1
|
Sinha S, Ng V, Novaj A, Zhu Y, Yazaki S, Pei X, Derakhshan F, Pareja F, Setton J, Naulin F, Beltrán-Visiedo M, Shin E, Longhini ALF, Gardner R, Ma J, Ma K, Roulston A, Morris S, Koehler M, Powell S, Rosen E, Galluzzi L, Reis-Filho J, Khan A, Riaz N. The cold immunological landscape of ATM-deficient cancers. J Immunother Cancer 2025; 13:e010548. [PMID: 40350205 PMCID: PMC12067784 DOI: 10.1136/jitc-2024-010548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Mutations in genes encoding DNA repair factors, which facilitate mismatch repair, homologous recombination, or DNA polymerase functions, are known to enhance tumor immunogenicity. Ataxia telangiectasia mutated (ATM) is a central regulator of DNA double-strand break repair and is frequently affected by somatic or germline mutations in various cancer types, including breast, prostate, pancreatic, and lung cancer. However, the consequences of ATM loss on tumor immunogenicity are poorly understood. METHODS We generated isogenic ATM-null models using CRISPR in murine triple-negative breast (4T1) and colorectal (CT26) cancer cell lines. ATM inactivation was confirmed by PCR and western blot. Immune cell infiltrates were assessed by flow cytometry and immunohistochemistry in both murine tumors and human samples from breast and lung cancers (via The Cancer Genome Atlas and institutional cohorts). In vivo, the impact of ATM loss on tumor growth and response to immune checkpoint blockade (anti-programmed cell death protein-1 (PD-1)) was evaluated. Furthermore, we compared the effects of different DNA-damaging agents-including an ATR inhibitor (RP-3500), a PARP inhibitor (olaparib), and the topoisomerase II inhibitor etoposide-on interferon-stimulated gene (ISG) expression and immune modulation. RESULTS We find that-in contrast to other DNA repair defects-ATM deficiency (1) fails to encourage immune effector cell infiltration into tumors, and (2) does not enable immune cell recruitment via synthetic lethality strategies in clinical trials, such as with ATR inhibition. Assessing various DNA-damaging agents in Atm null tumors revealed a differential activation of type I interferon (IFN) signaling, with etoposide, a topoisomerase II inhibitor, emerging as the strongest activator of ISG under these conditions. Yet, PD-1-targeted immune checkpoint blockade does not bolster the therapeutic activity of etoposide in Atm-null syngeneic tumor models, nor does it modify the tumor microenvironment, suggesting that type I IFN signaling alone is insufficient to overcome immunosuppression in immunologically cold ATM null neoplasms. CONCLUSIONS ATM deficiency, while compromising DNA repair and enhancing sensitivity to radiation and ATR inhibition, does not increase tumor antigenicity or immunogenicity. Altogether, our results have important implications for the design of novel combination therapies for ATM null tumors and highlight the importance of antigenicity in the immunological consequences of defective DNA repair.
Collapse
Affiliation(s)
- Sonali Sinha
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Victor Ng
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ardijana Novaj
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Yingjei Zhu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Shu Yazaki
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Xin Pei
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jeremy Setton
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Flavie Naulin
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ethan Shin
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Rui Gardner
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jennifer Ma
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kevin Ma
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | | | - Simon Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jorge Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Atif Khan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
2
|
Skelly DA, Graham JP, Cheng M, Furuta M, Walter A, Stoklasek TA, Yang H, Stearns TM, Poirion O, Zhang JG, Grassmann JDS, Luo D, Flynn WF, Courtois ET, Chang CH, Serreze DV, Menghi F, Reinholdt LG, Liu ET. Mapping the genetic landscape establishing a tumor immune microenvironment favorable for anti-PD-1 response. Cell Rep 2025; 44:115698. [PMID: 40343794 DOI: 10.1016/j.celrep.2025.115698] [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: 09/10/2024] [Revised: 01/03/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying host genetic factors modulating immune checkpoint inhibitor (ICI) efficacy is experimentally challenging. Our approach, utilizing the Collaborative Cross mouse genetic resource, fixes the tumor genomic configuration while varying host genetics. We find that response to anti-PD-1 (aPD1) immunotherapy is significantly heritable in four distinct murine tumor models (H2: 0.18-0.40). For the MC38 colorectal carcinoma system, we map four significant ICI response quantitative trait loci (QTLs) with significant epistatic interactions. The differentially expressed genes within these QTLs that define responder genetics are highly enriched for processes involving antigen processing and presentation, allograft rejection, and graft vs. host disease (all p < 1 × 10-10). Functional blockade of two top candidate immune targets, GM-CSF and IL-2RB, completely abrogates the MC38 transcriptional response to aPD1 therapy. Thus, our in vivo experimental platform is a powerful approach for discovery of host genetic factors that establish the tumor immune microenvironment propitious for ICI response.
Collapse
Affiliation(s)
- Daniel A Skelly
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - John P Graham
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | - Mayuko Furuta
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Andrew Walter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | | | - Timothy M Stearns
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Olivier Poirion
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ji-Gang Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Jessica D S Grassmann
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Diane Luo
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - William F Flynn
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Elise T Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; OB/Gyn Department, UConn Health, Farmington, CT 06032, USA
| | - Chih-Hao Chang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - David V Serreze
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Laura G Reinholdt
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.
| |
Collapse
|
3
|
Kaplan Z, Prezioso E, Jain A, Lavu H, Yeo CJ, Bowne WB, Nevler A. Clinical Implications of Mismatch Repair Deficiency in Pancreatic Ductal Adenocarcinoma. Cancer Med 2025; 14:e70960. [PMID: 40366030 PMCID: PMC12076359 DOI: 10.1002/cam4.70960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive and lethal disease, characterized by a limited response to chemotherapy and overall poor prognosis. Pancreatic cancers with a distinct mismatch repair deficiency, although relatively rare, have been shown to be associated with markedly better outcomes in comparison. Furthermore, whereas pancreatic cancers are generally unresponsive to current immunotherapy, this specific group of tumors has been shown to have a notable susceptibility to immune checkpoint inhibitors. AIMS In this review, we aim to summarize the relevant literature regarding mismatch-repair associated pancreatic cancers, the impacted biological mechanisms, and the resulting vulnerabilities for potential opportunistic immunotherapeutic treatment approaches. We will also review the current clinical studies assessing survival outcomes of mismatch repair deficient pancreatic cancers and ongoing clinical trials in this emerging field. RESULTS AND CONCLUSIONS Patients with dMMR/MSI-H pancreatic cancers harbor a distinct phenotype that has increased immune activation, greater responsiveness to immune checkpoint inhibitor therapy and better overall survival when compared to other pancreatic cancers. Although this molecular subtype makes up a small minority of cases, emerging data suggest immunotherapy may offer benefit to these patients.
Collapse
Affiliation(s)
- Zachary Kaplan
- Sidney Kimmel Medical CollegePhiladelphiaPennsylvaniaUSA
| | | | - Aditi Jain
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Harish Lavu
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Charles J. Yeo
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Wilbur B. Bowne
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Avinoam Nevler
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
4
|
Xia F, Verbiest MA, Lundström O, Sonay TB, Baudis M, Anisimova M. Multicancer analyses of short tandem repeat variations reveal shared gene regulatory mechanisms. Brief Bioinform 2025; 26:bbaf219. [PMID: 40401350 PMCID: PMC12096010 DOI: 10.1093/bib/bbaf219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/23/2025] Open
Abstract
Short tandem repeats (STRs) have been reported to influence gene expression across various human tissues. While STR variations are enriched in colorectal, stomach, and endometrial cancers, particularly in microsatellite instable tumors, their functional effects and regulatory mechanisms on gene expression remain poorly understood across these cancer types. Here, we leverage whole-exome sequencing and gene expression data to identify STRs for which repeat lengths are associated with the expression of nearby genes (eSTRs) in colorectal, stomach, and endometrial tumors. While most eSTRs are cancer-specific, shared eSTRs across multiple cancers exhibit consistent effects on gene expression. Notably, coding-region eSTRs identified in all three cancer types show positive correlations with nearby gene expression. We further validate the functional effects of eSTRs by demonstrating associations between somatic eSTR mutations and gene expression changes during the transition from normal to tumor tissues, suggesting their potential roles in tumorigenesis. Combined with DNA methylation data, we perform the first quantitative analysis of the interplay between STR variations and DNA methylation in tumors. We identify eSTRs where repeat lengths are associated with methylation levels of nearby CpG sites (meSTRs) and show that >70% of eSTRs are significantly linked to local DNA methylation. Importantly, the effects of meSTRs on DNA methylation remain consistent across cancer types. Overall, our findings enhance the understanding of how functional STR variations influence gene expression and DNA methylation. Our study highlights shared regulatory mechanisms of STRs across multiple cancers, offering a foundation for future research into their broader implications in tumor biology.
Collapse
Affiliation(s)
- Feifei Xia
- Institute of Computational Life Sciences, Zurich University of Applied Sciences, Schloss 4, 8820 Wädenswil, Switzerland
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
- Swiss Institute of Bioinformatics, Amphipôle, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland
| | - Max Adriaan Verbiest
- Institute of Computational Life Sciences, Zurich University of Applied Sciences, Schloss 4, 8820 Wädenswil, Switzerland
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
- Swiss Institute of Bioinformatics, Amphipôle, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland
| | - Oxana Lundström
- Department of Computer Science and Media Technology, Linnaeus University, Universitetsplatsen 1, 352 52 Växjö, Sweden
| | - Tugce Bilgin Sonay
- Institute of Computational Life Sciences, Zurich University of Applied Sciences, Schloss 4, 8820 Wädenswil, Switzerland
- Swiss Institute of Bioinformatics, Amphipôle, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland
| | - Michael Baudis
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
- Swiss Institute of Bioinformatics, Amphipôle, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland
| | - Maria Anisimova
- Institute of Computational Life Sciences, Zurich University of Applied Sciences, Schloss 4, 8820 Wädenswil, Switzerland
- Swiss Institute of Bioinformatics, Amphipôle, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland
| |
Collapse
|
5
|
Zheng S, Wang H, Wang Y. Thyroid hormone receptor interacting protein 13 is associated with prognosis and immunotherapy efficacy in human cancers: a pan-cancer analysis. Discov Oncol 2025; 16:580. [PMID: 40253660 PMCID: PMC12009794 DOI: 10.1007/s12672-025-02385-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
Thyroid hormone receptor-interacting protein 13 (TRIP13) is involved in the regulation of mitosis and is overexpressed in multiple cancers. However, there is no systematic assessment of the role of TRIP13 in the immunotherapy response across human cancers. Therefore, a pan-cancer analysis involving expression, prognosis, immune-related mechanisms, and biomarker values was performed to explore the associations between TRIP13 expression and the immunotherapy response. TRIP13 is highly expressed in various types of cancer, increasing patient outcomes in eight types of cancer. TRIP13 expression was correlated with significant tumor mutation burden and microsatellite instability, and its mutations were linked with poor prognosis in patients with adrenocortical carcinoma. TRIP13 promoted endothelial cell and hematopoietic stem cell infiltration in human cancers. Additionally, TRIP13 mutation significantly increased the infiltration of CD8 + T cells in kidney renal clear cell carcinoma, which might contribute to poor prognosis. Furthermore, three key genes that interact with TRIP13 were identified: CDC20, RAD1, and MAD2L1, which are related to the cell cycle and ultimately promote tumorigenesis and proliferation. The expression of TRIP13 was significantly greater in kidney renal clear cell carcinoma, liver hepatocellular carcinoma, and pancreatic adenocarcinoma cells than in corresponding normal cells according to qPCR. Taken together, these findings indicate that TRIP13 is associated with poor prognosis in eight human cancers and serves as a novel biomarker for predicting immunotherapy efficacy. Our first pan-cancer study contributes to personalized precision medicine in cancer immunotherapy, promoting subsequent clinical management and improving patient prognosis.
Collapse
Affiliation(s)
- ShengYao Zheng
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - HongYi Wang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingyi Wang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Menkiti FE, Okani CO, Onyiaorah IV, Ukah CO, Menkiti IO, Ihekwoaba EC, Okoye OA, Ofiaeli OC, Akpuaka FC. Immunohistochemical expression of PD-L1 in colorectal carcinoma among black patients and the clinicopathological correlates: a cross-sectional study. BMC Gastroenterol 2025; 25:277. [PMID: 40254571 PMCID: PMC12010690 DOI: 10.1186/s12876-025-03862-7] [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: 12/02/2024] [Accepted: 04/07/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) incidence is rising in Nigeria, with majority of patients presenting with advanced disease. Despite promising results of PD-L1 antibody therapy in clinical trials, efficacy data exclusively derives from Caucasian populations, leaving a critical knowledge gap for African populations. This study investigated PD-L1 expression in CRC among blacks, correlating it with clinicopathologic parameters. METHODS The immunohistochemical expression of PD-L1 was evaluated in 96 cases of CRC diagnosed between February 2022 and January 2024, using formalin-fixed paraffin-embedded (FFPE) tissue blocks. Statistical analysis was performed using SPSS version 25. The relationships between the PD-L1 expression and the clinicopathological parameters of CRC patients were determined using the chi-square test and Spearman's rank correlation. p < 0.05 was considered to be statistically significant. RESULTS CRC showed a male: female ratio of 1:1.8, most occurred in the seventh decade and 54.17% were right-sided. Adenocarcinoma NOS accounted for 72.5%. The majority (n = 55, 57.3%) of the patients were diagnosed at an advanced stage. PD-L1 expression was observed in 86.46% of cases, significantly correlating with tumour Size (r = 0.263, p = 0.010*), histologic Grade (r = 0.446, p = 0.000*) and tumour Stage (r = 0.367, p = 0.000*). CONCLUSION This study highlighted the high frequency of PD-L1 expression in CRC among black patients, with significant associations to clinicopathologic parameters. The findings suggest the potential benefit of PD-1/PD-L1 targeted therapies and emphasize the need for enhanced early detection and screening in Nigeria.
Collapse
Affiliation(s)
- Felix Emeka Menkiti
- Department of Anatomic Pathology and Forensic Medicine, Faculty of Basic Clinical Sciences, Nnamdi Azikiwe University, Awka, Nigeria.
- Department of Histopathology, Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria.
| | - Chukwudi Onyeaghana Okani
- Department of Histopathology, Faculty of Basic Clinical Sciences, Chukwuemeka Odumegwu Ojukwu University, Awka, Anambra State, Nigeria
| | - Igwebuike Victor Onyiaorah
- Department of Anatomic Pathology and Forensic Medicine, Faculty of Basic Clinical Sciences, Nnamdi Azikiwe University, Awka, Nigeria
- Department of Histopathology, Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria
| | - Cornelius Ozobia Ukah
- Department of Anatomic Pathology and Forensic Medicine, Faculty of Basic Clinical Sciences, Nnamdi Azikiwe University, Awka, Nigeria
- Department of Histopathology, Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria
| | - Ifeoma Oluchukwu Menkiti
- Department of Nursing, College of Nursing, Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria
| | - Eric Chukwudi Ihekwoaba
- Department of Surgery, Faculty of Medicine, Nnamdi Azikiwe University, Nnewi Campus, Nnewi, Anambra State, Nigeria
| | - Odili Aloysius Okoye
- Department of Surgery, Faculty of Medicine, Nnamdi Azikiwe University, Nnewi Campus, Nnewi, Anambra State, Nigeria
| | - Ogochukwu Chioma Ofiaeli
- Department of Paediatrics, Nnamdi Azikiwe University, Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria
| | - Frank Chinedu Akpuaka
- Department of Surgery, Chukwuemeka Odumegwu Ojukwu University, Awka, Anambra State, Nigeria
| |
Collapse
|
7
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
8
|
Zhang C, Wang H, Li X, Jiang Y, Sun G, Yu H. Enhancing antitumor immunity: the role of immune checkpoint inhibitors, anti-angiogenic therapy, and macrophage reprogramming. Front Oncol 2025; 15:1526407. [PMID: 40260303 PMCID: PMC12009726 DOI: 10.3389/fonc.2025.1526407] [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: 11/11/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer treatment has long been hindered by the complexity of the tumor microenvironment (TME) and the mechanisms that tumors employ to evade immune detection. Recently, the combination of immune checkpoint inhibitors (ICIs) and anti-angiogenic therapies has emerged as a promising approach to improve cancer treatment outcomes. This review delves into the role of immunostimulatory molecules and ICIs in enhancing anti-tumor immunity, while also discussing the therapeutic potential of anti-angiogenic strategies in cancer. In particular, we highlight the critical role of endoplasmic reticulum (ER) stress in angiogenesis. Moreover, we explore the potential of macrophage reprogramming to bolster anti-tumor immunity, with a focus on restoring macrophage phagocytic function, modulating hypoxic tumor environments, and targeting cytokines and chemokines that shape immune responses. By examining the underlying mechanisms of combining ICIs with anti-angiogenic therapies, we also review recent clinical trials and discuss the potential of biomarkers to guide and predict treatment efficacy.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xinying Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuxin Jiang
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hanqing Yu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Ambrosini M, Manca P, Nasca V, Sciortino C, Ghelardi F, Seligmann JF, Taieb J, Pietrantonio F. Epidemiology, pathogenesis, biology and evolving management of MSI-H/dMMR cancers. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01015-z. [PMID: 40181086 DOI: 10.1038/s41571-025-01015-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
Deficiency in DNA mismatch repair (dMMR) is a common pathway of carcinogenesis across different tumour types and confers a characteristic microsatellite instability-high (MSI-H) molecular phenotype. The prevalence of the MSI-H/dMMR phenotype is highest in endometrial and colorectal cancers, and this phenotype is associated with a distinct tumour biology, prognosis and responsiveness to various anticancer treatments. In a minority of patients, MSI-H/dMMR cancers result from an inherited pathogenic variant in the context of Lynch syndrome, which has important implications for familial genetic screening. Whether these hereditary cancers have a different biology and clinical behaviour to their sporadic counterparts remains uncertain. Interest in this tumour molecular subtype has increased following the discovery of the high sensitivity of metastatic MSI-H/dMMR cancers to immune-checkpoint inhibitors (ICIs) in a histology-agnostic manner, which reflects the genomic hypermutation resulting from dMMR that renders these tumours highly immunogenic and immune infiltrated. This vulnerability is now also being exploited in early stage disease settings. Despite this common biological foundation, different MSI-H/dMMR cancers have histotype-specific features that correspond to their particular cell or tissue of origin, which might be associated with differences in prognosis and sensitivity to ICIs. In this Review, we provide an overview of the epidemiology, biology, pathogenesis, clinical diagnosis and treatment of MSI-H/dMMR tumours as a histology-agnostic cancer phenomenon. We also highlight peculiarities associated with specific pathogenetic alterations and histologies of MSI-H/dMMR tumours.
Collapse
Affiliation(s)
- Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Paolo Manca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vincenzo Nasca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carolina Sciortino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Ghelardi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Jenny F Seligmann
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Paris-Cité University, SIRIC CARPEM Comprehensive Cancer Center, Paris, France
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
10
|
Zhang H, Wu J, Cui L, Wang T, Jin H, Guo H, Xie C, Li L, Wang X, Wang Z. Pyrithione zinc alters mismatch repair to trigger tumor immunogenicity. Oncogene 2025; 44:983-995. [PMID: 39814851 DOI: 10.1038/s41388-024-03272-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Mismatch repair deficiency (dMMR) cancers are highly sensitive to immunotherapy, but only account for a small fraction of cancer patients. How to increase immunotherapy efficacy on MMR-proficient (pMMR) cancer is still a major challenge. This study demonstrates that pyrithione zinc (PYZ), an FDA-approved drug, can enhance tumor immunogenicity via altering MMR and activating STING signaling. Mechanistically, PYZ elevates levels of ROS, leading to the upregulation of HIF-1α and DNA damage, while also inhibiting the expression of DNA mismatch repair proteins MSH2 and MSH6, together promoting DNA damage accumulation. Therefore, the administration of PYZ results in the accumulation of DNA damage, leading to the activation of STING signaling, which enhances tumor immunogenicity. Knockout of Sting diminishes the activation of IFN-I signaling induced by PYZ and reduces tumor immunogenicity. Furthermore, in vivo administration of PYZ promotes the infiltration of CD8+ T cells into the tumor and inhibits tumor growth, an effect that is attenuated in Nude mice or mice with CD8+ T cell depletion or deficiency of Ifnar. Overall, our findings showed that pyrithione zinc could trigger tumor immunogenicity by downregulating MMR machinery and activating STING pathway in tumor cells, and provide a translational approach to improve immunotherapy on pMMR cancer.
Collapse
Affiliation(s)
- Huanling Zhang
- Guangzhou Institute of Clinical Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Jiaxin Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lei Cui
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tiantian Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huan Jin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Guo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunyuan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lin Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaojuan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zining Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
11
|
Balaban R, Cohen M. Decoding multicellular interaction networks-a new horizon in tumor microenvironment research. Mol Oncol 2025; 19:957-960. [PMID: 39906955 PMCID: PMC11977648 DOI: 10.1002/1878-0261.13810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025] Open
Abstract
The tumor microenvironment (TME) milieu directs a plethora of tumor-modulating functions. Recent years have seen pivotal breakthroughs in our understanding of the TME's role in tumor initiation and progression, with tangible clinical applications. Individual components of the TME exert their function predominantly by cell-cell crosstalk, both in the form of physical interaction and secreted factors. Notably, different spatial niches represent exclusive signaling hubs in the TME, propagating pro- or antitumoral functions. The exploration of these interactions has been vastly facilitated by novel molecular technologies, each of which provides a different perspective on this intricate intercellular communication network. Together, these complementary methods paint a detailed, high-resolution map of the TME's interaction landscape. In this viewpoint, we explore how cellular interactions can unlock a new level of understanding of TME complexity, and discuss the promises and challenges of characterizing tumors based on their cellular interaction footprint.
Collapse
Affiliation(s)
- Roi Balaban
- Department of Clinical Microbiology and Immunology, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
| | - Merav Cohen
- Department of Clinical Microbiology and Immunology, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
| |
Collapse
|
12
|
Liu Y, Xie Y, Chen Y, Duan J, Bao C, Wang J, Feng H, Wang M, Ren Y, Li P, Luo Q, Xu J, Jiang M, Men Y, Wu Y, Li J, Wang G, Lu W. A protease-cleavable liposome for co-delivery of anti-PD-L1 and doxorubicin for colon cancer therapy in mice. Nat Commun 2025; 16:2854. [PMID: 40128211 PMCID: PMC11933685 DOI: 10.1038/s41467-025-57965-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Immune checkpoint blockade therapy using programmed cell death 1 (PD1) or programmed death ligand 1 (PD-L1) has made significant progress in the treatment of advanced cancers, with some patients achieving long-term remission without clinical recurrence. However, only a minority of colon cancer patients respond to the therapy. Here, we report a protease-cleavable anti-PD-L1 antibody liposome, eLipo anti-PD-L1, for enhancing colon cancer therapy. In vivo, eLipo anti-PD-L1 is cleaved by legumain at colon cancer site into pegylated anti-PD-L1 and cancer-homing doxorubicin liposome. Functional assessments show cancer-targeting, legumain-responding, tumor-penetrating, and immune-activating effects, as well as efficacy in treating colon cancer-bearing mice in vivo. Further mechanistic analysis implicates genes related to T cell differentiation and T cell receptor signaling as potential molecular mediators. Lastly, human colorectal cancer tissue evaluations verify expressions of PD-L1 and legumain, hinting a potential translatability. Our study thus suggests that eLipo anti-PD-L1 may be a feasible vector for co-delivery of immunochemotherapy for colon cancer.
Collapse
Affiliation(s)
- Yixuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jialun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Chunjie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jinling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Hexuan Feng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Mengjie Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuxin Ren
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Peishan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Qian Luo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jiarui Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Min Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yanchen Men
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yang Wu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Guiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Wanliang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
| |
Collapse
|
13
|
Tithi TI, Mo J, Borcherding N, Jo S, Kates HR, Cho E, Cash KE, Honda M, Wang L, Ahmed KK, Shirlekar K, Chen L, Gibson-Corley K, Weigel R, Spies M, Kolb R, Zhang W. The distinct roles of MSH2 and MLH1 in basal-like breast cancer and immune modulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.07.20.549745. [PMID: 37745359 PMCID: PMC10515760 DOI: 10.1101/2023.07.20.549745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The mismatch repair (MMR) pathway is known as a tumor suppressive pathway and genes involved in MMR are commonly mutated in hereditary colorectal or other cancer types. However, the function of MMR genes/proteins in breast cancer progression and metastasis are largely undefined. We found that MSH2, but not MLH1, is highly enriched in basal-like breast cancer (BLBC) and that its protein expression is inversely correlated with overall survival time (OS). MSH2 expression is frequently elevated due to genomic amplification or gain-of-expression in BLBC, which results in increased MSH2 protein to pair with MSH6 (collectively referred to as MutSα). Genetic deletion of MSH2 or MLH1 results in a contrasting phenotype in metastasis, with MSH2 -deletion leading to reduced metastasis and MLH1 -deletion to enhanced liver or lung metastasis. Mechanistically, MSH2 - but not MLH1 - binds to the promoter region of interferon α receptor 1 ( IFNAR1 ) and suppresses its expression in BLBC. Deletion of MSH2 initiates a chain of immune reactions via the upregulation of IFNAR1 expression and the activation of type 1 interferon signaling, which explains a highly immune active tumor microenvironment in tumors with MSH2-deficiency. Our study supports the contrasting functions of MSH2 and MLH1 in BLBC progression and metastasis due to the differential regulation of IFNAR1 expression, which challenges the paradigm of the MMR pathway as a universal tumor suppressive mechanism.
Collapse
|
14
|
Zhou J, Liu J, Yu Y, Nie H, Hong Y, Ning Y, Yang C, Lai J, Wang H, Tang X, Wang F, Zhao Q. Melanoma Cell Adhesion Molecule Plays a Pivotal Role in Proliferation, Migration, Tumor Immune Microenvironment, and Immunotherapy in Colorectal Cancer. Cancer Med 2025; 14:e70740. [PMID: 40042109 PMCID: PMC11880918 DOI: 10.1002/cam4.70740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 05/12/2025] Open
Abstract
INTRODUCTION MCAM, alternatively referred to as CD146, is an integral membrane glycoprotein belonging to the immunoglobulin superfamily. However, its importance in the tumorigenesis of colorectal cancer is still partially understood. Therefore, this study was designed to investigate the significance of MCAM in colorectal cancer. METHODS MCAM expression was analyzed by TCGA and GEO databases. qRT-PCR and IHC analysis were conducted to validate MCAM expression in patient tissues. The tumor-inhibiting ability of MCAM was further assessed by CCK-8 assay, colony formation assay, and wound-healing assay. qRT-PCR and WB analysis were conducted to evaluate the expression of EMT markers and MMP2/9. qRT-PCR analysis was utilized to detect the polarization status of macrophages. Kaplan-Meier curve, univariate, and multivariate cox analyses were employed to verify the ability of MCAM in prognosis prediction. TIDE scores were used to assess the impact of MCAM on immunotherapy. RESULTS The expression of MCAM was significantly downregulated in CRC, and low MCAM expression revealed poor prognosis in CRC patients. Moreover, MCAM overexpression inhibited the proliferation, migration, and invasive ability of CRC cells. Additionally, MCAM overexpression suppressed N-cadherin and MMP2/9 expression. Furthermore, MCAM impacted M1 macrophage polarization. MCAM is an independent predictor of CRC patient prognosis through Cox regression analysis. Lastly, TIDE score analysis indicated that elevated expression of MCAM increased immunotherapy efficacy. CONCLUSION The findings of this research suggest that MCAM impacts M1 macrophage polarization and enhances immunotherapy efficacy, underscoring its potential as a therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Jingkai Zhou
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jing Liu
- Emergency Medicine CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yali Yu
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Haihang Nie
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yuntian Hong
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yumei Ning
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Chao Yang
- Department of RadiologyXiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Jun Lai
- The Infirmary of Hangzhou Power Supply Company of State Grid, Zhejiang Electric Power Co., LtdHangzhouChina
| | - Haizhou Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xuelian Tang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Fan Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qiu Zhao
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
15
|
Ying L, Zhang L, Chen Y, Huang C, Zhou J, Xie J, Liu L. Predicting immunotherapy prognosis and targeted therapy sensitivity of colon cancer based on a CAF-related molecular signature. Sci Rep 2025; 15:6387. [PMID: 39984646 PMCID: PMC11845748 DOI: 10.1038/s41598-025-90899-z] [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: 11/12/2024] [Accepted: 02/17/2025] [Indexed: 02/23/2025] Open
Abstract
The role of cancer-associated fibroblasts (CAFs) in modulating the tumor microenvironment (TME) is gaining attention, yet their impact on prognosis and therapeutic response in colon cancer remains unclear. Here, we identified genes associated with CAF infiltration via weighted gene co-expression network analysis (WGCNA) utilizing data from The Cancer Genome Atlas (TCGA) and GSE39582 cohorts. Univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses were used to construct CAF molecular signatures (CAFscore). Patients were categorized into high and low CAFscore groups to analyze clinicopathological traits, somatic mutations, immune evasion, and treatment responses. In this study, a total of 244 genes were correlated with CAF infiltration, with 11 linked to overall survival. Notably, FSTL3, CRIP2, and SLC2A3 were selected for the CAFscore. A higher CAFscore was associated with poorer prognoses, increased malignancy, and therapeutic resistance, particularly among patients with high tumor mutation burden and microsatellite instability. Furthermore, elevated FSTL3 expression was associated with reduced CD8+ T cell infiltration, indicating a suppressive TME. Mechanistically, CAFs may promote immune evasion via NAMPT ligand-receptor interactions based on single-cell RNA sequencing data. Thus, the CAFscore is crucial for personalizing treatment strategies and identifying patients who require more aggressive management.
Collapse
Affiliation(s)
- Leqian Ying
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Lu Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Yanping Chen
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Chunchun Huang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Jingyi Zhou
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Jinbing Xie
- Department of Radiology, Nurturing Center of Jiangsu Province for the State Laboratory of AI Imaging and Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Lin Liu
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China.
| |
Collapse
|
16
|
Schoenfeld JD, Azad NS, Gross J, Chen L, Overman MJ, Kao K, Jackson L, Brunnquell D, Bu X, Coppola C, Guan P, Lee J, Sims D, Fuchs R, Weirather JL, Pfaff KL, Gunasti L, Ranasinghe S, Hamilton SR, Wang V, O’Dwyer PJ, Wu CJ, Rodig SJ, Patton DR, Harris L. Next-Generation Sequencing-Based MSI Scoring Predicts Benefit in Mismatch Repair-Deficient Tumors Treated with Nivolumab: Follow-up on NCI-MATCH Arm Z1D. Clin Cancer Res 2025; 31:667-677. [PMID: 39670863 PMCID: PMC11831103 DOI: 10.1158/1078-0432.ccr-24-0427] [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: 02/08/2024] [Revised: 06/17/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
PURPOSE Mismatch repair-deficient (dMMR) tumors have demonstrated favorable responses to immune checkpoint inhibition targeting PD-1. However, more in-depth identification of predictors of response could further refine patient selection for immunotherapy treatment. PATIENTS AND METHODS We undertook integrated evaluation performed on samples collected from 28 of 42 patients enrolled on the NCI-Molecular Analysis for Therapy Choice arm Z1D trial that evaluated PD-1 inhibition treatment with nivolumab in patients with noncolorectal dMMR tumors. Genomic analyses were performed using next-generation sequencing (NGS), whole-exome sequencing, and RNA sequencing and supplemented by multiplex immunofluorescence performed on tissue samples. RESULTS In this dMMR population, more extensive alterations of microsatellites as assessed by measures of NGS were associated with clinical benefit and tumor mutational burden. RNA sequencing further revealed associations between clinical benefit and immune infiltration index. Gene sets enriched in patients with clinical benefit included IFN signaling, antigen processing, and PI3K-AKT-mTOR signaling, whereas hedgehog signaling was found to be enriched in subjects lacking clinical benefit. CONCLUSIONS These genomic data highlight the importance of immune infiltration and antigen presentation in dMMR tumors that respond to immune checkpoint blockade. In addition, they suggest that, even within a dMMR population, NGS-based measures of microsatellite instability could serve as biomarkers of immunotherapy response.
Collapse
Affiliation(s)
- Jonathan D. Schoenfeld
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nilofer S. Azad
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Jacob Gross
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Li Chen
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katrina Kao
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Latifa Jackson
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Donna Brunnquell
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Xiangning Bu
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Christina Coppola
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ping Guan
- Cancer Diagnosis Program, National Cancer Institute, Bethesda, Maryland
| | - Jennifer Lee
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David Sims
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Rebecca Fuchs
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jason L. Weirather
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kathleen L. Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lauren Gunasti
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Srin Ranasinghe
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Victoria Wang
- Dana-Farber Cancer Institute–ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - Peter J. O’Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Catherine J. Wu
- Center for Hematologic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Scott J. Rodig
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - David R. Patton
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lyndsay Harris
- Cancer Diagnosis Program, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
17
|
Mestrallet G, Brown M, Vaninov N, Cho NW, Velazquez L, Ananthanarayanan A, Spitzer M, Vabret N, Cimen Bozkus C, Samstein RM, Bhardwaj N. Coordinated macrophage and T cell interactions mediate response to checkpoint blockade in colorectal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637954. [PMID: 40027748 PMCID: PMC11870396 DOI: 10.1101/2025.02.12.637954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Mismatch repair deficiency (MMRd), either due to inherited or somatic mutation, is prevalent in colorectal cancer (CRC) and other cancers. While anti-PD-1 therapy is utilized in both local and advanced disease, up to 50% of MMRd CRC fail to respond. Using animal and human models of MMRd, we determined that interactions between MHC+ C1Q+ CXCL9+ macrophages and TCF+ BHLHE40+ PRF1+ T cell subsets are associated with control of MMRd tumor growth, during anti-PD-1 treatment. In contrast, resistance is associated with upregulation of TIM3, LAG3, TIGIT, and PD-1 expression on T cells, and infiltration of the tumor with immunosuppressive TREM2+ macrophages and monocytes. By combining anti-PD-1 with anti-LAG3/CTLA4/TREM2, up to 100% tumor eradication was achieved in MMRd CRC and remarkably, in >70% in MMRp CRC. This study identifies key T cell and macrophage subsets mediating the efficacy of immunotherapy in overcoming immune escape in both MMRd and MMRp CRC settings. Abstract Figure Highlights Anti-PD-1 therapy leads to the accumulation and colocalization of MHCI/II+ C1Q+ CXCL9+ macrophages and DCs with TCF+ CCL5+ T cells that have high TCR diversity.Resistance to anti-PD-1 therapy involves multiple T cell checkpoints, TREM2+ macrophages, IL1B+ TREM1+ monocytes and neutrophils, and IFITM+ tumor cells.Simultaneous blockade of PD-1, LAG3, CTLA-4 and TREM2 dramatically prevents progression of both MMRd and MMRp tumors.Combination therapy completely eliminates tumors by leveraging MHC+ macrophage, CD4+ and CD8+ T cell interactions, facilitating durable anti-tumor effects.
Collapse
|
18
|
Cook AL, Sur S, Dobbyn L, Watson E, Cohen JD, Ptak B, Lee BS, Paul S, Hsiue E, Popoli M, Vogelstein B, Papadopoulos N, Bettegowda C, Gabrielson K, Zhou S, Kinzler KW, Wyhs N. Identification of nonsense-mediated decay inhibitors that alter the tumor immune landscape. eLife 2025; 13:RP95952. [PMID: 39960487 PMCID: PMC11832170 DOI: 10.7554/elife.95952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Despite exciting developments in cancer immunotherapy, its broad application is limited by the paucity of targetable antigens on the tumor cell surface. As an intrinsic cellular pathway, nonsense-mediated decay (NMD) conceals neoantigens through the destruction of the RNA products from genes harboring truncating mutations. We developed and conducted a high-throughput screen, based on the ratiometric analysis of transcripts, to identify critical mediators of NMD in human cells. This screen implicated disruption of kinase SMG1's phosphorylation of UPF1 as a potential disruptor of NMD. This led us to design a novel SMG1 inhibitor, KVS0001, that elevates the expression of transcripts and proteins resulting from human and murine truncating mutations in vitro and murine cells in vivo. Most importantly, KVS0001 concomitantly increased the presentation of immune-targetable human leukocyte antigens (HLA) class I-associated peptides from NMD-downregulated proteins on the surface of human cancer cells. KVS0001 provides new opportunities for studying NMD and the diseases in which NMD plays a role, including cancer and inherited diseases.
Collapse
Affiliation(s)
- Ashley L Cook
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Surojit Sur
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Laura Dobbyn
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Evangeline Watson
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Joshua D Cohen
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Blair Ptak
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Bum Seok Lee
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Suman Paul
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Emily Hsiue
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Maria Popoli
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Bert Vogelstein
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Howard Hughes Medical Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nickolas Papadopoulos
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Chetan Bettegowda
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurosurgery, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Kathy Gabrielson
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Shibin Zhou
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Kenneth W Kinzler
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nicolas Wyhs
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
19
|
Cho NW, Guldberg SM, Nabet BY, Yu JZ, Kim EJ, Hiam-Galvez KJ, Yee JL, DeBarge R, Tenvooren I, Ashitey NA, Lynce F, Dillon DA, Rosenbluth JM, Spitzer MH. T Cells Instruct Immune Checkpoint Inhibitor Therapy Resistance in Tumors Responsive to IL1 and TNFα Inflammation. Cancer Immunol Res 2025; 13:229-244. [PMID: 39404741 PMCID: PMC11790381 DOI: 10.1158/2326-6066.cir-24-0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/10/2024] [Accepted: 10/11/2024] [Indexed: 02/04/2025]
Abstract
Resistance to immune checkpoint inhibitors (ICI) is common, even in tumors with T-cell infiltration. We thus investigated consequences of ICI-induced T-cell infiltration in the microenvironment of resistant tumors. T cells and neutrophil numbers increased in ICI-resistant tumors following treatment, in contrast to ICI-responsive tumors. Resistant tumors were distinguished by high expression of IL1 receptor 1, enabling a synergistic response to IL1 and TNFα to induce G-CSF, CXCL1, and CXCL2 via NF-κB signaling, supporting immunosuppressive neutrophil accumulation in tumor. Perturbation of this inflammatory resistance circuit sensitized tumors to ICIs. Paradoxically, T cells drove this resistance circuit via TNFα both in vitro and in vivo. Evidence of this inflammatory resistance circuit and its impact also translated to human cancers. These data support a mechanism of ICI resistance, wherein treatment-induced T-cell activity can drive resistance in tumors responsive to IL1 and TNFα, with important therapeutic implications.
Collapse
Affiliation(s)
- Nam Woo Cho
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Sophia M. Guldberg
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Barzin Y. Nabet
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA *present address: Genentech Inc., South San Francisco, CA, USA
| | - Jie Zeng Yu
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Kamir J. Hiam-Galvez
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jacqueline L. Yee
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rachel DeBarge
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Iliana Tenvooren
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | - Naa Asheley Ashitey
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| | | | - Deborah A. Dillon
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA
| | - Jennifer M. Rosenbluth
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, and Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Matthew H. Spitzer
- Departments of Otolaryngology and Microbiology & Immunology, Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
20
|
Badani A, Ozair A, Khasraw M, Woodworth GF, Tiwari P, Ahluwalia MS, Mansouri A. Immune checkpoint inhibitors for glioblastoma: emerging science, clinical advances, and future directions. J Neurooncol 2025; 171:531-547. [PMID: 39570554 DOI: 10.1007/s11060-024-04881-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Glioblastoma (GBM), the most common and aggressive primary central nervous system (CNS) tumor in adults, continues to have a dismal prognosis. Across hundreds of clinical trials, few novel approaches have translated to clinical practice while survival has improved by only a few months over the past three decades. Randomized controlled trials of immune checkpoint inhibitors (ICIs), which have seen impressive success for advanced or metastatic extracranial solid tumors, have so far failed to demonstrate a clinical benefit for patients with GBM. This has been secondary to GBM heterogeneity, the unique immunosuppressive CNS microenvironment, immune-evasive strategies by cancer cells, and the rapid evolution of tumor on therapy. This review aims to summarize findings from major clinical trials of ICIs for GBM, review historic failures, and describe currently promising avenues of investigation. We explore the biological mechanisms driving ICI responses, focusing on the role of the tumor microenvironment, immune evasion, and molecular biomarkers. Beyond conventional monotherapy approaches targeting PD-1, PD-L1, CTLA-4, we describe emerging approaches for GBM, such as dual-agent ICIs, and combination of ICIs with oncolytic virotherapy, antigenic peptide vaccines, chimeric antigenic receptor (CAR) T-cell therapy, along with nanoparticle-based delivery systems to enhance ICI efficacy. We highlight potential strategies for improving patient selection and treatment personalization, along with real-time, longitudinal monitoring of therapeutic responses through advanced imaging and liquid biopsy techniques. Integrated radiomics, tissue, and plasma-based analyses, may potentially uncover immunotherapeutic response signatures, enabling early, adaptive therapeutic adjustments. By specifically targeting current therapeutic challenges, outcomes for GBM patients may potentially be improved.
Collapse
Affiliation(s)
- Aarav Badani
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neuroscience, University of California, Berkeley, CA, USA
| | - Ahmad Ozair
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mustafa Khasraw
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, NC, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Brain Tumor Center, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
- University of Maryland - Medicine Institute for Neuroscience Discovery (UM-MIND), Baltimore, MD, USA
| | - Pallavi Tiwari
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- William S. Middleton Memorial Veterans Affairs (VA) Healthcare, Madison, WI, USA
| | - Manmeet S Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
21
|
Ding J, Wang T, Lin Z, Li Z, Yang J, Li F, Rong Y, Chen X, He C. Chiral polypeptide hydrogels regulating local immune microenvironment and anti-tumor immune response. Nat Commun 2025; 16:1222. [PMID: 39890820 PMCID: PMC11785995 DOI: 10.1038/s41467-025-56137-w] [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: 01/09/2025] [Indexed: 02/03/2025] Open
Abstract
The impact of chirality on immune response has attracted great interest in cancer vaccine research recently. However, the study of chiral synthetic polypeptide hydrogels as cancer vaccines as well as of the impact of biomaterials themselves for antitumor immunotherapy has rarely been reported. Here, we show the key role of residue chirality of polypeptide hydrogels in antitumor immunity and local immune microenvironment regulation. Compared to poly(γ-ethyl-L-glutamate)-based hydrogels (L-Gel), poly(γ-ethyl-D-glutamate)-based hydrogels (D-Gel) induces enhanced level of immune cell infiltration. However, D-Gel causes higher levels of suppressive markers on antigen-presenting cells and even induces stronger T cell exhaustion than L-Gel. Finally, D-Gel establishes a local chronic inflammatory and immunosuppressive microenvironment and shows insufficient anti-tumor effects. Conversely, the milder host immune responses induced by L-Gel leads to more effective tumor inhibition. This study provides insights on the role of residue chirality in the regulation of local immune microenvironment and affecting antitumor immune response.
Collapse
Affiliation(s)
- Junfeng Ding
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Tianran Wang
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Zhiqiang Lin
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Zhenyu Li
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Jiaxuan Yang
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Fujiang Li
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Yan Rong
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
22
|
Liu X, Wang S, Lv H, Chen E, Yan L, Yu J. Advances in the relationship of immune checkpoint inhibitors and DNA damage repair. Curr Res Transl Med 2025; 73:103494. [PMID: 39824061 DOI: 10.1016/j.retram.2025.103494] [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: 01/22/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Cancer immunotherapy, alongside surgery, radiation therapy, and chemotherapy, has emerged as a key treatment modality. Immune checkpoint inhibitors (ICIs) represent a promising immunotherapy that plays a critical role in the management of various solid tumors. However, the limited efficacy of ICI monotherapy and the development of primary or secondary resistance to combination therapy remain a challenge. Consequently, identifying molecular markers for predicting ICI efficacy has become an area of active clinical research. Notably, the correlation between DNA damage repair (DDR) mechanisms and the effectiveness of ICI treatment has been established. This review outlines the two primary pathways of DDR, namely, the homologous recombination repair pathway and the mismatch repair pathway. The relationship between these key genes and ICIs has been discussed and the potential of these genes as molecular markers for predicting ICI efficacy summarized.
Collapse
Affiliation(s)
- Xiaolin Liu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Shan Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Hongwei Lv
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Enli Chen
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Li Yan
- School of Humanities, Beijing University of Chinese Medicine, Beijing, PR China
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
23
|
Robles-Oteíza C, Hastings K, Choi J, Sirois I, Ravi A, Expósito F, de Miguel F, Knight JR, López-Giráldez F, Choi H, Socci ND, Merghoub T, Awad M, Getz G, Gainor J, Hellmann MD, Caron É, Kaech SM, Politi K. Hypoxia is linked to acquired resistance to immune checkpoint inhibitors in lung cancer. J Exp Med 2025; 222:e20231106. [PMID: 39585348 PMCID: PMC11602551 DOI: 10.1084/jem.20231106] [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: 06/28/2023] [Revised: 04/29/2024] [Accepted: 09/27/2024] [Indexed: 11/26/2024] Open
Abstract
Despite the established use of immune checkpoint inhibitors (ICIs) to treat non-small cell lung cancer (NSCLC), only a subset of patients benefit from treatment and ∼50% of patients whose tumors respond eventually develop acquired resistance (AR). To identify novel drivers of AR, we generated murine Msh2 knock-out (KO) lung tumors that initially responded but eventually developed AR to anti-PD-1, alone or in combination with anti-CTLA-4. Resistant tumors harbored decreased infiltrating T cells and reduced cancer cell-intrinsic MHC-I and MHC-II levels, yet remained responsive to IFNγ. Resistant tumors contained extensive regions of hypoxia, and a hypoxia signature derived from single-cell transcriptional profiling of resistant cancer cells was associated with decreased progression-free survival in a cohort of NSCLC patients treated with anti-PD-1/PD-L1 therapy. Targeting hypoxic tumor regions using a hypoxia-activated pro-drug delayed AR to ICIs in murine Msh2 KO tumors. Thus, this work provides a rationale for targeting tumor metabolic features, such as hypoxia, in combination with immune checkpoint inhibition.
Collapse
Affiliation(s)
| | | | - Jungmin Choi
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | | | - Arvind Ravi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - James R. Knight
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | | | - Hyejin Choi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas D. Socci
- Marie-Josee and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program & Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Mark Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Gad Getz
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research and Department of Pathology, Massachusetts Genral Hospital, Boston, MA, USA
| | - Justin Gainor
- Center for Thoracic Cancers, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew D. Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Étienne Caron
- CHU Sainte-Justine Research Center, Montreal, Canada
- Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute, La Jolla, CA, USA
| | - Katerina Politi
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Departments of Pathology and Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Wu Y, Jiang X, Yu Z, Xing Z, Ma Y, Qing H. Mechanisms of Anti-PD Therapy Resistance in Digestive System Neoplasms. Recent Pat Anticancer Drug Discov 2025; 20:1-25. [PMID: 38305306 PMCID: PMC11865675 DOI: 10.2174/0115748928269276231120103256] [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: 06/27/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 02/03/2024]
Abstract
Digestive system neoplasms are highly heterogeneous and exhibit complex resistance mechanisms that render anti-programmed cell death protein (PD) therapies poorly effective. The tumor microenvironment (TME) plays a pivotal role in tumor development, apart from supplying energy for tumor proliferation and impeding the body's anti-tumor immune response, the TME actively facilitates tumor progression and immune escape via diverse pathways, which include the modulation of heritable gene expression alterations and the intricate interplay with the gut microbiota. In this review, we aim to elucidate the mechanisms underlying drug resistance in digestive tumors, focusing on immune-mediated resistance, microbial crosstalk, metabolism, and epigenetics. We will highlight the unique characteristics of each digestive tumor and emphasize the significance of the tumor immune microenvironment (TIME). Furthermore, we will discuss the current therapeutic strategies that hold promise for combination with cancer immune normalization therapies. This review aims to provide a thorough understanding of the resistance mechanisms in digestive tumors and offer insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuxia Wu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zongrui Xing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yong Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Huiguo Qing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
25
|
Pan W, Jia Z, Zhao X, Chang K, Liu W, Tan W. Identification of immunogenic cell death gene-related subtypes and risk model predicts prognosis and response to immunotherapy in ovarian cancer. PeerJ 2024; 12:e18690. [PMID: 39686988 PMCID: PMC11648682 DOI: 10.7717/peerj.18690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Background Immunogenic cell death (ICD) has been associated with enhanced anti-tumor immunotherapy by stimulating adaptive immune responses and remodeling the immune microenvironment in tumors. Nevertheless, the role of ICD-related genes in ovarian cancer (OC) and tumor microenvironment remains unexplored. Methods In this study, high-throughput transcriptomic data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases as training and validation sets separately were obtained and proceeded to explore ICD-related clusters, and an ICD-related risk signature was conducted based on the least absolute shrinkage and selection operator (LASSO) Cox regression model by iteration. Multiple tools including CIBERSORT, ESTIMATE, GSEA, TIDE, and immunohistochemistry were further applied to illustrate the biological roles of ICD-related genes as well as the prognostic capacity of ICD risk signature in OC. Results Two ICD-related subtypes were identified, with the ICD-high subtype showing more intense immune cell infiltration and higher activities of immune response signaling, along with a favorable prognosis. Additionally, four candidate ICD genes (IFNG, NLRP3, FOXP3, and IL1B) were determined to potentially impact OC prognosis, with an upregulated expression of NLRP3 in OC and metastatic omental tissues. A prognostic model based on these genes was established, which could predict overall survival (OS) and response to immunotherapy for OC patients, with lower-risk patients benefiting more from immunotherapy. Conclusion Our research conducted a prognostic and prediction of immunotherapy response model based on ICD genes, which could be instrumental in assessing prognosis and assigning immunotherapeutic strategies for OC patients. NLRP3 is a promising target for prognosis in OC.
Collapse
Affiliation(s)
- Wenjing Pan
- Department of Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhaoyang Jia
- Department of Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xibo Zhao
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University of Medical Sciences, Guangzhou, China
| | - Kexin Chang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Wei Liu
- Department of Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenhua Tan
- Department of Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
26
|
Sako C, Duan C, Maresca K, Kent S, Schmidt TG, Aerts HJWL, Parikh RB, Simon GR, Jordan P. Real-World and Clinical Trial Validation of a Deep Learning Radiomic Biomarker for PD-(L)1 Immune Checkpoint Inhibitor Response in Advanced Non-Small Cell Lung Cancer. JCO Clin Cancer Inform 2024; 8:e2400133. [PMID: 39671539 DOI: 10.1200/cci.24.00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 12/15/2024] Open
Abstract
PURPOSE This study developed and validated a novel deep learning radiomic biomarker to estimate response to immune checkpoint inhibitor (ICI) therapy in advanced non-small cell lung cancer (NSCLC) using real-world data (RWD) and clinical trial data. MATERIALS AND METHODS Retrospective RWD of 1,829 patients with advanced NSCLC treated with PD-(L)1 ICIs were collected from 10 academic and community institutions in the United States and Europe. The RWD included data sets for discovery (Data Set A-Discovery, n = 1,173) and independent test (Data Set B, n = 458). A radiomic pipeline, containing a deep learning feature extractor and a survival model, generated the computed tomography (CT) response score (CTRS) applied to the pretreatment routine CT/positron emission tomography (PET)-CT scan. An enhanced CTRS (eCTRS) also incorporated age, sex, treatment line, and lesion annotations. Performance was evaluated against progression-free survival (PFS) and overall survival (OS). Biomarker generalizability was further evaluated using a secondary analysis of a prospective clinical trial (ClinicalTrials.gov identifier: NCT02573259) evaluating the PD-1 inhibitor sasanlimab in second or later line of treatment (Data Set C, n = 54). RESULTS In RWD Test Data Set B, the CTRS identified patients with a high probability of response to ICI with a PFS hazard ratio (HR) of 0.46 (95% CI, 0.26 to 0.82) and an OS HR of 0.50 (95% CI, 0.28 to 0.92) in the first-line ICI monotherapy cohort, after adjustment for baseline covariates including the PD-L1 tumor proportion score. In Clinical Trial Data Set C, the CTRS demonstrated an adjusted PFS HR of 1.03 (95% CI, 0.43 to 2.47) and an OS HR of 0.33 (95% CI, 0.14 to 0.91). The CTRS and eCTRS outperformed traditional imaging biomarkers of lesion size in PFS and OS for RWD Test Data Set B and in OS for the Clinical Trial Data Set. CONCLUSION The study developed and validated a deep learning radiomic biomarker using pretreatment routine CT/PET-CT scans to identify ICI benefit in advanced NSCLC.
Collapse
|
27
|
Zhang X, Wang H, Li Y, Yan W, Chen Y, Song S, Liu N, Zhang C, Niu Z, Hou H. Neoadjuvant PD-(L)1 blockade with or without chemotherapy versus chemotherapy alone in mismatch repair-deficient, potentially resectable stage III-IV A gastric cancer patients: a single-center retrospective study. World J Surg Oncol 2024; 22:313. [PMID: 39593126 PMCID: PMC11590301 DOI: 10.1186/s12957-024-03601-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Currently, PD-(L)1 blockade-based neoadjuvant treatment has shown promising outcomes in patients with potentially resectable gastric cancer. In this real-world study, we aimed to retrospectively observe the efficacy including tumor response and event-free survival (EFS), and safety of PD-(L)1 blockade-based neoadjuvant treatment versus chemotherapy alone in potentially resectable gastric cancer patients with microsatellite instability-high (MSI-H) or mismatch-repair deficient (dMMR) status. METHODS We retrospectively collected the clinical data of patients with potentially resectable gastric cancer and MSI-H/dMMR status who received neoadjuvant treatment followed by D2 gastrectomy at the Affiliated Hospital of Qingdao University from January 2019 to June 2023. The outcomes of interest mainly included overall complete response (CR) rates, radiographical and pathological tumor response, treatment-related adverse events (TRAEs), and EFS. RESULTS In total, 30 patients were included in the analysis; 23 patients received neoadjuvant PD-(L)1 blockade plus chemotherapy or PD-(L)1 blockade monotherapy, and seven patients received neoadjuvant chemotherapy. In the PD-(L)1 blockade-based treatment group, 7 of 23 patients (30.4%, 95% CI 0.141-0.530) achieved pathological CR (pCR), while three patients with radiographical CR did not undergo surgery. In contrast, 1 of 7 (14.3%) patients in the neoadjuvant chemotherapy group achieved pCR. The overall CR rate was 43.5% (10 of 23, 95% CI 0.239-0.651) in the PD-(L)1 blockade-based treatment group and 14.3% (1 of 7, 95% CI 0.026-0.513) in the chemotherapy group. The neoadjuvant PD-(L)1 blockade-based treatment regimen was mild and well tolerated. By the latest follow-up, median EFS time was not reached in both cohorts. CONCLUSION In potentially resectable gastric cancer patients with MSI-H/dMMR status, PD-(L)1 blockade-based neoadjuvant treatment regimen provided promising clinical benefits and was well tolerated.
Collapse
Affiliation(s)
- Xuchen Zhang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266035, China
| | - Huiyun Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, No. 7 Jiaxing Road, Qingdao, Shandong, 266031, China
| | - Yi Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China
| | - Weihua Yan
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China
| | - Yunqing Chen
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China
| | - Shanai Song
- Department of Oncology, The Affiliated Hospital of Qingdao University, No. 7 Jiaxing Road, Qingdao, Shandong, 266031, China
| | - Ning Liu
- Department of Oncology, The Affiliated Hospital of Qingdao University, No. 7 Jiaxing Road, Qingdao, Shandong, 266031, China
| | - Chuantao Zhang
- Department of Oncology, The Affiliated Hospital of Qingdao University, No. 7 Jiaxing Road, Qingdao, Shandong, 266031, China
| | - Zhaojian Niu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266003, China.
| | - Helei Hou
- Department of Oncology, The Affiliated Hospital of Qingdao University, No. 7 Jiaxing Road, Qingdao, Shandong, 266031, China.
| |
Collapse
|
28
|
Wang Y, Guan Y, Lai X, Liu Y, Chang Z, Wang X, Wang Q, Liu J, Zhao J, Yang S, Wang J, Song X. THOR: a TMB heterogeneity-adaptive optimization model predicts immunotherapy response using clonal genomic features in group-structured data. Brief Bioinform 2024; 26:bbae648. [PMID: 39679440 DOI: 10.1093/bib/bbae648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/04/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024] Open
Abstract
With the increasing number of indications for immune checkpoint inhibitors in early and advanced cancers, the prospect of a tumor-agnostic biomarker to prioritize patients is compelling. Tumor mutation burden (TMB) is a widely endorsed biomarker that quantifies nonsynonymous mutations within tumor DNA, essential for neoantigen production, which, in turn, correlates with the immune response and guides decision-making. However, the general clinical application of TMB-relying on simple mutational counts targeted at a single endpoint-does not adequately capture the complex clonal structure of tumors nor the multifaceted nature of prognostic indicators. This recognition has spurred the exploration of sophisticated high-dimensional regression techniques. Unfortunately, the limited cohort sizes in immunotherapy trials have hindered the full potential of these advanced methods. Our approach considers patient subgroups as related yet distinct entities, enabling precise tailoring and refinement to address subgroup-specific dynamics. Given the deficiencies and the constraints, we introduce a TMB heterogeneity-optimized regression (THOR). This innovative model enhances the predictive capabilities of TMB by integrating tumor clonality and a diverse spectrum of clinical endpoints, further augmented by fusion techniques across subgroups to facilitate robust data sharing and interpretation. Our simulations validate THOR's superiority in parameter estimation for statistical inference. Clinically, we assess the utility of THOR in a structured cohort of 238 cancer patients undergoing immunotherapy, supplemented by 2212 patients across 19 subgroups from public datasets. The forecast of the responses and comparison of survival hazards demonstrate that THOR significantly enhances patient stratification and prognostic predictions by incorporating complex immunogenetic biology and subgroup-specific dynamics.
Collapse
Affiliation(s)
- Yixuan Wang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Jiangning, Nanjing 211106, China
| | - Yanfang Guan
- School of Computer Science and Technology, Faculty of Electronics and Information Engineering, Xi'an Jiaotong University, 28 Xianning West Road, Beilin, Xi'an 710049, China
- Geneplus-Beijing Institute, 8 Life Park Road, Changping, Beijing 102206, China
| | - Xin Lai
- School of Computer Science and Technology, Faculty of Electronics and Information Engineering, Xi'an Jiaotong University, 28 Xianning West Road, Beilin, Xi'an 710049, China
| | - Yuqian Liu
- School of Computer Science and Technology, Faculty of Electronics and Information Engineering, Xi'an Jiaotong University, 28 Xianning West Road, Beilin, Xi'an 710049, China
| | - Zhili Chang
- School of Computer Science and Technology, Faculty of Electronics and Information Engineering, Xi'an Jiaotong University, 28 Xianning West Road, Beilin, Xi'an 710049, China
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., 128 Huakang Road, Pukou, Nanjing 210032, China
| | - Xiaonan Wang
- School of Computer Science and Technology, Faculty of Electronics and Information Engineering, Xi'an Jiaotong University, 28 Xianning West Road, Beilin, Xi'an 710049, China
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., 128 Huakang Road, Pukou, Nanjing 210032, China
| | - Quan Wang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Jiangning, Nanjing 211106, China
| | - Jingjing Liu
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Jiangning, Nanjing 211106, China
| | - Jian Zhao
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Jiangning, Nanjing 211106, China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 3 Shangqin Road, Xincheng, Xi'an 710004, China
| | - Jiayin Wang
- School of Computer Science and Technology, Faculty of Electronics and Information Engineering, Xi'an Jiaotong University, 28 Xianning West Road, Beilin, Xi'an 710049, China
| | - Xiaofeng Song
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Jiangning, Nanjing 211106, China
| |
Collapse
|
29
|
Tang E, Tang C, Lin X, Chen Y, Zhou Y, Pan C, Jiang H. Clinical performance of the TrueMark™ MSI assay for microsatellite instability detection in a Chinese colorectal cancer cohort. Gene 2024; 927:148745. [PMID: 38969248 DOI: 10.1016/j.gene.2024.148745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Microsatellite instability (MSI) and mismatch repair (MMR) detection is valuable in assessing prognosis and treatment options. However, the conventional detection methods such as immunohistochemistry (IHC) are limited by not fully consistent results as well as a long turnaround time. TrueMark™ MSI Assay is a novel solution for MSI analysis, but lack of research support in the Chinese colorectal cancer (CRC) patients. MATERIALS AND METHODS 60 dMMR and 60 pMMR CRC samples identified by IHC were collected and their MSI status were detected using TrueMark™ MSI assay with an expanded panel of 13 markers. The overall performance and diagnostic concordance between TrueMark™ MSI test and MMR IHC analysis were assessed and analyzed. RESULTS According to the TrueMark™ test, 55 out of the 120 (45.8 %) CRCs were identified as MSI-high (MSI-H) with an instability at ≥ 4/13 markers. Compared with the MMR IHC analysis, an overall percent agreement of 94.2 % and a Kappa of 0.883 were achieved. For the seven inconsistent samples, tumor mutation burden analysis was performed and the results supported the diagnosis by TrueMark™ test. To confirm the robustness of the above findings, a validation was performed in an independent cohort comprising 51 consecutive CRCs. Furthermore, an optimized panel composed of NR-21, NR-24, NR-27, ABI-16, ABI-17 and ABI-20B was developed by multivariate logistic regression model, and showed 100 % agreement with the 13-marker panel for MSI detection in both the derivation and validation sets. CONCLUSION TrueMark™ MSI provides a fast, reliable and highly automated solution to MSI detection in Chinese CRC patients, and the new 6-marker panel we established shows promise deserving further evaluation.
Collapse
Affiliation(s)
- Erjiang Tang
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University, Shanghai, China
| | - Cui Tang
- Department of Radiology, Yangpu Hospital, Tongji University, Shanghai, China
| | - Xuejing Lin
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University, Shanghai, China
| | - Ying Chen
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University, Shanghai, China
| | - Yi Zhou
- Department of Medical Oncology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenyu Pan
- Department of General Surgery, Yangpu Hospital, Tongji University, Shanghai, China.
| | - Huihong Jiang
- Department of General Surgery, Yangpu Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
30
|
Alban TJ, Riaz N, Parthasarathy P, Makarov V, Kendall S, Yoo SK, Shah R, Weinhold N, Srivastava R, Ma X, Krishna C, Mok JY, van Esch WJE, Garon E, Akerley W, Creelan B, Aanur N, Chowell D, Geese WJ, Rizvi NA, Chan TA. Neoantigen immunogenicity landscapes and evolution of tumor ecosystems during immunotherapy with nivolumab. Nat Med 2024; 30:3209-3222. [PMID: 39349627 PMCID: PMC12066197 DOI: 10.1038/s41591-024-03240-y] [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: 11/07/2023] [Accepted: 08/08/2024] [Indexed: 11/16/2024]
Abstract
Neoantigen immunoediting drives immune checkpoint blockade efficacy, yet the molecular features of neoantigens and how neoantigen immunogenicity shapes treatment response remain poorly understood. To address these questions, 80 patients with non-small cell lung cancer were enrolled in the biomarker cohort of CheckMate 153 (CA209-153), which collected radiographic guided biopsy samples before treatment and during treatment with nivolumab. Early loss of mutations and neoantigens during therapy are both associated with clinical benefit. We examined 1,453 candidate neoantigens, including many of which that had reduced cancer cell fraction after treatment with nivolumab, and identified 196 neopeptides that were recognized by T cells. Mapping these neoantigens to clonal dynamics, evolutionary trajectories and clinical response revealed a strong selection against immunogenic neoantigen-harboring clones. We identified position-specific amino acid and physiochemical features related to immunogenicity and developed an immunogenicity score. Nivolumab-induced microenvironmental evolution in non-small cell lung cancer shared some similarities with melanoma, yet critical differences were apparent. This study provides unprecedented molecular portraits of neoantigen landscapes underlying nivolumab's mechanism of action.
Collapse
Affiliation(s)
- Tyler J Alban
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prerana Parthasarathy
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sviatoslav Kendall
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Seong-Keun Yoo
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Rachna Shah
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nils Weinhold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raghvendra Srivastava
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xiaoxiao Ma
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | - Edward Garon
- Department of Thoracic Medical Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Wallace Akerley
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Benjamin Creelan
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Diego Chowell
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Naiyer A Rizvi
- Synthekine, Menlo Park, CA, USA
- Thoracic Oncology, Columbia University, New York, NY, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA.
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.
- National Center for Regenerative Medicine, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
31
|
Amodio V, Vitiello PP, Bardelli A, Germano G. DNA repair-dependent immunogenic liabilities in colorectal cancer: opportunities from errors. Br J Cancer 2024; 131:1576-1590. [PMID: 39271762 PMCID: PMC11554791 DOI: 10.1038/s41416-024-02848-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the major causes of cancer death worldwide. Chemotherapy continues to serve as the primary treatment modality, while immunotherapy is largely ineffective for the majority of CRC patients. Seminal discoveries have emphasized that modifying DNA damage response (DDR) mechanisms confers both cell-autonomous and immune-related vulnerabilities across various cancers. In CRC, approximately 15% of tumours exhibit alterations in the mismatch repair (MMR) machinery, resulting in a high number of neoantigens and the activation of the type I interferon response. These factors, in conjunction with immune checkpoint blockades, collectively stimulate anticancer immunity. Furthermore, although less frequently, somatic alterations in the homologous recombination (HR) pathway are observed in CRC; these defects lead to genome instability and telomere alterations, supporting the use of poly (ADP-ribose) polymerase (PARP) inhibitors in HR-deficient CRC patients. Additionally, other DDR inhibitors, such as Ataxia Telangiectasia and Rad3-related protein (ATR) inhibitors, have shown some efficacy both in preclinical models and in the clinical setting, irrespective of MMR proficiency. The aim of this review is to elucidate how preexisting or induced vulnerabilities in DNA repair pathways represent an opportunity to increase tumour sensitivity to immune-based therapies in CRC.
Collapse
Affiliation(s)
- V Amodio
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy
| | - P P Vitiello
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy
| | - A Bardelli
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy.
| | - G Germano
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Department of Medical Biotechnologies and Translational Medicine, University of Milano, 20133, Milan, Italy.
| |
Collapse
|
32
|
Holla VR, Kahle MP, Kim SH, Ronaghy A, Yang RK, Patel KP, Routbort MJ, Overman MJ, Dumbrava EE, Shaw KRM, Karp DD, Meric-Bernstam F. Genomic Alterations in DNA Mismatch Repair Genes Across Different Cancer Types. JCO Precis Oncol 2024; 8:e2400419. [PMID: 39576951 PMCID: PMC11825140 DOI: 10.1200/po-24-00419] [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: 06/21/2024] [Revised: 09/11/2024] [Accepted: 10/09/2024] [Indexed: 11/24/2024] Open
Abstract
PURPOSE PD-1 inhibition is effective in patients with mismatch repair deficient (dMMR) solid tumors in a tumor-agnostic fashion. However, dMMR testing by immunohistochemistry (IHC) is not routinely performed across tumor types. By contrast, next-generation sequencing (NGS) for somatic genomic alterations is frequently performed across tumor types. We hypothesized that NGS would identify patients with alterations in mismatch repair (MMR) genes and that these patients would have higher rates of MMR protein loss by IHC. This would support the utility of IHC reflex testing after NGS and potential matching to approved therapeutic options. METHODS From January 2016 to December 2021, 15,701 patients with solid tumors received NGS covering the MMR genes, and 4,994 patients had both IHC and NGS. Sequencing results were analyzed for mutations in MMR genes, tumor type distribution, and concordance with IHC results when available. RESULTS Six hundred and ninety-eight (4.4%) of 15,701 patients had mutations in one of the MMR genes. Mutations were found across tumor types. Three hundred and seventeen (6.3%) of 4,994 patients displayed IHC loss for at least one MMR protein. 33.8% patients (110/325) patients with MMR mutations had dMMR, compared with just 4.4% (207/4,669) patients without mutations (P < .001); dMMR rate varied by mutation type. CONCLUSION Mutations in MMR genes are found in multiple tumor types where IHC testing is not routine. Reflex IHC testing of patients carrying MMR gene mutations, especially those known or inferred to be inactivating, may identify more patients with dMMR and matched treatment options. However, dedicated IHC screening is needed to capture majority of the patients.
Collapse
Affiliation(s)
- Vijaykumar R. Holla
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Michael P Kahle
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Sun-Hee Kim
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Arash Ronaghy
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Richard K Yang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Keyur P Patel
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Mark J Routbort
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Kenna R Mills Shaw
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| |
Collapse
|
33
|
Su Z, He Y, You L, Chen J, Zhang G, Liu Z. SPP1+ macrophages and FAP+ fibroblasts promote the progression of pMMR gastric cancer. Sci Rep 2024; 14:26221. [PMID: 39482333 PMCID: PMC11528032 DOI: 10.1038/s41598-024-76298-w] [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: 02/12/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024] Open
Abstract
Immunotherapy has become a primary and secondary treatment for gastric cancer (GC) patients with mismatch repair deficiency (dMMR), and is used in both perioperative and advanced stages. The tumor immune microenvironment (TiME) is crucial for immunotherapy efficacy, yet the impact of MMR status on TiME remains understudied. We employed single-cell RNA sequencing (scRNA-seq) to analyze 33 fresh tissue samples from 25 patients, which included 10 normal tissues, 6 dMMR tumor tissues, and 17 pMMR tumor tissues, aiming to characterize the cellular and molecular components of the TiME. The proficient mismatch repair (pMMR) group displayed a significantly higher prevalence of a specific GC cell type, termed GC2, characterized by increased hypoxia, epithelial-mesenchymal transition (EMT), and angiogenic activities compared to the dMMR group. GC2 cells overexpressed BEX3 and GPC3, and they significantly correlated with poorer survival. The pMMR group also showed increased infiltration of SPP1 + macrophages and FAP + fibroblasts, exhibiting strong hypoxic and pro-angiogenic features. Furthermore, a higher proportion of E2 endothelial cells, involved in extracellular matrix (ECM) remodeling and showing heightened VEGF pathway, HIF pathway, and angiogenesis activity, were identified in pMMR patients. Intercellular communication analyses revealed that GC2 cells, SPP1 + macrophages, FAP + fibroblasts, and E2 endothelial cells interact through VEGF, SPP1, and MIF signals, forming a TiME characterized by hypoxia, pro-angiogenesis, and ECM remodeling. This study uncovered TiME heterogeneity among GC patients with different MMR states, highlighting that the pMMR TiME is distinguished by hypoxia, pro-angiogenesis, and ECM remodeling, driven by the presence of GC2 cells, SPP1 + macrophages, FAP + fibroblasts, and E2 endothelial cells. These findings are pivotal for developing targeted immunotherapies for GC patients with pMMR.
Collapse
Affiliation(s)
- Zhixiong Su
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Yufang He
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Lijie You
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Jingbo Chen
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China.
| | - Guifeng Zhang
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China.
| | - Zhenhua Liu
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
34
|
Budczies J, Kazdal D, Menzel M, Beck S, Kluck K, Altbürger C, Schwab C, Allgäuer M, Ahadova A, Kloor M, Schirmacher P, Peters S, Krämer A, Christopoulos P, Stenzinger A. Tumour mutational burden: clinical utility, challenges and emerging improvements. Nat Rev Clin Oncol 2024; 21:725-742. [PMID: 39192001 DOI: 10.1038/s41571-024-00932-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Tumour mutational burden (TMB), defined as the total number of somatic non-synonymous mutations present within the cancer genome, varies across and within cancer types. A first wave of retrospective and prospective research identified TMB as a predictive biomarker of response to immune-checkpoint inhibitors and culminated in the disease-agnostic approval of pembrolizumab for patients with TMB-high tumours based on data from the Keynote-158 trial. Although the applicability of outcomes from this trial to all cancer types and the optimal thresholds for TMB are yet to be ascertained, research into TMB is advancing along three principal avenues: enhancement of TMB assessments through rigorous quality control measures within the laboratory process, including the mitigation of confounding factors such as limited panel scope and low tumour purity; refinement of the traditional TMB framework through the incorporation of innovative concepts such as clonal, persistent or HLA-corrected TMB, tumour neoantigen load and mutational signatures; and integration of TMB with established and emerging biomarkers such as PD-L1 expression, microsatellite instability, immune gene expression profiles and the tumour immune contexture. Given its pivotal functions in both the pathogenesis of cancer and the ability of the immune system to recognize tumours, a profound comprehension of the foundational principles and the continued evolution of TMB are of paramount relevance for the field of oncology.
Collapse
Affiliation(s)
- Jan Budczies
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany.
- Center for Personalized Medicine (ZPM), Heidelberg, Germany.
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Michael Menzel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Susanne Beck
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Klaus Kluck
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Christian Altbürger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Michael Allgäuer
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Aysel Ahadova
- Department of Applied Tumour Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumour Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Solange Peters
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne University, Lausanne, Switzerland
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Petros Christopoulos
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumour Diseases at Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany.
- Center for Personalized Medicine (ZPM), Heidelberg, Germany.
| |
Collapse
|
35
|
Cai Y, Lu Z, Chen C, Zhu Y, Chen Z, Wu Z, Peng J, Zhu X, Liu Z, Li B, Zhang M, Huang J, Li Y, Liu Y, Ma Q, He C, Chen S, Tian W, Fan L, Ning C, Geng H, Xu B, Li H, Zhu X, Fang J, Wang X, Zhang S, Jin M, Huang C, Yang X, Tian J, Miao X. An atlas of genetic effects on cellular composition of the tumor microenvironment. Nat Immunol 2024; 25:1959-1975. [PMID: 39223350 DOI: 10.1038/s41590-024-01945-3] [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: 12/31/2022] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Deciphering the composition of the tumor microenvironment (TME) is critical for understanding tumorigenesis and to design immunotherapies. In the present study, we mapped genetic effects on cell-type proportions using single-cell and bulk RNA sequencing data, identifying 3,494 immunity quantitative trait loci (immunQTLs) across 23 cancer types from The Cancer Genome Atlas. Functional annotation revealed regulatory potential and we further assigned 1,668 genes that regulate TME composition. We constructed a combined immunQTL map by integrating data from European and Chinese colorectal cancer (CRC) samples. A polygenic risk score that incorporates these immunQTLs and hits on a genome-wide association study outperformed in CRC risk stratification within 447,495 multiethnic individuals. Using large-scale population cohorts, we identified that the immunQTL rs1360948 is associated with CRC risk and prognosis. Mechanistically, the rs1360948-G-allele increases CCL2 expression, recruiting regulatory T cells that can exert immunosuppressive effects on CRC progression. Blocking the CCL2-CCR2 axis enhanced anti-programmed cell death protein 1 ligand therapy. Finally, we have established a database (CancerlmmunityQTL2) to serve the research community and advance our understanding of immunogenomic interactions in cancer pathogenesis.
Collapse
Affiliation(s)
- Yimin Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Can Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Zhirui Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Zuyou Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Jingyi Peng
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Xuanyu Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Ziying Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Jinyu Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Yanmin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Yizhuo Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Qianying Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Chunyi He
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Shuoni Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Wen Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Linyun Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Caibo Ning
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Hui Geng
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Haijie Li
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Zhu
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Fang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaoyang Wang
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Shaokai Zhang
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Meng Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoqun Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xiaojun Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.
- Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
36
|
Taherifard E, Tran K, Saeed A, Yasin JA, Saeed A. Biomarkers for Immunotherapy Efficacy in Advanced Hepatocellular Carcinoma: A Comprehensive Review. Diagnostics (Basel) 2024; 14:2054. [PMID: 39335733 PMCID: PMC11431712 DOI: 10.3390/diagnostics14182054] [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: 07/26/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver malignancy and the sixth most common cancer globally, remains fatal for many patients with inappropriate responses to treatment. Recent advancements in immunotherapy have transformed the treatment landscape for advanced HCC. However, variability in patient responses to immunotherapy highlights the need for biomarkers that can predict treatment outcomes. This manuscript comprehensively reviews the evolving role of biomarkers in immunotherapy efficacy, spanning from blood-derived indicators-alpha-fetoprotein, inflammatory markers, cytokines, circulating tumor cells, and their DNA-to tissue-derived indicators-programmed cell death ligand 1 expression, tumor mutational burden, microsatellite instability, and tumor-infiltrating lymphocytes. The current body of evidence suggests that these biomarkers hold promise for improving patient selection and predicting immunotherapy outcomes. Each biomarker offers unique insights into disease biology and the immune landscape of HCC, potentially enhancing the precision of treatment strategies. However, challenges such as methodological variability, high costs, inconsistent findings, and the need for large-scale validation in well-powered two-arm trial studies persist, making them currently unsuitable for integration into standard care. Addressing these challenges through standardized techniques and implementation of further studies will be critical for the future incorporation of these biomarkers into clinical practice for advanced HCC.
Collapse
Affiliation(s)
- Erfan Taherifard
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Krystal Tran
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Ali Saeed
- Department of Medicine, Ochsner Lafayette General Medical Center, Lafayette, LA 70503, USA
| | - Jehad Amer Yasin
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
37
|
Cordts SC, Yuki K, Henao Echeverri MF, Narasimhan B, Kuo CJ, Tang SKY. Microdissection tools to generate organoids for modeling the tumor immune microenvironment. MICROSYSTEMS & NANOENGINEERING 2024; 10:126. [PMID: 39251611 PMCID: PMC11385579 DOI: 10.1038/s41378-024-00756-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024]
Abstract
Patient-derived tumor organoids have emerged as promising models for predicting personalized drug responses in cancer therapy, but they typically lack immune components. Preserving the in vivo association between tumor cells and endogenous immune cells is critical for accurate testing of cancer immunotherapies. Mechanical dissection of tumor specimens into tumor fragments, as opposed to enzymatic digestion into single cells, is essential for maintaining these native tumor-immune cell spatial relationships. However, conventional mechanical dissection relying on manual mincing is time-consuming and irreproducible. This study describes two microdissection devices, the µDicer and µGrater, to facilitate the generation of intact tumor fragments from mouse B16 melanoma, a common model of human melanoma. The µDicer- and µGrater-cut tumor fragments were used to generate air‒liquid interface (ALI) organoids that copreserve tumor cells with infiltrating immune subsets without artificial reconstitution. The µDicer, consisting of a hexagonal array of silicon microblades, was employed to investigate the effect of organoid size. The viability of ALI organoid immune cells appeared insensitive to organoid sizes exceeding ~400 µm but diminished in organoids ~200 µm in size. The µGrater, consisting of an array of submillimeter holes in stainless steel, was employed to accelerate dissection. For the samples studied, the µGrater was 4.5 times faster than manual mincing. Compared with those generated by manual mincing, ALI organoids generated by the µGrater demonstrated similar viability, immune cell composition, and responses to anti-PD-1 immunotherapy. With further optimization, the µGrater holds potential for integration into clinical workflows to support the advancement of personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Seth C Cordts
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | | | | | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | - Sindy K Y Tang
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
38
|
Okada M, Yamasaki S, Nakazato H, Hirahara Y, Ishibashi T, Kawamura M, Shimizu K, Fujii SI. ARID1A-Deficient Tumors Acquire Immunogenic Neoantigens during the Development of Resistance to Targeted Therapy. Cancer Res 2024; 84:2792-2805. [PMID: 39228255 DOI: 10.1158/0008-5472.can-23-2846] [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: 09/18/2023] [Revised: 01/23/2024] [Accepted: 06/17/2024] [Indexed: 09/05/2024]
Abstract
Neoantigen-based immunotherapy is an attractive potential treatment for previously intractable tumors. To effectively broaden the application of this approach, stringent biomarkers are crucial to identify responsive patients. ARID1A, a frequently mutated subunit of SWI/SNF chromatin remodeling complex, has been reported to determine tumor immunogenicity in some cohorts; however, mutations and deletions of ARID1A are not always linked to clinical responses to immunotherapy. In this study, we investigated immunotherapeutic responses based on ARID1A status in targeted therapy-resistant cancers. Mouse and human BRAFV600E melanomas with or without ARID1A expression were transformed into resistant to vemurafenib, an FDA-approved specific BRAFV600E inhibitor. Anti-PD-1 antibody treatment enhanced antitumor immune responses in vemurafenib-resistant ARID1A-deficient tumors but not in ARID1A-intact tumors or vemurafenib-sensitive ARID1A-deficient tumors. Neoantigens derived from accumulated somatic mutations during vemurafenib resistance were highly expressed in ARID1A-deficient tumors and promoted tumor immunogenicity. Furthermore, the newly generated neoantigens could be utilized as immunotherapeutic targets by vaccines. Finally, targeted therapy resistance-specific neoantigen in experimental human melanoma cells lacking ARID1A were validated to elicit T-cell receptor responses. Collectively, the classification of ARID1A-mutated tumors based on vemurafenib resistance as an additional indicator of immunotherapy response will enable a more accurate prediction to guide cancer treatment. Furthermore, the neoantigens that emerge with therapy resistance can be promising therapeutic targets for refractory tumors. Significance: Chemotherapy resistance promotes the acquisition of immunogenic neoantigens in ARID1A-deficient tumors that confer sensitivity to immune checkpoint blockade and can be utilized for developing antitumor vaccines, providing strategies to improve immunotherapy efficacy.
Collapse
Affiliation(s)
- Masahiro Okada
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroshi Nakazato
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yuhya Hirahara
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takuya Ishibashi
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masami Kawamura
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- RIKEN Program for Drug Discovery and Medical Technology Platforms, RIKEN, Yokohama, Japan
| |
Collapse
|
39
|
Ooki A, Osumi H, Yoshino K, Yamaguchi K. Potent therapeutic strategy in gastric cancer with microsatellite instability-high and/or deficient mismatch repair. Gastric Cancer 2024; 27:907-931. [PMID: 38922524 PMCID: PMC11335850 DOI: 10.1007/s10120-024-01523-4] [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: 04/17/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
Gastric cancer (GC) is a common malignancy that presents challenges in patient care worldwide. The mismatch repair (MMR) system is a highly conserved DNA repair mechanism that protects genome integrity during replication. Deficient MMR (dMMR) results in an increased accumulation of genetic errors in microsatellite sequences, leading to the development of a microsatellite instability-high (MSI-H) phenotype. Most MSI-H/dMMR GCs arise sporadically, mainly due to MutL homolog 1 (MLH1) epigenetic silencing. Unlike microsatellite-stable (MSS)/proficient MMR (pMMR) GCs, MSI-H/dMMR GCs are relatively rare and represent a distinct subtype with genomic instability, a high somatic mutational burden, favorable immunogenicity, different responses to treatment, and prognosis. dMMR/MSI-H status is a robust predictive biomarker for treatment with immune checkpoint inhibitors (ICIs) due to high neoantigen load, prominent tumor-infiltrating lymphocytes, and programmed cell death ligand 1 (PD-L1) overexpression. However, a subset of MSI-H/dMMR GC patients does not benefit from immunotherapy, highlighting the need for further research into predictive biomarkers and resistance mechanisms. This review provides a comprehensive overview of the clinical, molecular, immunogenic, and therapeutic aspects of MSI-H/dMMR GC, with a focus on the impact of ICIs in immunotherapy and their potential as neoadjuvant therapies. Understanding the complexity and diversity of the molecular and immunological profiles of MSI-H/dMMR GC will drive the development of more effective therapeutic strategies and molecular targets for future precision medicine.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Koichiro Yoshino
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| |
Collapse
|
40
|
Ma Y, Jiang Z, Pan L, Zhou Y, Xia R, Liu Z, Yuan L. Current development of molecular classifications of gastric cancer based on omics (Review). Int J Oncol 2024; 65:89. [PMID: 39092559 DOI: 10.3892/ijo.2024.5677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Gastric cancer (GC) is a complex and heterogeneous disease with significant phenotypic and genetic variation. Traditional classification systems rely mainly on the evaluation of clinical pathological features and conventional biomarkers and might not capture the diverse clinical processes of individual GCs. The latest discoveries in omics technologies such as next‑generation sequencing, proteomics and metabolomics have provided crucial insights into potential genetic alterations and biological events in GC. Clustering strategies for identifying subtypes of GC might offer new tools for improving GC treatment and clinical trial outcomes by enabling the development of therapies tailored to specific subtypes. However, the feasibility and therapeutic significance of implementing molecular classifications of GC in clinical practice need to addressed. The present review examines the current molecular classifications, delineates the prevailing landscape of clinically relevant molecular features, analyzes their correlations with traditional GC classifications, and discusses potential clinical applications.
Collapse
Affiliation(s)
- Yubo Ma
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhengchen Jiang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Libin Pan
- Department of Pharmacy, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310005, P.R. China
| | - Ying Zhou
- Department of Pharmacy, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310005, P.R. China
| | - Ruihong Xia
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhuo Liu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Li Yuan
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
41
|
Ma Y, Shao G, Xie Y, Yang D, Li K, Tan B, Wang C, Sun P, Cao J. Multiomics Analysis of Interleukin-21 as a Potential Immunologic and Biomarker in Hepatocellular Carcinoma. J Immunother 2024; 47:266-274. [PMID: 38832536 DOI: 10.1097/cji.0000000000000526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 04/18/2024] [Indexed: 06/05/2024]
Abstract
Interleukin-21 (IL-21) is an important antitumor cytokine that contributes to the proliferation and differentiation of CD8 + T cells. It has been proven to enhance the response to immune checkpoint inhibitors (ICIs) in various solid tumors. However, its role in hepatocellular carcinoma (HCC) has not yet been clarified. In this research, we aimed to investigate the antitumor effect of IL-21 in HCC and its effect on ICI treatment. Through transcriptome sequencing analysis and immunohistochemistry validation, we found that patients with high IL-21 expression had a better prognosis. HCCs with high expression of IL-21 had higher infiltration of CD8 + T cells, increased expression of immune checkpoints, and an improved response to ICI treatment. In conclusion, IL-21 can enhance the efficacy of ICI treatment and improve the prognosis of patients by promoting the infiltration of CD8 + T cells and the expression of immune checkpoint-related genes.
Collapse
Affiliation(s)
- Yonghui Ma
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Huang YS, Ou YC, Wu CH, Lan J, Huang CC, Fu HC, Huang SW, Huang SY, Wang SC, Lin H. A comparative analysis of MMR immunohistochemistry panels: Evaluating the utility of four-protein versus two-protein panels in endometrial cancer patients. J Formos Med Assoc 2024:S0929-6646(24)00396-6. [PMID: 39183142 DOI: 10.1016/j.jfma.2024.08.028] [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: 03/21/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
AIMS This study aimed to assess the accuracy of a two-protein panel for mismatch repair (MMR) immunohistochemistry (IHC) compared to a four-protein panel in a cohort of endometrial cancer patients. METHODS The study included patients diagnosed with endometrial cancer between January 2018 and December 2023 with patients underwent MMR IHC staining for the four-protein panel (MSH2, MSH6, MLH1, and PMS2) serving as the reference standard. Various combinations of two proteins were examined and evaluated for their accuracy against the four-protein panel. Sensitivity, negative predictive value (NPV), and negative likelihood ratio were calculated for each combination. McNemar's test was performed to assess discordance, and receiver operating characteristic (ROC) curves were generated to evaluate diagnostic accuracy. RESULTS Of 593 patients, MMR deficiency defined as at least one protein loss was observed in 146 patients (24.62%). When compared with four-protein panel, the highest sensitivity was observed with the MSH6/PMS2 combination (99.32%), followed sequentially by MSH6/MLH1 (97.26%), MSH2/PMS2 (93.15%), MSH2/MLH1 (91.10%), MLH1/PMS2 (79.45%), and MSH2/MSH6 (21.92%). The MSH6/PMS2 combination also demonstrated the best NPV of 99.78% and negative likelihood ratio of 0.01, while MSH6/MLH1 showed satisfactory NPV of 99.11% and negative likelihood ratio of 0.03. McNemar's test revealed no statistical difference between the four-protein panel and the MSH6/PMS2 panel (p = 1.000), and the MSH6/MLH1 panel (p = 0.125). CONCLUSIONS The two-protein panel, particularly MSH6/PMS2, offers high sensitivity and negative predictive value, suggesting its potential as a cost-effective alternative to the four-protein panel in MMR testing for endometrial cancer patients.
Collapse
Affiliation(s)
- Yu-Sheng Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Chen-Hsuan Wu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jui Lan
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chao-Cheng Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-Chun Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Szu-Wei Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Szu-Yu Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shao-Chi Wang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
43
|
Fan W, Liu H, Stachelek GC, Begum A, Davis CE, Dorado TE, Ernst G, Reinhold WC, Ozbek B, Zheng Q, De Marzo AM, Rajeshkumar NV, Barrow JC, Laiho M. Ribosomal RNA transcription governs splicing through ribosomal protein RPL22. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608201. [PMID: 39211199 PMCID: PMC11361076 DOI: 10.1101/2024.08.15.608201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Ribosome biosynthesis is a cancer vulnerability executed by targeting RNA polymerase I (Pol I) transcription. We developed advanced, specific Pol I inhibitors to identify drivers of this sensitivity. By integrating multi-omics features and drug sensitivity data from a large cancer cell panel, we discovered that RPL22 frameshift mutation conferred Pol I inhibitor sensitivity in microsatellite instable cancers. Mechanistically, RPL22 directly interacts with 28S rRNA and mRNA splice junctions, functioning as a splicing regulator. RPL22 deficiency, intensified by 28S rRNA sequestration, promoted the splicing of its paralog RPL22L1 and p53 negative regulator MDM4. Chemical and genetic inhibition of rRNA synthesis broadly remodeled mRNA splicing controlling hundreds of targets. Strikingly, RPL22-dependent alternative splicing was reversed by Pol I inhibition revealing a ribotoxic stress-initiated tumor suppressive pathway. We identify a mechanism that robustly connects rRNA synthesis activity to splicing and reveals their coordination by ribosomal protein RPL22.
Collapse
|
44
|
Sun Y, Patterson-Fortin J, Han S, Li Z, Nowicka Z, Hirohashi Y, Kilgas S, Yi JK, Spektor A, Fendler W, Konstantinopoulos PA, Chowdhury D. 53BP1 loss elicits cGAS-STING-dependent antitumor immunity in ovarian and pancreatic cancer. Nat Commun 2024; 15:6676. [PMID: 39107288 PMCID: PMC11303708 DOI: 10.1038/s41467-024-50999-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
53BP1 nucleates the anti-end resection machinery at DNA double-strand breaks, thereby countering BRCA1 activity. Loss of 53BP1 leads to DNA end processing and homologous recombination in BRCA1-deficient cells. Consequently, BRCA1-mutant tumors, typically sensitive to PARP inhibitors (PARPi), become resistant in the absence of 53BP1. Here, we demonstrate that the 'leaky' DNA end resection in the absence of 53BP1 results in increased micronuclei and cytoplasmic double-stranded DNA, leading to activation of the cGAS-STING pathway and pro-inflammatory signaling. This enhances CD8+ T cell infiltration, activates macrophages and natural killer cells, and impedes tumor growth. Loss of 53BP1 correlates with a response to immune checkpoint blockade (ICB) and improved overall survival. Immunohistochemical assessment of 53BP1 in two malignancies, high grade serous ovarian cancer and pancreatic ductal adenocarcinoma, which are refractory to ICBs, reveals that lower 53BP1 levels correlate with an increased adaptive and innate immune response. Finally, BRCA1-deficient tumors that develop resistance to PARPi due to the loss of 53BP1 are susceptible to ICB. Therefore, we conclude that 53BP1 is critical for tumor immunogenicity and underpins the response to ICB. Our results support including 53BP1 expression as an exploratory biomarker in ICB trials for malignancies typically refractory to immunotherapy.
Collapse
MESH Headings
- Tumor Suppressor p53-Binding Protein 1/metabolism
- Tumor Suppressor p53-Binding Protein 1/genetics
- Female
- Nucleotidyltransferases/metabolism
- Nucleotidyltransferases/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Humans
- Animals
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/metabolism
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Mice
- Cell Line, Tumor
- DNA Breaks, Double-Stranded
- BRCA1 Protein/metabolism
- BRCA1 Protein/genetics
- Signal Transduction
- CD8-Positive T-Lymphocytes/immunology
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/pharmacology
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
- Mice, Inbred C57BL
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Mice, Knockout
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Immunity, Innate
Collapse
Affiliation(s)
- Yajie Sun
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jeffrey Patterson-Fortin
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sen Han
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhe Li
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zuzanna Nowicka
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Yuna Hirohashi
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Susan Kilgas
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jae Kyo Yi
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alexander Spektor
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Dipanjan Chowdhury
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
45
|
Liu W, Zhou H, Lai W, Hu C, Xu R, Gu P, Luo M, Zhang R, Li G. The immunosuppressive landscape in tumor microenvironment. Immunol Res 2024; 72:566-582. [PMID: 38691319 DOI: 10.1007/s12026-024-09483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Recent advances in cancer immunotherapy, especially immune checkpoint inhibitors (ICIs), have revolutionized the clinical outcome of many cancer patients. Despite the fact that impressive progress has been made in recent decades, the response rate remains unsatisfactory, and many patients do not benefit from ICIs. Herein, we summarized advanced studies and the latest insights on immune inhibitory factors in the tumor microenvironment. Our in-depth discussion and updated landscape of tumor immunosuppressive microenvironment may provide new strategies for reversing tumor immune evasion, enhancing the efficacy of ICIs therapy, and ultimately achieving a better clinical outcome.
Collapse
Affiliation(s)
- Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Huyue Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rufu Xu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Peng Gu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Menglin Luo
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| |
Collapse
|
46
|
Chang TG, Cao Y, Sfreddo HJ, Dhruba SR, Lee SH, Valero C, Yoo SK, Chowell D, Morris LGT, Ruppin E. LORIS robustly predicts patient outcomes with immune checkpoint blockade therapy using common clinical, pathologic and genomic features. NATURE CANCER 2024; 5:1158-1175. [PMID: 38831056 PMCID: PMC11962634 DOI: 10.1038/s43018-024-00772-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 04/24/2024] [Indexed: 06/05/2024]
Abstract
Despite the revolutionary impact of immune checkpoint blockade (ICB) in cancer treatment, accurately predicting patient responses remains challenging. Here, we analyzed a large dataset of 2,881 ICB-treated and 841 non-ICB-treated patients across 18 solid tumor types, encompassing a wide range of clinical, pathologic and genomic features. We developed a clinical score called LORIS (logistic regression-based immunotherapy-response score) using a six-feature logistic regression model. LORIS outperforms previous signatures in predicting ICB response and identifying responsive patients even with low tumor mutational burden or programmed cell death 1 ligand 1 expression. LORIS consistently predicts patient objective response and short-term and long-term survival across most cancer types. Moreover, LORIS showcases a near-monotonic relationship with ICB response probability and patient survival, enabling precise patient stratification. As an accurate, interpretable method using a few readily measurable features, LORIS may help improve clinical decision-making in precision medicine to maximize patient benefit. LORIS is available as an online tool at https://loris.ccr.cancer.gov/ .
Collapse
Affiliation(s)
- Tian-Gen Chang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yingying Cao
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Hannah J Sfreddo
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Saugato Rahman Dhruba
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Se-Hoon Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Cristina Valero
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Seong-Keun Yoo
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diego Chowell
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luc G T Morris
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
47
|
Li C, Wang B, Tu J, Liu C, Wang Y, Chen J, Huang Y, Liu B, Yuan X. ATM inhibition enhance immunotherapy by activating STING signaling and augmenting MHC Class I. Cell Death Dis 2024; 15:519. [PMID: 39033176 PMCID: PMC11271473 DOI: 10.1038/s41419-024-06911-3] [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: 04/02/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Accumulating evidence supports the concept that DNA damage response targeted therapies can improve antitumor immune response by increasing the immunogenicity of tumor cells and improving the tumor immune microenvironment. Ataxia telangiectasia mutated (ATM) is a core component of the DNA repair system. Although the ATM gene has a significant mutation rate in many human cancers, including colorectal, prostate, lung, and breast, it remains understudied compared with other DDR-involved molecules such as PARP and ATR. Here, we found that either gene knockout or drug intervention, ATM inhibition activated the cGAS/STING pathway and augmented MHC class I in CRC cells, and these effects could be amplified by radiation. Furthermore, we found that MHC class I upregulation induced by ATM inhibition is dependent on the activation of the NFκB/IRF1/NLRC5 pathway and independent of STING. Animal experiments have shown increasing infiltration and cytotoxic function of T cells and better survival in ATM-deficient tumors. This work indicated that ATM nonsense mutation predicted the clinical benefits of radiotherapy combined with immune checkpoint blockade for patients with CRC. It also provides a molecular mechanism rationale for ATM-targeted agents for patients with CRC.
Collapse
Affiliation(s)
- Chunya Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Boyu Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaofan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
48
|
Richardson TE, Walker JM, Hambardzumyan D, Brem S, Hatanpaa KJ, Viapiano MS, Pai B, Umphlett M, Becher OJ, Snuderl M, McBrayer SK, Abdullah KG, Tsankova NM. Genetic and epigenetic instability as an underlying driver of progression and aggressive behavior in IDH-mutant astrocytoma. Acta Neuropathol 2024; 148:5. [PMID: 39012509 PMCID: PMC11252228 DOI: 10.1007/s00401-024-02761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/17/2024]
Abstract
In recent years, the classification of adult-type diffuse gliomas has undergone a revolution, wherein specific molecular features now represent defining diagnostic criteria of IDH-wild-type glioblastomas, IDH-mutant astrocytomas, and IDH-mutant 1p/19q-codeleted oligodendrogliomas. With the introduction of the 2021 WHO CNS classification, additional molecular alterations are now integrated into the grading of these tumors, given equal weight to traditional histologic features. However, there remains a great deal of heterogeneity in patient outcome even within these established tumor subclassifications that is unexplained by currently codified molecular alterations, particularly in the IDH-mutant astrocytoma category. There is also significant intercellular genetic and epigenetic heterogeneity and plasticity with resulting phenotypic heterogeneity, making these tumors remarkably adaptable and robust, and presenting a significant barrier to the design of effective therapeutics. Herein, we review the mechanisms and consequences of genetic and epigenetic instability, including chromosomal instability (CIN), microsatellite instability (MSI)/mismatch repair (MMR) deficits, and epigenetic instability, in the underlying biology, tumorigenesis, and progression of IDH-mutant astrocytomas. We also discuss the contribution of recent high-resolution transcriptomics studies toward defining tumor heterogeneity with single-cell resolution. While intratumoral heterogeneity is a well-known feature of diffuse gliomas, the contribution of these various processes has only recently been considered as a potential driver of tumor aggressiveness. CIN has an independent, adverse effect on patient survival, similar to the effect of histologic grade and homozygous CDKN2A deletion, while MMR mutation is only associated with poor overall survival in univariate analysis but is highly correlated with higher histologic/molecular grade and other aggressive features. These forms of genomic instability, which may significantly affect the natural progression of these tumors, response to therapy, and ultimately clinical outcome for patients, are potentially measurable features which could aid in diagnosis, grading, prognosis, and development of personalized therapeutics.
Collapse
Affiliation(s)
- Timothy E Richardson
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA.
| | - Jamie M Walker
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, NY, 10029, USA
- Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, 10029, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kimmo J Hatanpaa
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Mariano S Viapiano
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Balagopal Pai
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Melissa Umphlett
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
| | - Oren J Becher
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, NY, 10029, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health, New York, NY, 10016, USA
| | - Samuel K McBrayer
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Nadejda M Tsankova
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15.238, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
49
|
Skelly DA, Graham JP, Cheng M, Furuta M, Walter A, Stoklasek TA, Yang H, Stearns TM, Poirion O, Zhang JG, Grassmann JDS, Luo D, Flynn WF, Courtois ET, Chang CH, Serreze DV, Menghi F, Reinholdt LG, Liu ET. Mapping the genetic landscape establishing a tumor immune microenvironment favorable for anti-PD-1 response in mice and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603136. [PMID: 39071392 PMCID: PMC11275897 DOI: 10.1101/2024.07.11.603136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Identifying host genetic factors modulating immune checkpoint inhibitor (ICI) efficacy has been experimentally challenging because of variations in both host and tumor genomes, differences in the microbiome, and patient life exposures. Utilizing the Collaborative Cross (CC) multi-parent mouse genetic resource population, we developed an approach that fixes the tumor genomic configuration while varying host genetics. With this approach, we discovered that response to anti-PD-1 (aPD1) immunotherapy was significantly heritable in four distinct murine tumor models (H2 between 0.18-0.40). For the MC38 colorectal carcinoma system (H2 = 0.40), we mapped four significant ICI response quantitative trait loci (QTL) localized to mouse chromosomes (mChr) 5, 9, 15 and 17, and identified significant epistatic interactions between specific QTL pairs. Differentially expressed genes within these QTL were highly enriched for immune genes and pathways mediating allograft rejection and graft vs host disease. Using a cross species analytical approach, we found a core network of 48 genes within the four QTLs that showed significant prognostic value for overall survival in aPD1 treated human cohorts that outperformed all other existing validated immunotherapy biomarkers, especially in human tumors of the previously defined immune subtype 4. Functional blockade of two top candidate immune targets within the 48 gene network, GM-CSF and high affinity IL-2/IL-15 signaling, completely abrogated the MC38 tumor transcriptional response to aPD1 therapy in vivo. Thus, we have established a powerful cross species in vivo platform capable of uncovering host genetic factors that establish the tumor immune microenvironment configuration propitious for ICI response.
Collapse
Affiliation(s)
- Daniel A. Skelly
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - John P. Graham
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | - Mayuko Furuta
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Andrew Walter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | | | | | - Olivier Poirion
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ji-Gang Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | - Diane Luo
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - William F. Flynn
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Elise T. Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- OB/Gyn Department, UConn Health, Farmington, CT, USA
| | - Chih-Hao Chang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - David V. Serreze
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Edison T. Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| |
Collapse
|
50
|
Shukla S, Patel H, Chen S, Sun R, Wei L, Chen ZS. Dostarlimab in the treatment of mismatch repair deficient recurrent or advanced endometrial cancer. CANCER PATHOGENESIS AND THERAPY 2024; 2:135-141. [PMID: 39027143 PMCID: PMC11252537 DOI: 10.1016/j.cpt.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 07/20/2024]
Abstract
Dostarlimab, a programmed death receptor-1 (PD-1)-blocking IgG4 humanized monoclonal antibody, gained accelerated approval from the US Food and Drug Administration (FDA) in April 2021, and received a full approval in February 2023. Dostarlimab was approved for treating adult patients with mismatch repair deficient (dMMR) recurrent or advanced endometrial cancer (EC) that progressed during or after prior treatment who have no other suitable treatment options. Herein, we review the structure-based mechanism of action of dostarlimab and the results of a clinical study (GARNET; NCT02715284) to comprehensively clarify the efficacy and toxicity of the drug. The efficacy and safety of dostarlimab as monotherapy was assessed in a non-randomized, multicenter, open-label, multi-cohort trial that included 209 patients with dMMR recurrent or advanced solid tumors after receiving systemic therapy. Patients received 500 mg of dostarlimab intravenously every three weeks until they were given four doses. Then, patients received 1000 mg dostarlimab intravenously every six weeks until disease progression or unacceptable toxicity. The overall response rate, as determined by shrinkage in tumor size, was 41.6% (95% confidence interval [CI]; 34.9, 48.6), with 34.7 months as the median response duration. In conclusion, dostarlimab is an immunotherapy-based drug that has shown promising results in adult patients with recurrent or advanced dMMR EC. However, its efficacy in other cancer subtypes, the development of resistance to monotherapy, and efficacy and safety in combination with other immunotherapeutic drugs have not yet been studied.
Collapse
Affiliation(s)
- Siddhant Shukla
- Institute for Biotechnology, St. John's University, New York, New York 11439, United States
| | - Harsh Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, New York 11439, United States
| | - Shuzhen Chen
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 261053, China
| | - Rainie Sun
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, New York 11439, United States
- Stuyvesant High School, New York, New York 10282, United States
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 261053, China
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John's University, New York, New York 11439, United States
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, New York 11439, United States
| |
Collapse
|