30 expression in tumor cells displayed high PD-L1 expression on corresponding cells, alongside a diminished ratio of CD8 + T cell infiltration within the tumor microenvironment. Furthermore, ectopic knockdown of IFI30 in both tumor cells and macrophages resulted in a reduction of PD-L1 expression, while conversely, overexpression of IFI30 led to an increase in PD-L1 expression. CONCLUSIONS This study offers new insights into the involvement of IFI30 in breast cancer, elucidating its interplay with the PD-L1 axis and immune response dynamics. Our findings suggest that modulation of the IFI30-PD-L1 axis could serve as a promising strategy for regulating T cells infiltration in breast cancer thus treating breast cancer.
Collapse
Affiliation(s)
- Lei Li
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China
| | - Yinjiao Fei
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China
| | - Tianfu Dong
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China; Lianyungang Clinical College of Nanjing Medical University, The First People Hospital of Lianyungang City, Lianyungang, Jiangsu 222061, PR China
| | - Yuxin Song
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China
| | - Xiu Chen
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China
| | - Heda Zhang
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China
| | - Honglei Zhou
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China.
| | - Mingxing Liang
- Department of Thyroid Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, PR China.
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, 300 Guanzhou Road, Nanjing 210029, PR China.
| |
Collapse
|
10
|
Xu L, Saunders K, Huang SP, Knutsdottir H, Martinez-Algarin K, Terrazas I, Chen K, McArthur HM, Maués J, Hodgdon C, Reddy SM, Roussos Torres ET, Xu L, Chan IS. A comprehensive single-cell breast tumor atlas defines epithelial and immune heterogeneity and interactions predicting anti-PD-1 therapy response. Cell Rep Med 2024; 5:101511. [PMID: 38614094 PMCID: PMC11148512 DOI: 10.1016/j.xcrm.2024.101511] [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: 03/09/2023] [Revised: 02/20/2024] [Accepted: 03/20/2024] [Indexed: 04/15/2024]
Abstract
We present an integrated single-cell RNA sequencing atlas of the primary breast tumor microenvironment (TME) containing 236,363 cells from 119 biopsy samples across eight datasets. In this study, we leverage this resource for multiple analyses of immune and cancer epithelial cell heterogeneity. We define natural killer (NK) cell heterogeneity through six subsets in the breast TME. Because NK cell heterogeneity correlates with epithelial cell heterogeneity, we characterize epithelial cells at the level of single-gene expression, molecular subtype, and 10 categories reflecting intratumoral transcriptional heterogeneity. We develop InteractPrint, which considers how cancer epithelial cell heterogeneity influences cancer-immune interactions. We use T cell InteractPrint to predict response to immune checkpoint inhibition (ICI) in two breast cancer clinical trials testing neoadjuvant anti-PD-1 therapy. T cell InteractPrint was predictive of response in both trials versus PD-L1 (AUC = 0.82, 0.83 vs. 0.50, 0.72). This resource enables additional high-resolution investigations of the breast TME.
Collapse
Affiliation(s)
- Lily Xu
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kaitlyn Saunders
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shao-Po Huang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hildur Knutsdottir
- Department of Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA
| | - Kenneth Martinez-Algarin
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Isabella Terrazas
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kenian Chen
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Heather M McArthur
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Sangeetha M Reddy
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Evanthia T Roussos Torres
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Isaac S Chan
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
11
|
Sammut SJ, Galson JD, Minter R, Sun B, Chin SF, De Mattos-Arruda L, Finch DK, Schätzle S, Dias J, Rueda OM, Seoane J, Osbourn J, Caldas C, Bashford-Rogers RJM. Predictability of B cell clonal persistence and immunosurveillance in breast cancer. Nat Immunol 2024; 25:916-924. [PMID: 38698238 PMCID: PMC11065701 DOI: 10.1038/s41590-024-01821-0] [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: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 05/05/2024]
Abstract
B cells and T cells are important components of the adaptive immune system and mediate anticancer immunity. The T cell landscape in cancer is well characterized, but the contribution of B cells to anticancer immunosurveillance is less well explored. Here we show an integrative analysis of the B cell and T cell receptor repertoire from individuals with metastatic breast cancer and individuals with early breast cancer during neoadjuvant therapy. Using immune receptor, RNA and whole-exome sequencing, we show that both B cell and T cell responses seem to coevolve with the metastatic cancer genomes and mirror tumor mutational and neoantigen architecture. B cell clones associated with metastatic immunosurveillance and temporal persistence were more expanded and distinct from site-specific clones. B cell clonal immunosurveillance and temporal persistence are predictable from the clonal structure, with higher-centrality B cell antigen receptors more likely to be detected across multiple metastases or across time. This predictability was generalizable across other immune-mediated disorders. This work lays a foundation for prioritizing antibody sequences for therapeutic targeting in cancer.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/immunology
- B-Lymphocytes/immunology
- Immunologic Surveillance
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- T-Lymphocytes/immunology
- Monitoring, Immunologic
- Exome Sequencing
- Antigens, Neoplasm/immunology
- Neoplasm Metastasis
- Clone Cells
Collapse
Affiliation(s)
- Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
- The Royal Marsden Hospital NHS Foundation Trust, London, UK.
| | | | | | - Bo Sun
- Wellcome Centre for Human Genetics, Oxford, UK
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Leticia De Mattos-Arruda
- IrsiCaixa, Germans Trias i Pujol University Hospital, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | | | | | | | - Oscar M Rueda
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Joan Seoane
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Institució Catalana de Recerca i Estudis Avançats (ICREA), Universitat Autònoma de Barcelona (UAB), CIBERONC, Barcelona, Spain
| | | | - Carlos Caldas
- School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| | - Rachael J M Bashford-Rogers
- Wellcome Centre for Human Genetics, Oxford, UK.
- Department of Biochemistry, University of Oxford, Oxford, UK.
- Oxford Cancer Centre, Oxford, UK.
| |
Collapse
|
12
|
Seager RJ, Ko H, Pabla S, Senosain MF, Kalinski P, Van Roey E, Gao S, Strickland KC, Previs RA, Nesline MK, Hastings S, Zhang S, Conroy JM, Jensen TJ, Eisenberg M, Caveney B, Severson EA, Ramkissoon S, Gandhi S. Immunologic Factors Associated with Differential Response to Neoadjuvant Chemoimmunotherapy in Triple-Negative Breast Cancer. J Pers Med 2024; 14:481. [PMID: 38793063 PMCID: PMC11122407 DOI: 10.3390/jpm14050481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Background: KEYNOTE-522 resulted in FDA approval of the immune checkpoint inhibitor pembrolizumab in combination with neoadjuvant chemotherapy for patients with early-stage, high-risk, triple-negative breast cancer (TNBC). Unfortunately, pembrolizumab is associated with several immune-related adverse events (irAEs). We aimed to identify potential tumor microenvironment (TME) biomarkers which could predict patients who may attain pathological complete response (pCR) with chemotherapy alone and be spared the use of anti-PD-1 immunotherapy. Methods: Comprehensive immune profiling, including RNA-seq gene expression assessment of 395 immune genes, was performed on matched FFPE tumor samples from 22 stage I-III TNBC patients (14 patients treated with neoadjuvant chemotherapy alone (NAC) and 8 treated with neoadjuvant chemotherapy combined with pembrolizumab (NAC+I)). Results: Differential gene expression analysis revealed that in the NAC group, IL12B and IL13 were both significantly associated with pCR. In the NAC+I group, LCK and TP63 were significantly associated with pCR. Patients in both treatment groups exhibiting pCR tended to have greater tumor inflammation than non-pCR patients. In the NAC+I group, patients with pCR tended to have greater cell proliferation and higher PD-L1 expression, while in the NAC group, patients with pCR tended to have lower cancer testis antigen expression. Additionally, the NAC+I group trended toward a lower relative dose intensity averaged across all chemotherapy drugs, suggesting that more dose reductions or treatment delays occurred in the NAC+I group than the NAC group. Conclusions: A comprehensive understanding of immunologic factors could potentially predict pCR to chemotherapy alone, enabling the avoidance of the unnecessary treatment of these patients with checkpoint inhibitors.
Collapse
Affiliation(s)
- Robert J. Seager
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Heidi Ko
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Sarabjot Pabla
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Maria-Fernanda Senosain
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Pawel Kalinski
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Erik Van Roey
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Shuang Gao
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Kyle C. Strickland
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Department of Pathology, Duke University Medical Center, Duke Cancer Institute, Durham, NC 27710, USA
| | - Rebecca Ann Previs
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Department of Obstetrics & Gynecology, Duke University Medical Center, Duke Cancer Institute, Division of Gynecologic Oncology, Durham, NC 27710, USA
| | - Mary K. Nesline
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Stephanie Hastings
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Shengle Zhang
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Jeffrey M. Conroy
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Taylor J. Jensen
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | | | | | - Eric A. Severson
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Shakti Ramkissoon
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27710, USA
| | - Shipra Gandhi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| |
Collapse
|
13
|
Massa C, Karn T, Weber K, Schneeweiss A, Hanusch C, Uwe Blohmer J, Zahm D, Jackisch C, van Mackelenbergh M, Thomalla J, Marmé F, Huober J, Müller V, Schem C, Müller A, Stickeler E, Biehl K, Fasching PA, Untch M, Loibl S, Denkert C, Seliger B. Baseline CD4 + and expansion of γδ T cells correlate with response to durvalumab in triple-negative breast cancer patients. Clin Transl Med 2024; 14:e1617. [PMID: 38664548 PMCID: PMC11045558 DOI: 10.1002/ctm2.1617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 04/29/2024] Open
Affiliation(s)
- Chiara Massa
- Institute of Medical ImmunologyMartin Luther University Halle‐WittenbergHalleGermany
- Institute for Translational ImmunologyBrandenburg Medical School Theodor FontaneBrandenburg an der HavelGermany
| | - Thomas Karn
- Department of Obstetrics and GynecologyGoethe UniversityFrankfurtGermany
| | - Karsten Weber
- German Breast Group, GBG Forschungs GmbHNeu‐IsenburgGermany
| | - Andreas Schneeweiss
- Nationales Centrum für TumorerkrankungenUniversitätsklinikum und Deutsches KrebsforschungszentrumHeidelbergGermany
| | | | - Jens Uwe Blohmer
- Gynäkologie mit Brustzentrum der Charite CCMCharité‐Universitätsmedizin BerlinBerlinGermany
| | | | - Christian Jackisch
- Department of Obstetrics and GynecologySana Klinikum OffenbachOffenbachGermany
| | | | | | - Frederik Marmé
- UniversitätsfrauenklinikMedizinische Fakultät Mannheim der Universität HeidelbergHeidelbergGermany
| | - Jens Huober
- Breast CancerCantonal Hospital St.GallenSt. GallenSwitzerland
| | - Volkmar Müller
- Department of Obstetrics and GynecologyUniversitätsklinikum Hamburg‐EppendorfHamburgGermany
| | | | - Anja Müller
- Institute of Medical ImmunologyMartin Luther University Halle‐WittenbergHalleGermany
| | - Elmar Stickeler
- Klinik für Gynäkologie und GeburtsmedizinUniklinik RWTH AachenAachenGermany
| | - Katharina Biehl
- Institute of Medical ImmunologyMartin Luther University Halle‐WittenbergHalleGermany
| | - Peter A. Fasching
- Department of Obstetrics and GynecologyUniversitätsklinikum ErlangenErlangenGermany
| | - Michael Untch
- Department of Obstetrics and GynecologyHELIOS Klinikum Berlin BuchBerlinGermany
| | - Sibylle Loibl
- German Breast Group, GBG Forschungs GmbHNeu‐IsenburgGermany
| | - Carsten Denkert
- Institute of PathologyPhilipps‐University Marburg and University Hospital Marburg (UKGM)MarburgGermany
| | - Barbara Seliger
- Institute of Medical ImmunologyMartin Luther University Halle‐WittenbergHalleGermany
- Institute for Translational ImmunologyBrandenburg Medical School Theodor FontaneBrandenburg an der HavelGermany
| |
Collapse
|
14
|
Song F, Tarantino P, Garrido-Castro A, Lynce F, Tolaney SM, Schlam I. Immunotherapy for Early-Stage Triple Negative Breast Cancer: Is Earlier Better? Curr Oncol Rep 2024; 26:21-33. [PMID: 38198112 DOI: 10.1007/s11912-023-01487-1] [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] [Accepted: 12/11/2023] [Indexed: 01/11/2024]
Abstract
PURPOSE OF REVIEW In this narrative review, we discuss the optimal timing of immune checkpoint inhibitors (ICI) in early triple negative breast cancer (TNBC), the landscape of predictive biomarkers for the use of immunotherapy, and the mounting literature suggesting a benefit for an early use of ICI. RECENT FINDINGS TNBC is associated with a poor prognosis relative to other breast cancer subtypes, and until recently, the treatment of TNBC was limited to cytotoxic chemotherapy. In 2021, the immune-checkpoint inhibitor, pembrolizumab, was approved in combination with neoadjuvant chemotherapy for patients with high-risk early stage TNBC. This approval changed the treatment paradigm of early TNBC concomitantly raised several challenges in clinical practice, pertaining to patient selection, toxicity management, and post-neoadjuvant treatment, among others. The introduction of neoadjuvant chemoimmunotherapy has transformed the treatment landscape for early TNBC. However, several challenges, including patient selection, toxicity management, and the identification of predictive biomarkers, need to be addressed. Future research should focus on refining the timing and duration of immunotherapy, optimizing the chemotherapy partner, and exploring novel predictive biomarkers of response or toxicity.
Collapse
Affiliation(s)
- Fei Song
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Ana Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Filipa Lynce
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ilana Schlam
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA.
- Tufts University, Boston, MA, USA.
| |
Collapse
|
15
|
Gallagher RI, Wulfkuhle J, Wolf DM, Brown-Swigart L, Yau C, O'Grady N, Basu A, Lu R, Campbell MJ, Magbanua MJ, Coppé JP, Asare SM, Sit L, Matthews JB, Perlmutter J, Hylton N, Liu MC, Symmans WF, Rugo HS, Isaacs C, DeMichele AM, Yee D, Pohlmann PR, Hirst GL, Esserman LJ, van 't Veer LJ, Petricoin EF. Protein signaling and drug target activation signatures to guide therapy prioritization: Therapeutic resistance and sensitivity in the I-SPY 2 Trial. Cell Rep Med 2023; 4:101312. [PMID: 38086377 PMCID: PMC10772394 DOI: 10.1016/j.xcrm.2023.101312] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 07/03/2023] [Accepted: 11/14/2023] [Indexed: 12/22/2023]
Abstract
Molecular subtyping of breast cancer is based mostly on HR/HER2 and gene expression-based immune, DNA repair deficiency, and luminal signatures. We extend this description via functional protein pathway activation mapping using pre-treatment, quantitative expression data from 139 proteins/phosphoproteins from 736 patients across 8 treatment arms of the I-SPY 2 Trial (ClinicalTrials.gov: NCT01042379). We identify predictive fit-for-purpose, mechanism-of-action-based signatures and individual predictive protein biomarker candidates by evaluating associations with pathologic complete response. Elevated levels of cyclin D1, estrogen receptor alpha, and androgen receptor S650 associate with non-response and are biomarkers for global resistance. We uncover protein/phosphoprotein-based signatures that can be utilized both for molecularly rationalized therapeutic selection and for response prediction. We introduce a dichotomous HER2 activation response predictive signature for stratifying triple-negative breast cancer patients to either HER2 or immune checkpoint therapy response as a model for how protein activation signatures provide a different lens to view the molecular landscape of breast cancer and synergize with transcriptomic-defined signatures.
Collapse
Affiliation(s)
- Rosa I Gallagher
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA.
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA.
| | - Denise M Wolf
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lamorna Brown-Swigart
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nicholas O'Grady
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amrita Basu
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ruixiao Lu
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Michael J Campbell
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark J Magbanua
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jean-Philippe Coppé
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Laura Sit
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey B Matthews
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Nola Hylton
- Department of Radiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Minetta C Liu
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hope S Rugo
- Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Angela M DeMichele
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas Yee
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Paula R Pohlmann
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gillian L Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA.
| |
Collapse
|
16
|
James JL, Taylor BC, Axelrod ML, Sun X, Guerin LN, Gonzalez-Ericsson PI, Wang Y, Sanchez V, Fahey CC, Sanders ME, Xu Y, Hodges E, Johnson DB, Balko JM. Polycomb repressor complex 2 suppresses interferon-responsive MHC-II expression in melanoma cells and is associated with anti-PD-1 resistance. J Immunother Cancer 2023; 11:e007736. [PMID: 38315170 PMCID: PMC10660662 DOI: 10.1136/jitc-2023-007736] [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] [Accepted: 10/17/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Despite the remarkable success of immunotherapy in treating melanoma, understanding of the underlying mechanisms of resistance remains limited. Emerging evidence suggests that upregulation of tumor-specific major histocompatibility complex-II (tsMHC-II) serves as a predictive marker for the response to anti-programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) therapy in various cancer types. The genetic and epigenetic pathways modulating tsMHC-II expression remain incompletely characterized. Here, we provide evidence that polycomb repressive complex 2 (PRC2)/EZH2 signaling and resulting H3K27 hypermethylation suppresses tsMHC-II. METHODS RNA sequencing data from tumor biopsies from patients with cutaneous melanoma treated with or without anti-PD-1, targeted inhibition assays, and assays for transposase-accessible chromatin with sequencing were used to observe the relationship between EZH2 inhibition and interferon (IFN)-γ inducibility within the MHC-II pathway. RESULTS We find that increased EZH2 pathway messenger RNA (mRNA) expression correlates with reduced mRNA expression of both presentation and T-cell genes. Notably, targeted inhibition assays revealed that inhibition of EZH2 influences the expression dynamics and inducibility of the MHC-II pathway following IFN-γ stimulation. Additionally, our analysis of patients with metastatic melanoma revealed a significant inverse association between PRC2-related gene expression and response to anti-PD-1 therapy. CONCLUSIONS Collectively, our findings demonstrate that EZH2 inhibition leads to enhanced MHC-II expression potentially resulting from improved chromatin accessibility at CIITA, the master regulator of MHC-II. These insights shed light on the molecular mechanisms involved in tsMHC-II suppression and highlight the potential of targeting EZH2 as a therapeutic strategy to improve immunotherapy efficacy.
Collapse
Affiliation(s)
- Jamaal L James
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brandie C Taylor
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Margaret L Axelrod
- Department of Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Xiaopeng Sun
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lindsey N Guerin
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Paula I Gonzalez-Ericsson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Violeta Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Catherine C Fahey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Melinda E Sanders
- Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Emily Hodges
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
- Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Zhang S, Li N, Wang F, Liu H, Zhang Y, Xiao J, Qiu W, Zhang C, Fan X, Qiu M, Li M, Tang H, Fan S, Wang J, Luo H, Li X, Lin J, Huang Y, Liang L. Characterization of the tumor microenvironment and identification of spatially predictive biomarkers associated with beneficial neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Pharmacol Res 2023; 197:106974. [PMID: 37898442 DOI: 10.1016/j.phrs.2023.106974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Neoadjuvant chemoradiotherapy (nCRT) has become the standard treatment for patients with locally advanced rectal cancer (LARC). However, 20-40% of patients with LARC show little to no response to nCRT. Thus, comprehensively understanding the tumor microenvironment (TME), which might influence therapeutic efficacy, and identifying robust predictive biomarkers is urgently needed. Pre-treatment tumor biopsy specimens from patients with LARC were evaluated in detail through digital spatial profiling (DSP), public RNA sequencing datasets, and multiplex immunofluorescence (mIF). DSP analysis revealed distinct characteristics of the tumor stroma compared to the normal stroma and tumor compartments. We identified high levels of human leukocyte antigen-DR/major histocompatibility complex class II (HLA-DR/MHC-II) in the tumor compartment and B cells in the stroma as potential spatial predictors of nCRT efficacy in the Discovery cohort. Public datasets validated their predictive capacity for clinical outcomes. Using mIF in an independent nCRT cohort and/or the total cohort, we validated that a high density of HLA-DR/MHC-II+ cells in the tumor and CD20 + B cells in the stroma was associated with nCRT efficacy (all p ≤ 0.021). Spatial profiling successfully characterized the LARC TME and identified robust biomarkers with the potential to accurately predict nCRT response. These findings have important implications for individualized therapy.
Collapse
Affiliation(s)
- Shifen Zhang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China; Department of Pathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Na Li
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China.
| | - Feifei Wang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Hailing Liu
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Yuhan Zhang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Jinyuan Xiao
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Weihao Qiu
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Ceng Zhang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Xinjuan Fan
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Mingxin Qiu
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Mingzhou Li
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Hongzhen Tang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Shiheng Fan
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Jiaqian Wang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Haitao Luo
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen 518000, China
| | - Xiangzhao Li
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China.
| | - Li Liang
- Department of Pathology, Nanfang Hospital/School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
18
|
Goodman RS, Jung S, Balko JM, Johnson DB. Biomarkers of immune checkpoint inhibitor response and toxicity: Challenges and opportunities. Immunol Rev 2023; 318:157-166. [PMID: 37470280 PMCID: PMC10528475 DOI: 10.1111/imr.13249] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023]
Abstract
Immune checkpoint inhibitors have transformed cancer therapy, but their optimal use is still constrained by lack of response and toxicity. Biomarkers of response may facilitate drug development by allowing appropriate therapy selection and focusing clinical trial enrollment. However, aside from PD-L1 staining in a subset of tumors and rarely mismatch repair deficiency, no biomarkers are routinely used in the clinic. In addition, severe toxicities may cause severe morbidity, therapy discontinuation, and even death. Here, we review the state of the field with a focus on our research in therapeutic biomarkers and toxicities from immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Seungyeon Jung
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Justin M. Balko
- Department of Medicine, Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas B. Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
19
|
Wang XQ, Danenberg E, Huang CS, Egle D, Callari M, Bermejo B, Dugo M, Zamagni C, Thill M, Anton A, Zambelli S, Russo S, Ciruelos EM, Greil R, Győrffy B, Semiglazov V, Colleoni M, Kelly CM, Mariani G, Del Mastro L, Biasi O, Seitz RS, Valagussa P, Viale G, Gianni L, Bianchini G, Ali HR. Spatial predictors of immunotherapy response in triple-negative breast cancer. Nature 2023; 621:868-876. [PMID: 37674077 PMCID: PMC10533410 DOI: 10.1038/s41586-023-06498-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/28/2023] [Indexed: 09/08/2023]
Abstract
Immune checkpoint blockade (ICB) benefits some patients with triple-negative breast cancer, but what distinguishes responders from non-responders is unclear1. Because ICB targets cell-cell interactions2, we investigated the impact of multicellular spatial organization on response, and explored how ICB remodels the tumour microenvironment. We show that cell phenotype, activation state and spatial location are intimately linked, influence ICB effect and differ in sensitive versus resistant tumours early on-treatment. We used imaging mass cytometry3 to profile the in situ expression of 43 proteins in tumours from patients in a randomized trial of neoadjuvant ICB, sampled at three timepoints (baseline, n = 243; early on-treatment, n = 207; post-treatment, n = 210). Multivariate modelling showed that the fractions of proliferating CD8+TCF1+T cells and MHCII+ cancer cells were dominant predictors of response, followed by cancer-immune interactions with B cells and granzyme B+ T cells. On-treatment, responsive tumours contained abundant granzyme B+ T cells, whereas resistant tumours were characterized by CD15+ cancer cells. Response was best predicted by combining tissue features before and on-treatment, pointing to a role for early biopsies in guiding adaptive therapy. Our findings show that multicellular spatial organization is a major determinant of ICB effect and suggest that its systematic enumeration in situ could help realize precision immuno-oncology.
Collapse
Affiliation(s)
- Xiao Qian Wang
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Esther Danenberg
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Chiun-Sheng Huang
- National Taiwan University Hospital, College of Medicine, National Taiwan University and Taiwan Breast Cancer Consortium, Taipei, Taiwan
| | - Daniel Egle
- Department of Gynecology, Brust Gesundheit Zentrum Tirol, Medical University Innsbruck, Innsbruck, Austria
| | | | - Begoña Bermejo
- Medical Oncology, Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, Valencia, Spain
- Medicine Department, Universidad de Valencia, Valencia, Spain
- Oncology Biomedical Research National Network (CIBERONC-ISCIII), Madrid, Spain
| | | | - Claudio Zamagni
- IRCCS Azienda Ospedaliero-universitaria di Bologna, Bologna, Italy
| | - Marc Thill
- Department of Gynecology and Gynecological Oncology, Agaplesion Markus Krankenhaus, Frankfurt am Main, Germany
| | - Anton Anton
- Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | - Stefania Russo
- Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | | | - Richard Greil
- 3rd Medical Department, Paracelsus Medical University Salzburg, Salzburg, Austria
- Salzburg Cancer Research Institute-CCCIT, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Cancer Biomarker Research Group, Research Centre for Natural Sciences, Institute of Enzymology, Budapest, Hungary
| | | | | | - Catherine M Kelly
- Mater Private Hospital, Dublin and Cancer Trials Ireland Breast Group, Dublin, Ireland
| | | | - Lucia Del Mastro
- IRCCS Ospedale Policlinico San Martino, UO Clinica di Oncologia Medica, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche (Di.M.I.), Università di Genova, Genoa, Italy
| | - Olivia Biasi
- IEO, Istituto Europeo di Oncologia, IRCCS, Milan, Italy
| | | | | | - Giuseppe Viale
- IEO, Istituto Europeo di Oncologia, IRCCS, Milan, Italy
- University of Milan, Milan, Italy
| | | | | | - H Raza Ali
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Department of Histopathology, Addenbrookes Hospital, Cambridge, UK.
| |
Collapse
|
20
|
Su C, Yang JC, Rong Z, Li F, Luo LX, Liu G, Cheng CY, Zhao MG, Yang L. Identification of CCDC115 as an adverse prognostic biomarker in liver cancer based on bioinformatics and experimental analyses. Heliyon 2023; 9:e19233. [PMID: 37674842 PMCID: PMC10477456 DOI: 10.1016/j.heliyon.2023.e19233] [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: 03/27/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 09/08/2023] Open
Abstract
Liver hepatocellular carcinoma (LIHC) is a major malignant tumor of the digestive system with a high incidence rate and poor early diagnosis. Coiled-coil domain-containing protein 115 (CCDC115), an accessory component of vacuolar-ATPase with dramatically abnormal expression, is associated with survival outcomes of cancer patients. However, the role of CCDC115 in LIHC remains unclear. In this study, we aimed to determine the functional role of CCDC115 in LIHC by examining CCDC115 expression, and its influence on LIHC prognosis. Through extensive statistical analyses, using LIHC patient databases, we observed that CCDC115 expression significantly increased in tumor tissues of LIHC patients. In addition, CCDC115 expression correlated with the poor prognosis. Additionally, CCDC115 was found to be involved in several cancer-related pathways, specifically the PI3K-Akt pathway. The expression of CCDC115 was positively correlated with human leukocyte antigen molecules as well as with immune checkpoint molecules in LIHC patients. We performed in vitro experiments and confirmed that the expression of CCDC115 significantly affects the proliferation potential, metastasis and sorafenib resistance of liver cancer cells, as well as some key protein expression in PI3K-Akt pathway. These results indicate that CCDC115 could serve as a diagnostic and prognostic biomarker of LIHC, and targeting CCDC115 may provide a potential strategy to enhance the efficacy of liver cancer therapy.
Collapse
Affiliation(s)
- Chang Su
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
- Shaanxi Provincial Corps, Chinese People's Armed Police Force, Xi'an, China
| | - Jing-cheng Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Zheng Rong
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Fei Li
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Lan-xin Luo
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Guan Liu
- Department of General Surgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Cai-yan Cheng
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Ming-gao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
21
|
Yang K, Halima A, Chan TA. Antigen presentation in cancer - mechanisms and clinical implications for immunotherapy. Nat Rev Clin Oncol 2023; 20:604-623. [PMID: 37328642 DOI: 10.1038/s41571-023-00789-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/18/2023]
Abstract
Over the past decade, the emergence of effective immunotherapies has revolutionized the clinical management of many types of cancers. However, long-term durable tumour control is only achieved in a fraction of patients who receive these therapies. Understanding the mechanisms underlying clinical response and resistance to treatment is therefore essential to expanding the level of clinical benefit obtained from immunotherapies. In this Review, we describe the molecular mechanisms of antigen processing and presentation in tumours and their clinical consequences. We examine how various aspects of the antigen-presentation machinery (APM) shape tumour immunity. In particular, we discuss genomic variants in HLA alleles and other APM components, highlighting their influence on the immunopeptidomes of both malignant cells and immune cells. Understanding the APM, how it is regulated and how it changes in tumour cells is crucial for determining which patients will respond to immunotherapy and why some patients develop resistance. We focus on recently discovered molecular and genomic alterations that drive the clinical outcomes of patients receiving immune-checkpoint inhibitors. An improved understanding of how these variables mediate tumour-immune interactions is expected to guide the more precise administration of immunotherapies and reveal potentially promising directions for the development of new immunotherapeutic approaches.
Collapse
Affiliation(s)
- Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA.
- National Center for Regenerative Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
22
|
Gandhi S. Novel Biomarkers to Guide Immunotherapy De-Escalation in the Neoadjuvant Setting in Triple-Negative Breast Cancer. J Pers Med 2023; 13:1313. [PMID: 37763081 PMCID: PMC10532861 DOI: 10.3390/jpm13091313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has the highest incidence of disease recurrence and distant metastases among breast cancer subtypes, leading to significant rates of morbidity and mortality [...].
Collapse
Affiliation(s)
- Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
23
|
Boydell E, Sandoval JL, Michielin O, Obeid M, Addeo A, Friedlaender A. Neoadjuvant Immunotherapy: A Promising New Standard of Care. Int J Mol Sci 2023; 24:11849. [PMID: 37511609 PMCID: PMC10380420 DOI: 10.3390/ijms241411849] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Neoadjuvant immunotherapy has emerged as a promising approach in the treatment of various malignancies, with preclinical studies showing improved immune responses in the preoperative setting. FDA-approved neoadjuvant-immunotherapy-based approaches include triple-negative breast cancer and early non-small cell lung cancer on the basis of improvement in pathological response and event free survival. Nevertheless, current trials have only shown benefits in a fraction of patients. It is therefore crucial to identify predictive biomarkers to improve patient selection for such approaches. This review aims to provide an overview of potential biomarkers of neoadjuvant immunotherapy in early triple-negative breast cancer, bladder cancer, melanoma, non-small cell lung cancer, colorectal cancer and gastric cancer. By the extrapolation of the metastatic setting, we explore known predictive biomarkers, i.e., PD-L1, mismatch repair deficiency and tumour mutational burden, as well as potential early-disease-specific biomarkers. We also discuss the challenges of identifying reliable biomarkers and the need for standardized protocols and guidelines for their validation and clinical implementation.
Collapse
Affiliation(s)
- Emma Boydell
- University Hospital of Geneva, 1205 Geneva, Switzerland
| | | | | | - Michel Obeid
- University Hospital of Lausanne, 1005 Lausanne, Switzerland
| | - Alfredo Addeo
- University Hospital of Geneva, 1205 Geneva, Switzerland
| | - Alex Friedlaender
- University Hospital of Geneva, 1205 Geneva, Switzerland
- Clinique Générale Beaulieu, 1206 Geneva, Switzerland
| |
Collapse
|
24
|
Kotteas E, Bielo LB, Valenza C, Santoro C, Koukoutzeli C, Trapani D, Curigliano G. Treatment optimization in early triple negative breast cancer. Expert Rev Anticancer Ther 2023; 23:1107-1116. [PMID: 37873652 DOI: 10.1080/14737140.2023.2268840] [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: 08/10/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION The treatment of early-stage triple-negative breast cancer (TNBC) has radically changed in recent years. Response to neoadjuvant treatment has provided prognostic information, and the achievement of a pathological complete response (pCR) is associated with improved prognosis. An exact treatment algorithm that embraces the trade-off of efficacy and toxicity in a risk-adapted manner has, however, not been consolidated. AREAS COVERED In this review, we focused on the current treatments used for patients with early triple negative breast cancer, aiming at framing a therapeutic approach toward risk-adapted treatment optimization. We reviewed the clinical trials and other evidence at the foundation of the current clinical practice in early TNBC and identified possible areas of clinical implementation. EXPERT OPINION In our opinion, treatment optimization will ensure improved patient-centric outcomes, with less toxicities, better long-term quality of life and risk-adapted treatment modulation. Presently, treatment modulation can be applied in some patients through de-intensification, for small TNBC, informed by novel biomarkers and based on the response to neoadjuvant treatments, especially in the case of pCR. Innovative approaches should incorporate baseline risk and cancer biology, treatment response, and post-surgery biomarkers of prognosis, to deliver risk-adapted treatments for patients with early TNBC.
Collapse
Affiliation(s)
- Elias Kotteas
- Oncology Unit, 3rd Department of Internal Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Celeste Santoro
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Chrysanthi Koukoutzeli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
25
|
Bawden E, Gebhardt T. The multifaceted roles of CD4 + T cells and MHC class II in cancer surveillance. Curr Opin Immunol 2023; 83:102345. [PMID: 37245413 DOI: 10.1016/j.coi.2023.102345] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/29/2023] [Accepted: 05/01/2023] [Indexed: 05/30/2023]
Abstract
CD4+ T cells exhibit diverse functions in cancer surveillance. Concordantly, single-cell transcriptional analyses have revealed several distinct CD4+ T-cell differentiation states in tumours, including cytotoxic and regulatory subsets associated with favourable or unfavourable outcomes, respectively. These transcriptional states are determined and further shaped by dynamic interactions of CD4+ T cells with different types of immune cells, stromal cells and cancer cells. Therefore, we discuss the cellular networks in the tumour microenvironment (TME) that either promote or impede CD4+ T-cell cancer surveillance. We consider antigen/Major histocompatibility complexclass-II (MHC-II)-dependent interactions of CD4+ T cells with both professional antigen-presenting cells and cancer cells, the latter of which can directly express MHC-II, at least in some tumours. Additionally, we examine recent single-cell RNA sequencing studies that have shed light on the phenotype and functions of cancer-specific CD4+ T cells in human tumours.
Collapse
Affiliation(s)
- Emma Bawden
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Thomas Gebhardt
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
26
|
Vaz SC, Graff SL, Ferreira AR, Debiasi M, de Geus-Oei LF. PET/CT in Patients with Breast Cancer Treated with Immunotherapy. Cancers (Basel) 2023; 15:cancers15092620. [PMID: 37174086 PMCID: PMC10177398 DOI: 10.3390/cancers15092620] [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: 03/29/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Significant advances in breast cancer (BC) treatment have been made in the last decade, including the use of immunotherapy and, in particular, immune checkpoint inhibitors that have been shown to improve the survival of patients with triple negative BC. This narrative review summarizes the studies supporting the use of immunotherapy in BC. Furthermore, the usefulness of 2-deoxy-2-[18F]fluoro-D-glucose (2-[18F]FDG) positron emission/computerized tomography (PET/CT) to image the tumor heterogeneity and to assess treatment response is explored, including the different criteria to interpret 2-[18F]FDG PET/CT imaging. The concept of immuno-PET is also described, by explaining the advantages of mapping treatment targets with a non-invasive and whole-body tool. Several radiopharmaceuticals in the preclinical phase are referred too, and, considering their promising results, translation to human studies is needed to support their use in clinical practice. Overall, this is an evolving field in BC treatment, despite PET imaging developments, the future trends also include expanding immunotherapy to early-stage BC and using other biomarkers.
Collapse
Affiliation(s)
- Sofia C Vaz
- Nuclear Medicine-Radiopharmacology, Champalimaud Center for the Unkown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Department of Radiology, Leiden University Medical Center, P.O. Box 9600-2300 RC Leiden, The Netherlands
| | - Stephanie L Graff
- Division of Hematology/Oncology, Lifespan Cancer Institute, Providence, RI 02903, USA
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Arlindo R Ferreira
- Católica Medical School, Universidade Católica Portuguesa, 2635-631 Lisbon, Portugal
| | - Márcio Debiasi
- Breast Cancer Unit, Champalimaud Center for the Unkown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, P.O. Box 9600-2300 RC Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, P.O. Box 217-7500 AE Enschede, The Netherlands
- Department of radiation Science & Technology, Delft University of Technology, P.O. Postbus 5 2600 AA Delft, The Netherlands
| |
Collapse
|
27
|
Licata L, Mariani M, Rossari F, Viale G, Notini G, Naldini MM, Bosi C, Piras M, Dugo M, Bianchini G. Tissue- and liquid biopsy-based biomarkers for immunotherapy in breast cancer. Breast 2023; 69:330-341. [PMID: 37003065 PMCID: PMC10070181 DOI: 10.1016/j.breast.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy and now represent the mainstay of treatment for many tumor types, including triple-negative breast cancer and two agnostic registrations. However, despite impressive durable responses suggestive of an even curative potential in some cases, most patients receiving ICIs do not derive a substantial benefit, highlighting the need for more precise patient selection and stratification. The identification of predictive biomarkers of response to ICIs may play a pivotal role in optimizing the therapeutic use of such compounds. In this Review, we describe the current landscape of tissue and blood biomarkers that could serve as predictive factors for ICI treatment in breast cancer. The integration of these biomarkers in a "holistic" perspective aimed at developing comprehensive panels of multiple predictive factors will be a major step forward towards precision immune-oncology.
Collapse
Affiliation(s)
- Luca Licata
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Mariani
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Federico Rossari
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy; San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Viale
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Notini
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Maria Naldini
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy; San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Bosi
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Piras
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
28
|
Abu-Khalaf MM, Alex Hodge K, Hatzis C, Baldelli E, El Gazzah E, Valdes F, Sikov WM, Mita MM, Denduluri N, Murphy R, Zelterman D, Liotta L, Dunetz B, Dunetz R, Petricoin EF, Pierobon M. AKT/mTOR signaling modulates resistance to endocrine therapy and CDK4/6 inhibition in metastatic breast cancers. NPJ Precis Oncol 2023; 7:18. [PMID: 36797347 PMCID: PMC9935518 DOI: 10.1038/s41698-023-00360-5] [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: 11/07/2022] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Endocrine therapy (ET) in combination with CDK4/6 inhibition is routinely used as first-line treatment for HR+/HER2- metastatic breast cancer (MBC) patients. However, 30-40% of patients quickly develop disease progression. In this open-label multicenter clinical trial, we utilized a hypothesis-driven protein/phosphoprotein-based approach to identify predictive markers of response to ET plus CDK4/6 inhibition in pre-treatment tissue biopsies. Pathway-centered signaling profiles were generated from microdissected tumor epithelia and surrounding stroma/immune cells using the reverse phase protein microarray. Phosphorylation levels of the CDK4/6 downstream substrates Rb (S780) and FoxM1 (T600) were higher in patients with progressive disease (PD) compared to responders (p = 0.02). Systemic PI3K/AKT/mTOR activation in tumor epithelia and stroma/immune cells was detected in patients with PD. This activation was not explained by underpinning genomic alterations alone. As the number of FDA-approved targeted compounds increases, functional protein-based signaling analyses may become a critical component of response prediction and treatment selection for MBC patients.
Collapse
Affiliation(s)
- Maysa M. Abu-Khalaf
- grid.415231.00000 0004 0577 7855Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, PA USA
| | - K. Alex Hodge
- grid.22448.380000 0004 1936 8032School of Systems Biology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA USA
| | | | - Elisa Baldelli
- grid.22448.380000 0004 1936 8032School of Systems Biology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA USA
| | - Emna El Gazzah
- grid.22448.380000 0004 1936 8032School of Systems Biology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA USA
| | - Frances Valdes
- grid.419791.30000 0000 9902 6374Sylvester Comprehensive Cancer Center (UM SCCC), University of Miami, Miami, FL USA
| | - William M. Sikov
- grid.241223.4Women and Infants Hospital of Rhode Island, Providence, RI USA
| | - Monica M. Mita
- grid.50956.3f0000 0001 2152 9905Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Neelima Denduluri
- grid.492966.60000 0004 0481 8256Virginia Cancer Specialists, Fairfax, VA USA
| | - Rita Murphy
- grid.415231.00000 0004 0577 7855Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, PA USA
| | | | - Lance Liotta
- grid.22448.380000 0004 1936 8032School of Systems Biology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA USA
| | | | - Rick Dunetz
- grid.490989.5Side Out Foundation, Fairfax, VA USA
| | - Emanuel F. Petricoin
- grid.22448.380000 0004 1936 8032School of Systems Biology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA USA
| | - Mariaelena Pierobon
- School of Systems Biology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, VA, USA.
| |
Collapse
|
29
|
Korotaeva AA, Borunova AA, Kuzevanova AY, Zabotina TN, Alimov AA. [Molecular mechanisms of impaired antigenic presentation as a cause of tumor escape from immune surveillance]. Arkh Patol 2023; 85:76-83. [PMID: 38010642 DOI: 10.17116/patol20238506176] [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] [Indexed: 11/29/2023]
Abstract
The review summarizes data on the features of antigen presentation in tumor cells. The molecular mechanisms of the antitumor immune response are considered with an emphasis on the ability of tumor cells to avoid the action of immune surveillance. The features of expression of MHC molecules depending on treatment regimens are provided. Ways to improve existing and create new treatment regimens aimed at elimination of tumor cells because of antitumor immune response are discussed.
Collapse
Affiliation(s)
- A A Korotaeva
- Research Centre for Medical Genetics, Moscow, Russia
| | - A A Borunova
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | | | - T N Zabotina
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - A A Alimov
- Research Centre for Medical Genetics, Moscow, Russia
| |
Collapse
|
30
|
Mechanisms and Strategies to Overcome PD-1/PD-L1 Blockade Resistance in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 15:cancers15010104. [PMID: 36612100 PMCID: PMC9817764 DOI: 10.3390/cancers15010104] [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: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a high rate of systemic metastasis, insensitivity to conventional treatment and susceptibility to drug resistance, resulting in a poor patient prognosis. The immune checkpoint inhibitors (ICIs) represented by antibodies of programmed death receptor 1 (PD-1) and programmed death receptor ligand 1 (PD-L1) have provided new therapeutic options for TNBC. However, the efficacy of PD-1/PD-L1 blockade monotherapy is suboptimal immune response, which may be caused by reduced antigen presentation, immunosuppressive tumor microenvironment, interplay with other immune checkpoints and aberrant activation of oncological signaling in tumor cells. Therefore, to improve the sensitivity of TNBC to ICIs, suitable patients are selected based on reliable predictive markers and treated with a combination of ICIs with other therapies such as chemotherapy, radiotherapy, targeted therapy, oncologic virus and neoantigen-based therapies. This review discusses the current mechanisms underlying the resistance of TNBC to PD-1/PD-L1 inhibitors, the potential biomarkers for predicting the efficacy of anti-PD-1/PD-L1 immunotherapy and recent advances in the combination therapies to increase response rates, the depth of remission and the durability of the benefit of TNBC to ICIs.
Collapse
|
31
|
Zhang X, Liu X, Zhu K, Zhang X, Li N, Sun T, Fan S, Dai L, Zhang J. CD5L-associated gene analyses highlight the dysregulations, prognostic effects, immune associations, and drug-sensitivity predicative potentials of LCAT and CDC20 in hepatocellular carcinoma. Cancer Cell Int 2022; 22:393. [PMID: 36494696 PMCID: PMC9733014 DOI: 10.1186/s12935-022-02820-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The dysregulation of CD5L has been reported in hepatocellular carcinoma (HCC). However, its functions in HCC were controversial. In this study, we aimed to identify CD5L-associated pathways and markers and explore their values in HCC diagnosis, prognosis and treatment. METHODS HCC datasets with gene expression profiles and clinical data in TCGA and ICGC were downloaded. The immune/stroma cell infiltrations were estimated with xCell. CD5L-associated pathways and CD5L-associated genes (CD5L-AGs) were identified with gene expression comparisons and gene set enrichment analysis (GSEA). Cox regression, Kaplan-Meier survival analysis, and least absolute shrinkage and selection operator (LASSO) regression analysis were performed. The correlations of the key genes with immune/stroma infiltrations, immunoregulators, and anti-cancer drug sensitivities in HCC were investigated. At protein level, the key genes dysregulations, their correlations and prognostic values were validated in clinical proteomic tumor analysis consortium (CPTAC) database. Serum CD5L and LCAT activity in 50 HCC and 30 normal samples were evaluated and compared. The correlations of serum LCAT activity with alpha-fetoprotein (AFP), albumin (ALB) and high-density lipoprotein (HDL) in HCC were also investigated. RESULTS Through systemic analyses, 14 CD5L-associated biological pathways, 256 CD5L-AGs and 28 CD5L-associated prognostic and diagnostic genes (CD5L-APDGs) were identified. A risk model consisting of LCAT and CDC20 was constructed for HCC overall survival (OS), which could discriminate HCC OS status effectively in both the training and the validation sets. CD5L, LCAT and CDC20 were shown to be significantly correlated with immune/stroma cell infiltrations, immunoregulators and 31 anti-cancer drug sensitivities in HCC. At protein level, the dysregulations of CD5L, LCAT and CDC20 were confirmed. LCAT and CDC20 were shown to be significantly correlated with proliferation marker MKI67. In serum, no significance of CD5L was shown. However, the lower activity of LCAT in HCC serum was obvious, as well as its significant positive correlations ALB and HDL concentrations. CONCLUSIONS CD5L, LCAT and CDC20 were dysregulated in HCC both at mRNA and protein levels. The LCAT-CDC20 signature might be new predicator for HCC OS. The associations of the three genes with HCC microenvironment and anti-cancer drug sensitivities would provide new clues for HCC immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Xiaoli Liu
- grid.414011.10000 0004 1808 090XLaboratory Department, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Keke Zhu
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Xue Zhang
- grid.207374.50000 0001 2189 3846Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ningning Li
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Tao Sun
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Shasha Fan
- grid.477407.70000 0004 1806 9292Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People’s Hospital, Changsha, China ,grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, China
| | - Liping Dai
- grid.207374.50000 0001 2189 3846Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinzhong Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| |
Collapse
|
32
|
Farheen J, Hosmane NS, Zhao R, Zhao Q, Iqbal MZ, Kong X. Nanomaterial-assisted CRISPR gene-engineering - A hallmark for triple-negative breast cancer therapeutics advancement. Mater Today Bio 2022; 16:100450. [PMID: 36267139 PMCID: PMC9576993 DOI: 10.1016/j.mtbio.2022.100450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/16/2022] [Accepted: 10/02/2022] [Indexed: 11/05/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most violent class of tumor and accounts for 20-24% of total breast carcinoma, in which frequently rare mutation occurs in high frequency. The poor prognosis, recurrence, and metastasis in the brain, heart, liver and lungs decline the lifespan of patients by about 21 months, emphasizing the need for advanced treatment. Recently, the adaptive immunity mechanism of archaea and bacteria, called clustered regularly interspaced short palindromic repeats (CRISPR) combined with nanotechnology, has been utilized as a potent gene manipulating tool with an extensive clinical application in cancer genomics due to its easeful usage and cost-effectiveness. However, CRISPR/Cas are arguably the efficient technology that can be made efficient via organic material-assisted approaches. Despite the efficacy of the CRISPR/Cas@nano complex, problems regarding successful delivery, biodegradability, and toxicity remain to render its medical implications. Therefore, this review is different in focus from past reviews by (i) detailing all possible genetic mechanisms of TNBC occurrence; (ii) available treatments and gene therapies for TNBC; (iii) overview of the delivery system and utilization of CRISPR-nano complex in TNBC, and (iv) recent advances and related toxicity of CRISPR-nano complex towards clinical trials for TNBC.
Collapse
Affiliation(s)
- Jabeen Farheen
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Narayan S. Hosmane
- Department of Chemistry & Biochemistry, Northern Illinois University, DeKalb, IL, 60115, USA
| | - Ruibo Zhao
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Department of Materials, Imperial College London, London, SW7 2AZ, UK
| | - Qingwei Zhao
- Research Center for Clinical Pharmacy & Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - M. Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
- Zhejiang-Mauritius Joint Research Centre for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| |
Collapse
|
33
|
Wang Z, Zheng W, Chen Z, Wu S, Chang H, Cai M, Cai H. Pan-Cancer analysis shows that ACO2 is a potential prognostic and immunotherapeutic biomarker for multiple cancer types including hepatocellular carcinoma. Front Oncol 2022; 12:1055376. [PMID: 36531056 PMCID: PMC9748622 DOI: 10.3389/fonc.2022.1055376] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/10/2022] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND Recent evidence increasingly suggests key roles for the tricarboxylic acid cycle and fatty acid metabolism in tumor progression and metastasis. Aconitase 2 (ACO2) is a component of the tricarboxylic acid cycle and represents a key cellular metabolic hub that promotes de novo fatty acid biosynthesis. However, there have been few reports on the role of ACO2 in tumorigenesis and cancer progression. METHODS Through the comprehensive use of datasets from The Cancer Genome Atlas, Genotype-Tissue Expression Project, cBioPortal, Human Protein Atlas, UALCAN, Gene Expression Profiling Interactive Analysis, DNA Methylation Interactive Visualization Database, and TIMER2, we adopted bioinformatics methods to uncover the potential carcinogenic roles of ACO2, including by analysing ACO2 expression and correlations between prognosis, genetic mutations, immune cell infiltration, DNA methylation, tumor mutational burden, and microsatellite instability in different tumors. Additionally, the expression level and tumor-promoting effect of ACO2 were verified in hepatocellular carcinoma (HCC) cells. To explore the underlying mechanisms of ACO2 in human cancer, ACO2-related gene enrichment analysis and lipid metabolomics were performed using LM3 cells with or without ACO2 knockdown. RESULTS The results indicated that ACO2 was highly expressed in most cancers, showing early diagnostic value in six tumor types, and was positively or negatively associated with prognosis in different tumors. Moreover, ACO2 expression was associated with immune cell infiltration, such as CD8+ T cells and tumor-associated neutrophils, in some cancers. For most cancer types, there was a significant association between immune checkpoint-associated genes and ACO2 expression. Compared with normal hepatocytes, ACO2 was upregulated in HCC cells, which promoted their proliferation and migration. Furthermore, to explore the underlying molecular mechanism, we performed KEGG pathway enrichment analysis of ACO2-associated genes and lipidomics using LM3 cells with or without ACO2 knockdown, which screened 19 significantly altered metabolites, including 17 with reduced levels and 2 with increased levels. CONCLUSION Through pan-cancer analysis, we discovered for the first time and verified that ACO2 could be a useful diagnostic biomarker for cancer detection. Additionally, ACO2 could be used as an auxiliary prognostic marker or as a marker for immunotherapy in some tumor types.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Pharmacy, Anhui Provincial Children’s Hospital, Hefei, China
| | - Wanqun Zheng
- Department of Chinese Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhen Chen
- Department of Pharmacy, The Third People’s Hospital of Hefei, Hefei, China
| | - Shilun Wu
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Haoxiao Chang
- China National Clinical Research Center for Neurological Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Anhui Acupuncture and Moxibustion Clinical Medicine Research Center, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Heping Cai
- Department of Pharmacy, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
34
|
Sharma P. Time to Optimize Deescalation Strategies in Triple-Negative Breast Cancer? Clin Cancer Res 2022; 28:4840-4842. [PMID: 36083131 DOI: 10.1158/1078-0432.ccr-22-2140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/16/2022] [Accepted: 08/31/2022] [Indexed: 01/24/2023]
Abstract
Pathologic response serves as a tool to optimize escalation and deescalation of adjuvant treatment. Neoadjuvant carboplatin-taxane combination is a promising chemotherapy deescalation strategy for triple-negative breast cancer. Many key points, such as trial design/patient selection, response biomarkers, role of immunotherapy, and patient advocate input, will have to be carefully considered in order to advance neoadjuvant chemotherapy deescalation investigations. See related article by Gluz et al., p. 4995.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| |
Collapse
|
35
|
Massa D, Tosi A, Rosato A, Guarneri V, Dieci MV. Multiplexed In Situ Spatial Protein Profiling in the Pursuit of Precision Immuno-Oncology for Patients with Breast Cancer. Cancers (Basel) 2022; 14:4885. [PMID: 36230808 PMCID: PMC9562913 DOI: 10.3390/cancers14194885] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many solid tumors. In breast cancer (BC), immunotherapy is currently approved in combination with chemotherapy, albeit only in triple-negative breast cancer. Unfortunately, most patients only derive limited benefit from ICIs, progressing either upfront or after an initial response. Therapeutics must engage with a heterogeneous network of complex stromal-cancer interactions that can fail at imposing cancer immune control in multiple domains, such as in the genomic, epigenomic, transcriptomic, proteomic, and metabolomic domains. To overcome these types of heterogeneous resistance phenotypes, several combinatorial strategies are underway. Still, they can be predicted to be effective only in the subgroups of patients in which those specific resistance mechanisms are effectively in place. As single biomarker predictive performances are necessarily suboptimal at capturing the complexity of this articulate network, precision immune-oncology calls for multi-omics tumor microenvironment profiling in order to identify unique predictive patterns and to proactively tailor combinatorial treatments. Multiplexed single-cell spatially resolved tissue analysis, through precise epitope colocalization, allows one to infer cellular functional states in view of their spatial organization. In this review, we discuss-through the lens of the cancer-immunity cycle-selected, established, and emerging markers that may be evaluated in multiplexed spatial protein panels to help identify prognostic and predictive patterns in BC.
Collapse
Affiliation(s)
- Davide Massa
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| |
Collapse
|
36
|
So JY, Ohm J, Lipkowitz S, Yang L. Triple negative breast cancer (TNBC): Non-genetic tumor heterogeneity and immune microenvironment: Emerging treatment options. Pharmacol Ther 2022; 237:108253. [PMID: 35872332 PMCID: PMC9378710 DOI: 10.1016/j.pharmthera.2022.108253] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/01/2022] [Accepted: 07/18/2022] [Indexed: 12/17/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by extensive intra-tumoral heterogeneity, and frequently develops resistance to therapies. Tumor heterogeneity and lack of biomarkers are thought to be some of the most difficult challenges driving therapeutic resistance and relapse. This review will summarize current therapy for TNBC, studies in treatment resistance and relapse, including data from recent single cell sequencing. We will discuss changes in both the transcriptome and epigenome of TNBC, and we will review mechanisms regulating the immune microenvironment. Lastly, we will provide new perspective in patient stratification, and treatment options targeting transcriptome dysregulation and the immune microenvironment of TNBC patients.
Collapse
Affiliation(s)
- Jae Young So
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joyce Ohm
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Stan Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Yang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
37
|
Liang Y, Xu Q, Liu S, Li J, Wang F, Li Z, Liao L, Lu Y, Li Y, Mu F, Sun HX, Zhu L. Single-Cell Transcriptomics Reveals Killing Mechanisms of Antitumor Cytotoxic CD4+ TCR-T Cells. Front Immunol 2022; 13:939940. [PMID: 35928827 PMCID: PMC9343810 DOI: 10.3389/fimmu.2022.939940] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/21/2022] [Indexed: 11/19/2022] Open
Abstract
T cell receptor-engineered T cells (TCR-Ts) have emerged as potent cancer immunotherapies. While most research focused on classical cytotoxic CD8+ T cells, the application of CD4+ T cells in adoptive T cell therapy has gained much interest recently. However, the cytotoxic mechanisms of CD4+ TCR-Ts have not been fully revealed. In this study, we obtained an MHC class I-restricted MART-127-35-specific TCR sequence based on the single-cell V(D)J sequencing technology, and constructed MART-127-35-specific CD4+ TCR-Ts and CD8+ TCR-Ts. The antitumor effects of CD4+ TCR-Ts were comparable to those of CD8+ TCR-Ts in vitro and in vivo. To delineate the killing mechanisms of cytotoxic CD4+ TCR-Ts, we performed single-cell RNA sequencing and found that classical granule-dependent and independent cytolytic pathways were commonly used in CD4+ and CD8+ TCR-Ts, while high expression of LTA and various costimulatory receptors were unique features for cytotoxic CD4+ TCR-Ts. Further signaling pathway analysis revealed that transcription factors Runx3 and Blimp1/Tbx21 were crucial for the development and killing function of cytotoxic CD4+ T cells. Taken together, we report the antitumor effects and multifaceted killing mechanisms of CD4+ TCR-Ts, and also indicate that MHC class I-restricted CD4+ TCR-Ts could serve as potential adoptive T cell therapies.
Collapse
Affiliation(s)
- Yanling Liang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Qumiao Xu
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
- *Correspondence: Qumiao Xu, ; Feng Mu, ; Hai-Xi Sun, ; Linnan Zhu,
| | - Songming Liu
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Jie Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Fei Wang
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Ziyi Li
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Lijuan Liao
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Yuting Lu
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Yijian Li
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Feng Mu
- Beijing Genomics Institute (BGI), Shenzhen, China
- *Correspondence: Qumiao Xu, ; Feng Mu, ; Hai-Xi Sun, ; Linnan Zhu,
| | - Hai-Xi Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
- Beijing Genomics Institute (BGI)-Beijing, Beijing, China
- *Correspondence: Qumiao Xu, ; Feng Mu, ; Hai-Xi Sun, ; Linnan Zhu,
| | - Linnan Zhu
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
- *Correspondence: Qumiao Xu, ; Feng Mu, ; Hai-Xi Sun, ; Linnan Zhu,
| |
Collapse
|
38
|
Vathiotis IA, Trontzas I, Gavrielatou N, Gomatou G, Syrigos NK, Kotteas EA. Immune Checkpoint Blockade in Hormone Receptor-Positive Breast Cancer: Resistance Mechanisms and Future Perspectives. Clin Breast Cancer 2022; 22:642-649. [PMID: 35906130 DOI: 10.1016/j.clbc.2022.06.004] [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: 11/17/2021] [Revised: 05/21/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Anti-programmed cell death protein 1 immunotherapy has been incorporated in the treatment algorithm of triple-negative breast cancer (TNBC). However, clinical trial results for patients with hormone receptor (HR)-positive disease appear less compelling. HR-positive tumors exhibit lower levels of programmed death-ligand 1 expression in comparison with their triple-negative counterparts. Moreover, signaling through estrogen receptor alters the immune microenvironment, rendering such tumors immunologically "cold." To explain differential responses to immune checkpoint blockade, this review interrogates differences between HR-positive and TNBC. Starting from distinct genomic features, we further present disparities concerning the tumor microenvironment and finally, we summarize early-phase clinical trial results on promising novel immunotherapy combinations.
Collapse
Affiliation(s)
- Ioannis A Vathiotis
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece; Department of Pathology, Yale University School of Medicine, New Haven, CT.
| | - Ioannis Trontzas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Niki Gavrielatou
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Georgia Gomatou
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Nikolaos K Syrigos
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Elias A Kotteas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| |
Collapse
|
39
|
Fracasso PM, Goodner SA, Wildi JD, Naughton MJ, Linette GP, Govindan R, Tan BR, Blum KA, Jones GJ, Pearce TE, Levitt DJ, Clamon GH. A Phase I Study of Apolizumab, an Anti-HLA-DR ß-chain Monoclonal Antibody, in Patients With Solid Tumor Malignancies. Am J Clin Oncol 2022; 45:294-297. [PMID: 35700081 PMCID: PMC9219582 DOI: 10.1097/coc.0000000000000924] [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] [Indexed: 11/25/2022]
Abstract
BACKGROUND Human leukocyte antigen (HLA)-DR, a member of the major histocompatibility complex class II antigen family, is a target for antibody-based therapeutics. Apolizumab (Hu1D10, Remitogen), a humanized IgG1 monoclonal anti-HLA-DR ß-chain antibody targets the antigen, 1D10, expressed on a wide variety of hematologic and solid tumor malignancies. In this Phase 1 trial, the maximum tolerated dose and dose-limiting toxicity of weekly apolizumab in patients with advanced solid tumor malignancies were determined. PATIENTS AND METHODS Eligible patients with refractory solid tumors were initially screened for ID10 Ag on their tumor. Patients whose tumors expressed 1D10 were administered apolizumab 0.5, 1.0, 1.5, or 3.0 mg/kg intravenously over 90 minutes weekly for 4 consecutive weeks, followed by a 4-week break, and assessment of response. Patients whose disease had not progressed were offered additional treatment. RESULTS Tumors from 75 patients were screened for 1D10 Ag of which 17 patients were positive and underwent treatment. The first 3 dose levels were well-tolerated. Dose-limiting toxicities of grade 3 infusion-related hypersensitivity reactions and grade 3 headache and hypertension occurred in 2 patients, respectively, at apolizumab 3.0 mg/kg. Four patients, 1 each with breast carcinoma, melanoma, renal cell carcinoma, and sarcoma had stable disease for a median of 15 weeks (range: 12 to 19 wk). CONCLUSION Apolizumab can be administered safely at a maximum tolerated dose of 1.5 mg/kg for 4 consecutive weeks. Adverse events and limited clinical data in both hematologic and solid tumor malignancies resulted in discontinuation of clinical development of apolizumab. HLA-DR remains an interesting immunotherapeutic target.
Collapse
Affiliation(s)
- Paula M. Fracasso
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Sherry A. Goodner
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Jonathan D. Wildi
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J. Naughton
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Gerald P. Linette
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Ramaswamy Govindan
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Benjamin R. Tan
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Kristie A. Blum
- Department of Medicine, Washington University School of Medicine and the Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110
| | - Gary J. Jones
- PDL BioPharma, Inc., 34801 Campus Drive, Freemont, CA 94555
| | | | | | - Gerald H. Clamon
- Department of Internal Medicine, University of Iowa Hospital and Clinics, Iowa City, IA 52242
| |
Collapse
|
40
|
Giugliano F, Valenza C, Tarantino P, Curigliano G. Immunotherapy for triple negative breast cancer: How can pathologic responses to experimental drugs in early-stage disease be enhanced? Expert Opin Investig Drugs 2022; 31:855-874. [PMID: 35762248 DOI: 10.1080/13543784.2022.2095260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : The treatment landscape of early triple negative breast cancer (TNBC) has recently expanded after the Food and Drug Administration (FDA) approval of pembrolizumab in combination with neoadjuvant chemotherapy. The addition of this immune checkpoint inhibitor (ICI) has shown to significantly increased pathological complete response (pCR) rate and event free survival (EFS) in the KEYNOTE-522 phase 3 trial. Several additional studies are ongoing with the goal of further improving outcomes and achieving an optimal integration of ICIs in the treatment of TNBC. AREAS COVERED : The article examines pCR and survival rates in TNBC. It appraises clinical trials investigating neoadjuvant ICIs for TNBC and the improvement of pCR rates (biomarker-driven escalation of treatment, optimization of chemotherapy backbone and addition of locoregional treatments or innovative agents). Insights on the role of pCR as surrogate endpoint and the possibility of enhancing pCR rates for women affected by early TNBC are offered. EXPERT OPINION : The pharmacopoeia of early TNBC is growing and becoming more heterogeneous with the advent of ICIs; to enhance the clinical benefit of patients, it is necessary to develop response endpoints that consider the mechanism of action of experimental drugs, to optimize patient selection through validated biomarkers, and to compare the most promising treatment strategies in randomized clinical trials.
Collapse
Affiliation(s)
- Federica Giugliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| | - Paolo Tarantino
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy.,Breast Oncology Center, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| |
Collapse
|
41
|
Wolf DM, Yau C, Wulfkuhle J, Brown-Swigart L, Gallagher RI, Lee PRE, Zhu Z, Magbanua MJ, Sayaman R, O'Grady N, Basu A, Delson A, Coppé JP, Lu R, Braun J, Asare SM, Sit L, Matthews JB, Perlmutter J, Hylton N, Liu MC, Pohlmann P, Symmans WF, Rugo HS, Isaacs C, DeMichele AM, Yee D, Berry DA, Pusztai L, Petricoin EF, Hirst GL, Esserman LJ, van 't Veer LJ. Redefining breast cancer subtypes to guide treatment prioritization and maximize response: Predictive biomarkers across 10 cancer therapies. Cancer Cell 2022; 40:609-623.e6. [PMID: 35623341 PMCID: PMC9426306 DOI: 10.1016/j.ccell.2022.05.005] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/16/2022] [Accepted: 05/06/2022] [Indexed: 12/26/2022]
Abstract
Using pre-treatment gene expression, protein/phosphoprotein, and clinical data from the I-SPY2 neoadjuvant platform trial (NCT01042379), we create alternative breast cancer subtypes incorporating tumor biology beyond clinical hormone receptor (HR) and human epidermal growth factor receptor-2 (HER2) status to better predict drug responses. We assess the predictive performance of mechanism-of-action biomarkers from ∼990 patients treated with 10 regimens targeting diverse biology. We explore >11 subtyping schemas and identify treatment-subtype pairs maximizing the pathologic complete response (pCR) rate over the population. The best performing schemas incorporate Immune, DNA repair, and HER2/Luminal phenotypes. Subsequent treatment allocation increases the overall pCR rate to 63% from 51% using HR/HER2-based treatment selection. pCR gains from reclassification and improved patient selection are highest in HR+ subsets (>15%). As new treatments are introduced, the subtyping schema determines the minimum response needed to show efficacy. This data platform provides an unprecedented resource and supports the usage of response-based subtypes to guide future treatment prioritization.
Collapse
Affiliation(s)
- Denise M Wolf
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA.
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Lamorna Brown-Swigart
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Rosa I Gallagher
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Pei Rong Evelyn Lee
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Zelos Zhu
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark J Magbanua
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Rosalyn Sayaman
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Nicholas O'Grady
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amrita Basu
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amy Delson
- Breast Science Advocacy Core, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jean Philippe Coppé
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Ruixiao Lu
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Jerome Braun
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Laura Sit
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey B Matthews
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Nola Hylton
- Department of Radiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Minetta C Liu
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Paula Pohlmann
- MedStar Georgetown University Hospital, Georgetown University, Washington, DC 20057, USA
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hope S Rugo
- Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Angela M DeMichele
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas Yee
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Lajos Pusztai
- Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Gillian L Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA.
| |
Collapse
|
42
|
Blenman KRM, Marczyk M, Karn T, Qing T, Li X, Gunasekharan V, Yaghoobi V, Bai Y, Ibrahim EY, Park T, Silber A, Wolf DM, Reisenbichler E, Denkert C, Sinn BV, Rozenblit M, Foldi J, Rimm DL, Loibl S, Pusztai L. Predictive Markers of Response to Neoadjuvant Durvalumab with Nab-Paclitaxel and Dose-Dense Doxorubicin/Cyclophosphamide in Basal-Like Triple-Negative Breast Cancer. Clin Cancer Res 2022; 28:2587-2597. [PMID: 35377948 PMCID: PMC9464605 DOI: 10.1158/1078-0432.ccr-21-3215] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/04/2022] [Accepted: 04/01/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE We examined gene expression, germline variant, and somatic mutation features associated with pathologic response to neoadjuvant durvalumab plus chemotherapy in basal-like triple-negative breast cancer (bTNBC). EXPERIMENTAL DESIGN Germline and somatic whole-exome DNA and RNA sequencing, programmed death ligand 1 (PD-L1) IHC, and stromal tumor-infiltrating lymphocyte scoring were performed on 57 patients. We validated our results using 162 patients from the GeparNuevo randomized trial. RESULTS Gene set enrichment analysis showed that pathways involved in immunity (adaptive, humoral, innate), JAK-STAT signaling, cancer drivers, cell cycle, apoptosis, and DNA repair were enriched in cases with pathologic complete response (pCR), whereas epithelial-mesenchymal transition, extracellular matrix, and TGFβ pathways were enriched in cases with residual disease (RD). Immune-rich bTNBC with RD was enriched in CCL-3, -4, -5, -8, -23, CXCL-1, -3, -6, -10, and IL1, -23, -27, -34, and had higher expression of macrophage markers compared with immune-rich cancers with pCR that were enriched in IFNγ, IL2, -12, -21, chemokines CXCL-9, -13, CXCR5, and activated T- and B-cell markers (GZMB, CD79A). In the validation cohort, an immune-rich five-gene signature showed higher expression in pCR cases in the durvalumab arm (P = 0.040) but not in the placebo arm (P = 0.923) or in immune-poor cancers. Independent of immune markers, tumor mutation burden was higher, and PI3K, DNA damage repair, MAPK, and WNT/β-catenin signaling pathways were enriched in germline and somatic mutations in cases with pCR. CONCLUSIONS The TGFβ pathway is associated with immune-poor phenotype and RD in bTNBC. Among immune-rich bTNBC RD, macrophage/neutrophil chemoattractants dominate the cytokine milieu, and IFNγ and activated B cells and T cells dominate immune-rich cancers with pCR.
Collapse
Affiliation(s)
- Kim RM Blenman
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Michal Marczyk
- Yale Cancer Center, Yale University, New Haven, CT, USA
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | | | - Tao Qing
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Xiaotong Li
- Department of Computational Biology & Bioinformatics, Biological & Biomedical Sciences, Yale University, New Haven, CT, USA
| | - Vignesh Gunasekharan
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Vesal Yaghoobi
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Yalai Bai
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Eiman Y Ibrahim
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Tristen Park
- Department of Surgery, Yale University, New Haven, CT, USA
| | - Andrea Silber
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Denise M. Wolf
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | | | | | | | - Mariya Rozenblit
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Julia Foldi
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - David L Rimm
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | | | - Lajos Pusztai
- Department of Internal Medicine, Section of Medical Oncology, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
43
|
Cathcart AM, Smith H, Labrie M, Mills GB. Characterization of anticancer drug resistance by reverse-phase protein array: new targets and strategies. Expert Rev Proteomics 2022; 19:115-129. [PMID: 35466854 PMCID: PMC9215307 DOI: 10.1080/14789450.2022.2070065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Drug resistance is the main barrier to achieving cancer cures with medical therapy. Cancer drug resistance occurs, in part, due to adaptation of the tumor and microenvironment to therapeutic stress at a proteomic level. Reverse-phase protein arrays (RPPA) are well suited to proteomic analysis of drug resistance due to high sample throughput, sensitive detection of phosphoproteins, and validation for a large number of critical cellular pathways. AREAS COVERED This review summarizes contributions of RPPA to understanding and combating drug resistance. In particular, contributions of RPPA to understanding resistance to PARP inhibitors, BRAF inhibitors, immune checkpoint inhibitors, and breast cancer investigational therapies are discussed. Articles reviewed were identified by MEDLINE, Scopus, and Cochrane search for keywords 'proteomics,' 'reverse-phase protein array,' 'drug resistance,' 'PARP inhibitor,' 'BRAF inhibitor,' 'immune checkpoint inhibitor,' and 'I-SPY' spanning October 1, 1960 - October 1, 2021. EXPERT OPINION Precision oncology has thus far failed to convert the armament of targeted therapies into durable responses for most patients, highlighting that genetic sequencing alone is insufficient to guide therapy selection and overcome drug resistance. Combined genomic and proteomic analyses paired with creative drug combinations and dosing strategies hold promise for maturing precision oncology into an era of improved patient outcomes.
Collapse
Affiliation(s)
- Ann M Cathcart
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Hannah Smith
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Marilyne Labrie
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Department of Immunology and Cellular Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Gordon B Mills
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
44
|
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
|
45
|
Tarantino P, Corti C, Schmid P, Cortes J, Mittendorf EA, Rugo H, Tolaney SM, Bianchini G, Andrè F, Curigliano G. Immunotherapy for early triple negative breast cancer: research agenda for the next decade. NPJ Breast Cancer 2022; 8:23. [PMID: 35181659 PMCID: PMC8857212 DOI: 10.1038/s41523-022-00386-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/11/2022] [Indexed: 12/31/2022] Open
Abstract
For decades, the systemic treatment of localized triple negative breast cancer (TNBC) has exclusively relied on chemotherapy. Recent advancements, however, are rapidly reshaping the treatment algorithms for this disease. The addition of pembrolizumab to neoadjuvant chemotherapy has indeed shown to significantly improve event-free survival for stage II-III TNBC, leading to its establishment as new standard of care in this setting. This landmark advancement has however raised several important scientific questions. Indeed, we desperately need strategies to identify upfront patients deriving benefit from the addition of immunotherapy. Moreover, the best integration of pembrolizumab with further recent advancements (capecitabine, olaparib) is yet to be defined. Lastly, extensive efforts are needed to minimize the impact on patients of immune-related adverse events and financial toxicity. The next decade of clinical research will be key to overcome these challenges, and ultimately learn how to optimally integrate immunotherapy in the treatment landscape of TNBC.
Collapse
Affiliation(s)
- Paolo Tarantino
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Corti
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Peter Schmid
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Javier Cortes
- Oncology Department, International Breast Cancer Center (IBCC), Quiron Group, Barcelona, Spain
- Medica Scientia Innovation Research (MedSIR), Barcelona, Spain
- Breast Cancer Research Program, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hope Rugo
- University of California San Francisco, UCSF Helen Diller Family Comprehensive Cancer Center Precision Medicine Cancer Building, San Francisco, CA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Fabrice Andrè
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
46
|
Qiu X, Zhao T, Luo R, Qiu R, Li Z. Tumor-Associated Macrophages: Key Players in Triple-Negative Breast Cancer. Front Oncol 2022; 12:772615. [PMID: 35237507 PMCID: PMC8882594 DOI: 10.3389/fonc.2022.772615] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/24/2022] [Indexed: 12/19/2022] Open
Abstract
Triple negative breast cancer (TNBC) refers to the subtype of breast cancer which is negative for ER, PR, and HER-2 receptors. Tumor-associated macrophages (TAMs) refer to the leukocyte infiltrating tumor, derived from circulating blood mononuclear cells and differentiating into macrophages after exuding tissues. TAMs are divided into typical activated M1 subtype and alternately activated M2 subtype, which have different expressions of receptors, cytokines and chemokines. M1 is characterized by expressing a large amount of inducible nitric oxide synthase and TNF-α, and exert anti-tumor activity by promoting pro-inflammatory and immune responses. M2 usually expresses Arginase 1 and high levels of cytokines, growth factors and proteases to support their carcinogenic function. Recent studies demonstrate that TAMs participate in the process of TNBC from occurrence to metastasis, and might serve as potential biomarkers for prognosis prediction.
Collapse
Affiliation(s)
- Xia Qiu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianjiao Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Cell Biology, Wuhan Institute of Bioengineering, Wuhan, China
| | - Ran Luo
- Department of Cell Biology, Wuhan Institute of Bioengineering, Wuhan, China
| | - Ran Qiu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhaoming Li, ; Ran Qiu,
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhaoming Li, ; Ran Qiu,
| |
Collapse
|
47
|
Yi R, Hong S, Zhang Y, Lin A, Ying H, Zou W, Wang Q, Wei T, Cheng Q, Zhu W, Luo P, Zhang J. MHC-II Signature Correlates With Anti-Tumor Immunity and Predicts anti-PD-L1 Response of Bladder Cancer. Front Cell Dev Biol 2022; 10:757137. [PMID: 35223828 PMCID: PMC8873787 DOI: 10.3389/fcell.2022.757137] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/13/2022] [Indexed: 01/02/2023] Open
Abstract
A large proportion of anti-tumor immunity research is focused on major histocompatibility complex class I (MHC-I) molecules and CD8+ T cells. Despite mounting evidence has shown that CD4+ T cells play a major role in anti-tumor immunity, the role of the MHC-II molecules in tumor immunotherapy has not been thoroughly researched and reported. In this study, we defined a MHC-II signature for the first time by calculating the enrichment score of MHC-II protein binding pathway with a single sample gene set enrichment analysis (ssGSEA) algorithm. To evaluate and validate the predictive value of the MHC class II (MHC-II) signature, we collected the transcriptome, mutation data and matched clinical data of bladder cancer patients from IMvigor210, The Cancer Genome Atlas (TCGA) databases and Gene Expression Omnibus (GEO) databases. Comprehensive analyses of immunome, transcriptome, metabolome, genome and drugome were performed in order to determine the association of MHC-II signature and tumor immunotherapy. We identified that MHC-II signature is an independent and favorable predictor of immune response and the prognosis of bladder cancer treated with immune checkpoint inhibitors (ICIs), one that may be superior to tumor mutation burden. MHC-II signature was significantly associated with increased immune cell infiltration and levels of immune-related gene expression signatures. Additionally, transcriptomic analysis showed immune activation in the high-MHC-II signature subgroup, whereas it showed fatty acid metabolism and glucuronidation in the low-MHC-II signature subgroup. Moreover, exploration of corresponding genomic profiles highlighted the significance of tumor protein p53 (TP53) and fibroblast growth factor receptor 3 (FGFR3) alterations. Our results also allowed for the identification of candidate compounds for combined immunotherapy treatment that may be beneficial for patients with bladder cancer and a high MHC-II signature. In conclusion, this study provides a new perspective on MHC-II signature, as an independent and favorable predictor of immune response and prognosis of bladder cancer treated with ICIs.
Collapse
Affiliation(s)
- Ruibin Yi
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuo Hong
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yueming Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haoxuan Ying
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weidong Zou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiongyao Wang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, China
| | - Weiliang Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Weiliang Zhu, ; Peng Luo, ; Jian Zhang,
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Weiliang Zhu, ; Peng Luo, ; Jian Zhang,
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Weiliang Zhu, ; Peng Luo, ; Jian Zhang,
| |
Collapse
|
48
|
Zhang Y, Tong GH, Wei XX, Chen HY, Liang T, Tang HP, Wu CA, Wen GM, Yang WK, Liang L, Shen H. Identification of Five Cytotoxicity-Related Genes Involved in the Progression of Triple-Negative Breast Cancer. Front Genet 2022; 12:723477. [PMID: 35046993 PMCID: PMC8762060 DOI: 10.3389/fgene.2021.723477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Breast cancer is one of the deadly tumors in women, and its incidence continues to increase. This study aimed to identify novel therapeutic molecules using RNA sequencing (RNA-seq) data of breast cancer from our hospital. Methods: 30 pairs of human breast cancer tissue and matched normal tissue were collected and RNA sequenced in our hospital. Differentially expressed genes (DEGs) were calculated with raw data by the R package "edgeR", and functionally annotated using R package "clusterProfiler". Tumor-infiltrating immune cells (TIICs) were estimated using a website tool TIMER 2.0. Effects of key genes on therapeutic efficacy were analyzed using RNA-seq data and drug sensitivity data from two databases: the Cancer Cell Line Encyclopedia (CCLE) and the Cancer Therapeutics Response Portal (CTRP). Results: There were 2,953 DEGs between cancerous and matched normal tissue, as well as 975 DEGs between primary breast cancer and metastatic breast cancer. These genes were primarily enriched in PI3K-Akt signaling pathway, calcium signaling pathway, cAMP signaling pathway, and cell cycle. Notably, CD8+ T cell, M0 macrophage, M1 macrophage, regulatory T cell and follicular helper T cell were significantly elevated in cancerous tissue as compared with matched normal tissue. Eventually, we found five genes (GALNTL5, MLIP, HMCN2, LRRN4CL, and DUOX2) were markedly corelated with CD8+ T cell infiltration and cytotoxicity, and associated with therapeutic response. Conclusion: We found five key genes associated with tumor progression, CD8+ T cell and therapeutic efficacy. The findings would provide potential molecular targets for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University/Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, The First Affiliated Hospital of Guangdong University Of Pharmacy, Guangzhou, China.,Department of Pathology, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Gui-Hui Tong
- Department of Pathology,The first Affiliated Hospital,Guangzhou Medical University, Guangzhou, China
| | - Xu-Xuan Wei
- Department of Pathology, The First Affiliated Hospital of Guangdong University Of Pharmacy, Guangzhou, China
| | - Hai-Yang Chen
- Department of Pathology, The First Affiliated Hospital of Guangdong University Of Pharmacy, Guangzhou, China
| | - Tian Liang
- Department of Pathology, The First Affiliated Hospital of Guangdong University Of Pharmacy, Guangzhou, China
| | - Hong-Ping Tang
- Department of Pathology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Chuan-An Wu
- Department of Prevention and Health Care, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Guo-Ming Wen
- Department of Prevention and Health Care, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Wei-Kang Yang
- Department of Prevention and Health Care, Shenzhen Longhua District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Li Liang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University/Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Hong Shen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University/Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Lin J, Xu A, Jin J, Zhang M, Lou J, Qian C, Zhu J, Wang Y, Yang Z, Li X, Yu W, Liu B, Tao H. MerTK-mediated efferocytosis promotes immune tolerance and tumor progression in osteosarcoma through enhancing M2 polarization and PD-L1 expression. Oncoimmunology 2022; 11:2024941. [PMID: 35036076 PMCID: PMC8757471 DOI: 10.1080/2162402x.2021.2024941] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The poor progress of immunotherapy on osteosarcoma patients requires deeper delineation of immune tolerance mechanisms in the osteosarcoma microenvironment and a new therapeutic strategy. Clearance of apoptotic cells by phagocytes, a process termed “efferocytosis,” is ubiquitous in tumors and mediates the suppression of innate immune inflammatory response. Considering the massive infiltrated macrophages in osteosarcoma, efferocytosis probably serves as a potential target, but is rarely studied in osteosarcoma. Here, we verified M2 polarization and PD-L1 expression of macrophages following efferocytosis. Pharmacological inhibition and genetic knockdown were used to explore the underlying pathway. Moreover, tumor progression and immune landscape were evaluated following inhibition of efferocytosis in osteosarcoma model. Our study indicated that efferocytosis promoted PD-L1 expression and M2 polarization of macrophages. Ëfferocytosis was mediated by MerTK receptor in osteosarcoma and regulated the phenotypes of macrophages through the p38/STAT3 pathway. By establishing the murine osteosarcoma model, we emphasized that inhibition of MerTK suppressed tumor growth and enhanced the T cell cytotoxic function by increasing the infiltration of CD8+ T cells and decreasing their exhaustion. Our findings demonstrate that MerTK-mediated efferocytosis promotes osteosarcoma progression by enhancing M2 polarization of macrophages and PD-L1-induced immune tolerance, which were regulated through the p38/STAT3 pathway.
Collapse
Affiliation(s)
- Jinti Lin
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Ankai Xu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Jiakang Jin
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Man Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Jianan Lou
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Chao Qian
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Jian Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Yitian Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Zhengming Yang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Xiumao Li
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Wei Yu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Bing Liu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| | - Huimin Tao
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
50
|
Liotta LA, Pappalardo PA, Carpino A, Haymond A, Howard M, Espina V, Wulfkuhle J, Petricoin E. Laser Capture Proteomics: spatial tissue molecular profiling from the bench to personalized medicine. Expert Rev Proteomics 2021; 18:845-861. [PMID: 34607525 PMCID: PMC10720974 DOI: 10.1080/14789450.2021.1984886] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Laser Capture Microdissection (LCM) uses a laser to isolate, or capture, specific cells of interest in a complex heterogeneous tissue section, under direct microscopic visualization. Recently, there has been a surge of publications using LCM for tissue spatial molecular profiling relevant to a wide range of research topics. AREAS COVERED We summarize the many advances in tissue Laser Capture Proteomics (LCP) using mass spectrometry for discovery, and protein arrays for signal pathway network mapping. This review emphasizes: a) transition of LCM phosphoproteomics from the lab to the clinic for individualized cancer therapy, and b) the emerging frontier of LCM single cell molecular analysis combining proteomics with genomic, and transcriptomic analysis. The search strategy was based on the combination of MeSH terms with expert refinement. EXPERT OPINION LCM is complemented by a rich set of instruments, methodology protocols, and analytical A.I. (artificial intelligence) software for basic and translational research. Resolution is advancing to the tissue single cell level. A vision for the future evolution of LCM is presented. Emerging LCM technology is combining digital and AI guided remote imaging with automation, and telepathology, to a achieve multi-omic profiling that was not previously possible.
Collapse
Affiliation(s)
- Lance A. Liotta
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Philip A. Pappalardo
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Alan Carpino
- Fluidigm Corporation, South San Francisco, CA, USA
| | - Amanda Haymond
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Marissa Howard
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Virginia Espina
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Julie Wulfkuhle
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| | - Emanuel Petricoin
- Center For Applied Proteomics and Molecular Medicine (CAPMM) School of Systems Biology, College of Sciences, George Mason University, Manassas, VA 20110, USA
| |
Collapse
|