1
|
Akand M, Jatsenko T, Muilwijk T, Gevaert T, Joniau S, Van der Aa F. Deciphering the molecular heterogeneity of intermediate- and (very-)high-risk non-muscle-invasive bladder cancer using multi-layered -omics studies. Front Oncol 2024; 14:1424293. [PMID: 39497708 PMCID: PMC11532112 DOI: 10.3389/fonc.2024.1424293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/13/2024] [Indexed: 11/07/2024] Open
Abstract
Bladder cancer (BC) is the most common malignancy of the urinary tract. About 75% of all BC patients present with non-muscle-invasive BC (NMIBC), of which up to 70% will recur, and 15% will progress in stage and grade. As the recurrence and progression rates of NMIBC are strongly associated with some clinical and pathological factors, several risk stratification models have been developed to individually predict the short- and long-term risks of disease recurrence and progression. The NMIBC patients are stratified into four risk groups as low-, intermediate-, high-risk, and very high-risk by the European Association of Urology (EAU). Significant heterogeneity in terms of oncological outcomes and prognosis has been observed among NMIBC patients within the same EAU risk group, which has been partly attributed to the intrinsic heterogeneity of BC at the molecular level. Currently, we have a poor understanding of how to distinguish intermediate- and (very-)high-risk NMIBC with poor outcomes from those with a more benign disease course and lack predictive/prognostic tools that can specifically stratify them according to their pathologic and molecular properties. There is an unmet need for developing a more accurate scoring system that considers the treatment they receive after TURBT to enable their better stratification for further follow-up regimens and treatment selection, based also on a better response prediction to the treatment. Based on these facts, by employing a multi-layered -omics (namely, genomics, epigenetics, transcriptomics, proteomics, lipidomics, metabolomics) and immunohistopathology approach, we hypothesize to decipher molecular heterogeneity of intermediate- and (very-)high-risk NMIBC and to better stratify the patients with this disease. A combination of different -omics will provide a more detailed and multi-dimensional characterization of the tumor and represent the broad spectrum of NMIBC phenotypes, which will help to decipher the molecular heterogeneity of intermediate- and (very-)high-risk NMIBC. We think that this combinatorial multi-omics approach has the potential to improve the prediction of recurrence and progression with higher precision and to develop a molecular feature-based algorithm for stratifying the patients properly and guiding their therapeutic interventions in a personalized manner.
Collapse
Affiliation(s)
- Murat Akand
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Tatjana Jatsenko
- Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Tim Muilwijk
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Frank Van der Aa
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Kögl J, Pan TL, Marth C, Zeimet AG. The game-changing impact of POLE mutations in oncology-a review from a gynecologic oncology perspective. Front Oncol 2024; 14:1369189. [PMID: 39239272 PMCID: PMC11374733 DOI: 10.3389/fonc.2024.1369189] [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: 01/11/2024] [Accepted: 07/31/2024] [Indexed: 09/07/2024] Open
Abstract
Somatic mutations within the exonuclease proofreading domain (EDM) of the DNA polymerase Pol ϵ (POLE) gene are increasingly being discovered in ovarian, colorectal, urological, and, especially, endometrial carcinoma (EC), where these are found in up to 10% of the cases. In EC, there are five confirmed pathogenic somatic POLE-EDM mutations that are located at codons 286, 411, 297, 456, and 459, and these are called "hotspot" mutations. POLE mutant tumors are ultramutated entities with a frequency of base substitution mutations that is among the highest in human tumors. Interestingly, these mutations are associated with excellent clinical outcome in EC. An additional six "non-hotspot" POLE-EDM EC mutations are also considered pathogenic, and they also confer a favorable prognosis. Currently, de-escalation of adjuvant treatment is recommended for patients with EC with stage I-II tumors involving any of these 11 EDM mutations, even in patients with other clinicopathological risk factors. The high tumor mutational burden and the consequent increased infiltration of immune cells due to the overexpression of different neoantigens are probably responsible for the improved prognosis. Ongoing studies are examining POLE hotspot mutations among many non-gynecologic tumors, although the impact of such mutations on clinical outcomes is still a topic of debate. Therapeutic modalities for these hypermutated tumors are also an important consideration, including the need for or de-escalation of adjuvant treatments and the response to immune therapy. This review addresses the critical role of POLE mutations in gynecologic oncology and oncology in general, focusing on definitions, variants, underlying pathogenic mechanisms, upcoming developments in the field, and the clinic behavior associated with such mutations.
Collapse
Affiliation(s)
- Johanna Kögl
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Teresa L Pan
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Marth
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Alain G Zeimet
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
3
|
Bakaloudi DR, Talukder R, Makrakis D, Diamantopoulos L, Enright T, Leary JB, Patgunarajah U, Thomas VM, Swami U, Agarwal N, Jindal T, Koshkin VS, Brown JR, Barata P, Murgić J, Miletić M, Johnson J, Zakharia Y, Hui G, Drakaki A, Duran I, Buznego LA, Barrera RM, Castañeda DM, Rey-Cárdenas M, Castellano D, Nguyen CB, Park JJ, Alva A, McKay RR, Stewart TF, Epstein IB, Bellmunt J, Wright JL, Gupta S, Grivas P, Khaki AR. Association of Tumor Mutational Burden and Microsatellite Instability With Response and Outcomes in Patients With Urothelial Carcinoma Treated With Immune Checkpoint Inhibitor. Clin Genitourin Cancer 2024; 22:102198. [PMID: 39241315 DOI: 10.1016/j.clgc.2024.102198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Microsatellite Instability (MSI) and Tumor Mutational Burden (TMB) are associated with immune checkpoint inhibitor (ICI) efficacy. We examined the association between TMB and MSI status with survival in patients with urothelial carcinoma (UC) treated with ICI. METHODS Patients from 15 institutions were treated with ICI monotherapy. Primary endpoint was overall survival and secondary endpoints included observed response rate (ORR), and progression-free (PFS) calculated from ICI initiation. TMB was analyzed as dichotomous (≥10 vs. <10 mut/Mb) and continuous variable. RESULTS We identified 411 patients: 203 were treated with ICI 1L/upfront; 104 with 2 + L. For the 1L/upfront: median [m] OS was numerically longer in patients with TMB ≥10 versus TMB <10: mOS 35 versus 26 months (HR = 0.6) and with MSI-H and MSI-S (mOS NR vs. 22 months), though neither association was statistically significant. A statistically significant association was found between TMB (continuous variable) and OS (HR = 0.96, P = .01). For 2 + L: mOS was numerically longer in patients with TMB ≥10 versus TMB <10: (20 vs. 12 months; HR = 0.9); mOS was 12 and 17 months for patients with MSI-H and MSI-S, respectively. Eighty-nine patients received maintenance avelumab (mAV): mOS was longer in patients with TMB ≥10 versus TMB <10: 61 versus 17 months; (HR = 0.2, P = .02) and with MSI-H and MSI-S (NR vs. 24 months). CONCLUSIONS Although not reaching statistical significance in several subsets, patients with high TMB and MSI-H had numerically longer OS with ICI, especially with mAV. Further validation is needed.
Collapse
Affiliation(s)
| | - Rafee Talukder
- Department of Medicine, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
| | - Dimitrios Makrakis
- Department of Medicine, Jacobi Medical Center-Albert Einstein College of Medicine, Bronx, NY
| | | | - Thomas Enright
- Department of Medicine, University of Washington, Seattle, WA
| | - Jacob B Leary
- Department of Medicine, University of Washington, Seattle, WA
| | - Ubenthira Patgunarajah
- Department of Hematology and Oncology, Taussig cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Vinay M Thomas
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Umang Swami
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Neeraj Agarwal
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Tanya Jindal
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Helen Diller Family Cancer Center, San Francisco, CA
| | - Vadim S Koshkin
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Helen Diller Family Cancer Center, San Francisco, CA
| | - Jason R Brown
- Division of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Pedro Barata
- Division of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Jure Murgić
- Department of Oncology and Nuclear Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia; Catholic University of Croatia School of Medicine, Zagreb, Croatia
| | - Marija Miletić
- Department of Oncology and Nuclear Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Jeffrey Johnson
- Division of Oncology, Department of Medicine, University of Iowa, Iowa City, IA
| | - Yousef Zakharia
- Division of Oncology, Department of Medicine, University of Iowa, Iowa City, IA
| | - Gavin Hui
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Alexandra Drakaki
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Ignacio Duran
- Department of Oncology, University Hospital Marqués of Valdecilla, Cantabria, Spain
| | - Lucia A Buznego
- Department of Oncology, University Hospital Marqués of Valdecilla, Cantabria, Spain
| | - Rafael M Barrera
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David M Castañeda
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Charles B Nguyen
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Joseph J Park
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Ajjai Alva
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Tyler F Stewart
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Ilana B Epstein
- Dana-Farber Cancer Institute/Brigham and Women's Hospital Harvard Medical School, Boston, MA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute/Brigham and Women's Hospital Harvard Medical School, Boston, MA
| | | | - Shilpa Gupta
- Department of Hematology and Oncology, Taussig cancer Institute, Cleveland Clinic Foundation, Cleveland, OH.
| | - Petros Grivas
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA.
| | - Ali Raza Khaki
- Division of Oncology, Department of Medicine, Stanford University, Palo Alto, CA.
| |
Collapse
|
4
|
Thomsen LCV, Kleinmanns K, Anandan S, Gullaksen SE, Abdelaal T, Iversen GA, Akslen LA, McCormack E, Bjørge L. Combining Mass Cytometry Data by CyTOFmerge Reveals Additional Cell Phenotypes in the Heterogeneous Ovarian Cancer Tumor Microenvironment: A Pilot Study. Cancers (Basel) 2023; 15:5106. [PMID: 37894472 PMCID: PMC10605295 DOI: 10.3390/cancers15205106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
The prognosis of high-grade serous ovarian carcinoma (HGSOC) is poor, and treatment selection is challenging. A heterogeneous tumor microenvironment (TME) characterizes HGSOC and influences tumor growth, progression, and therapy response. Better characterization with multidimensional approaches for simultaneous identification and categorization of the various cell populations is needed to map the TME complexity. While mass cytometry allows the simultaneous detection of around 40 proteins, the CyTOFmerge MATLAB algorithm integrates data sets and extends the phenotyping. This pilot study explored the potential of combining two datasets for improved TME phenotyping by profiling single-cell suspensions from ten chemo-naïve HGSOC tumors by mass cytometry. A 35-marker pan-tumor dataset and a 34-marker pan-immune dataset were analyzed separately and combined with the CyTOFmerge, merging 18 shared markers. While the merged analysis confirmed heterogeneity across patients, it also identified a main tumor cell subset, additionally to the nine identified by the pan-tumor panel. Furthermore, the expression of traditional immune cell markers on tumor and stromal cells was revealed, as were marker combinations that have rarely been examined on individual cells. This study demonstrates the potential of merging mass cytometry data to generate new hypotheses on tumor biology and predictive biomarker research in HGSOC that could improve treatment effectiveness.
Collapse
Affiliation(s)
- Liv Cecilie Vestrheim Thomsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021 Bergen, Norway
- Norwegian Institute of Public Health, 5015 Bergen, Norway
| | - Katrin Kleinmanns
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Shamundeeswari Anandan
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Stein-Erik Gullaksen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Tamim Abdelaal
- Delft Bioinformatics Laboratory, Delft University of Technology, 2628XE Delft, The Netherlands
- Department of Radiology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
| | - Grete Alrek Iversen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Lars Andreas Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway
- Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Emmet McCormack
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Line Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
5
|
Ma X, Zhao Y, Shi C, Jiang H, Liu H, Wang H, Qin X, Wang Y, Han Z. Systematic pan-cancer analysis identified neuropilin 1 as an immunological and prognostic biomarker. Cell Biochem Funct 2023; 41:658-675. [PMID: 37306257 DOI: 10.1002/cbf.3821] [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: 04/14/2023] [Revised: 05/14/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Neuropilin 1 (NRP1) is a transmembrane glycoprotein, nontyrosine kinase receptor that plays an important role in axonal growth and angiogenesis in the nervous system. Although currently more and more studies have shown that NRP1 plays an important role in some cancers, no systematic pan-cancer analysis of NRP-1 has been performed to date. Therefore, we aimed to investigate the associated immune function and prognostic value of NRP1 in 33 tumors of various cancer types. In this study, based on The Cancer Genome Atlas, Cancer Cell Line Encyclopedia, Genotype Tissue Expression, cBioportal for cancer genomics, and Human Protein Atlas (HPA databases), various bioinformatics analysis methods were used to investigate the potential carcinogenic effects of NRP1 activation, pan-cancer analysis of NRP1 expression, and the relationship between NRP1 expression and prognosis indicators including overall survival, disease-specific survival, disease-free interval, and progression-free interval, tumor mutational burden (TMB), and microsatellite instability (MSI). The results showed that NRP1 was highly expressed in most tumors. In addition, NRP1 was found to be positively or negatively correlated with the prognosis of different tumors. Also, the expression of NRP1 was associated with TMB and MSI in in 27 and 21 different types of tumors, respectively, and with DNA methylation in almost all the various types of tumors. The expression of the NRP1 gene was negatively correlated with the infiltration levels of most immune cells. In addition, the correlation between the level of immune cell infiltration and NRP1 expression varied according to immune cell subtype. Our study suggests that NRP1 plays an important role in tumor development and tumor immunity and could potentially be used as a prognostic indicator in a variety of malignancies.
Collapse
Affiliation(s)
- Xiao Ma
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Zhao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Congcong Shi
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong Jiang
- Department of Oncology, Jiawang People's Hospital, Xuzhou, Jiangsu, China
| | - Haonan Liu
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongmei Wang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaobing Qin
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuqin Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhengxiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
6
|
Bahlinger V, Hartmann A, Eckstein M. Immunotherapy in Genitourinary Cancers: Role of Surgical Pathologist for Detection of Immunooncologic Predictive Factors. Adv Anat Pathol 2023; 30:203-210. [PMID: 36730368 PMCID: PMC10082065 DOI: 10.1097/pap.0000000000000383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Genitourinary malignancies include a broad spectrum of distinct tumor entities occurring in the kidney, the urinary tract, the prostate, the adrenal glands, the penis, and testicles. Each tumor entity presents with unique biological characteristics, especially in terms of immunobiology. The immune landscape of genitourinary malignancies differs between immunoreactive tumors like urothelial carcinoma or carcinomas of the kidney, for which several immunotherapeutic treatment options have been approved in the past years. In contrast, prostate cancer presents with low immunogenicity and previous trials exploring immune checkpoint inhibitors and other immunotherapeutic agents did not proof substantial survival benefits. In this review, we are presenting a streamlined overview on the role of surgical pathologists within the contemporary practice of immune oncology. It includes current indications for pathologic programmed death-ligand 1 (PD-L1) assessment and important pathologic considerations on PD-L1 testing harmonization including interassay and algorithm variabilities. In addition, we will discuss emerging biomarkers beyond PD-L1 and their potential to predict immunotherapy responses including tumor mutational burden, microsatellite instability, gene expression signatures, and histologic factors.
Collapse
Affiliation(s)
- Veronika Bahlinger
- Institute of Pathology, University Hospital Erlangen-Nürnberg, Friedrich-Alexander-Universität Erlangen-Nürnberg
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen-Nürnberg, Friedrich-Alexander-Universität Erlangen-Nürnberg
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen-Nürnberg, Friedrich-Alexander-Universität Erlangen-Nürnberg
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
7
|
Zhang X, Ji H, Huang Y, Zhu B, Xing Q. Elevated PTTG1 predicts poor prognosis in kidney renal clear cell carcinoma and correlates with immunity. Heliyon 2023; 9:e13201. [PMID: 36793955 PMCID: PMC9922818 DOI: 10.1016/j.heliyon.2023.e13201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Background PTTG1 has been reported to be linked with the prognosis and progression of various cancers, including kidney renal clear cell carcinoma (KIRC). In this article, we mainly investigated the associations between prognosis, immunity, and PTTG1 in KIRC patients. Method We downloaded transcriptome data from the TCGA-KIRC database. PCR and immunohistochemistry were used, respectively, to validate the expression of PTTG1 in KIRC at the cell line and the protein levels. Survival analyses as well as univariate or multivariate Cox hazard regression analyses were used to prove whether PTTG1 alone could affect the prognosis of KIRC. The most important point was to study the relationship between PTTG1 and immunity. Results The results of the paper revealed that the expression levels of PTTG1 were elevated in KIRC compared with para-cancerous normal tissues, validated by PCR and immunohistochemistry at the cell line and the protein levels (P < 0.05). High PTTG1 expression was related to shorter overall survival (OS) in patients with KIRC (P < 0.05). Through univariate or multivariate regression analysis, PTTG1 was confirmed to be an independent prognostic factor for OS of KIRC (P < 0.05), and its related seven pathways were obtained through gene set enrichment analysis (GSEA; P < 0.05). Moreover, tumor mutational burden (TMB) and immunity were found to be significantly connected with PTTG1 in KIRC (P < 0.05). Correlations between PTTG1 and immunotherapy responses implied that the low-PTTG1 group was more sensitive to immunotherapy (P < 0.05). Conclusions PTTG1 was closely associated with TMB or immunity, and it had a superior ability to forecast the prognosis of KIRC patients.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Hao Ji
- Department of Urology, Tumor Hospital Affiliated to Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Yeqing Huang
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Bingye Zhu
- Department of Urology, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, 226001, Jiangsu Province, China,Corresponding author. Department of Urology, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), No. 881 Yonghe Road, Nantong, 226001, Jiangsu Province, China.
| | - Qianwei Xing
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China,Corresponding author. Department of Urology, Affiliated Hospital of Nantong University, No.20 West Temple Road, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
8
|
Wu C, Zhong R, Sun X, Shi J. PSME2 identifies immune-hot tumors in breast cancer and associates with well therapeutic response to immunotherapy. Front Genet 2022; 13:1071270. [PMID: 36583022 PMCID: PMC9793949 DOI: 10.3389/fgene.2022.1071270] [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: 10/16/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BrCa) is a heterogeneous disease, which leads to unsatisfactory prognosis in females worldwide. Previous studies have proved that tumor immune microenvironment (TIME) plays crucial roles in oncogenesis, progression, and therapeutic resistance in Breast cancer. However, biomarkers related to TIME features have not been fully discovered. Proteasome activator complex subunit 2 (PSME2) is a member of proteasome activator subunit gene family, which is critical to protein degradation mediated by the proteasome. In the current research, we comprehensively analyzed the expression and immuno-correlations of Proteasome activator complex subunit 2 in Breast cancer. Proteasome activator complex subunit 2 was significantly upregulated in tumor tissues but associated with well prognosis. In addition, Proteasome activator complex subunit 2 was overexpressed in HER2-positive Breast cancer but not related to other clinicopathological features. Interestingly, Proteasome activator complex subunit 2 was positively related to immune-related processes and identified immuno-hot TIME in Breast cancer. Specifically, Proteasome activator complex subunit 2 was positively correlated with immunomodulators, tumor-infiltrating immune cells (TIICs), immune checkpoints, and tumor mutation burden (TMB) levels. Moreover, the positive correlation between Proteasome activator complex subunit 2 and PD-L1 expression was confirmed in a tissue microarray (TMA) cohort. Furthermore, in an immunotherapy cohort of Breast cancer, patients with pathological complete response (pCR) expressed higher Proteasome activator complex subunit 2 compared with those with non-pathological complete response. In conclusion, Proteasome activator complex subunit 2 is upregulated in tumor tissues and correlated with the immuno-hot tumor immune microenvironment, which can be a novel biomarker for the recognition of tumor immune microenvironment features and immunotherapeutic response in Breast cancer.
Collapse
Affiliation(s)
- Cen Wu
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Ren Zhong
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Xiaofei Sun
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Jiajie Shi
- Departments of Breast Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
9
|
Sevillano Fernández E, Madurga de Lacalle R, Rodriguez Moreno JF, Barquín García A, Yagüe Fernández M, Navarro Alcaraz P, Barba Llacer M, Quiralte Pulido M, García-Donás Jiménez J. Prognostic Value and Clinical Significance of FGFR Genomic Alterations (GAs) in Metastatic Urothelial Cancer Patients. J Clin Med 2022; 11:jcm11154483. [PMID: 35956100 PMCID: PMC9369263 DOI: 10.3390/jcm11154483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Fibroblast growth factor receptor (FGFR) genomic alterations (GAs) represent an actionable target, key to the pathogenesis of some urothelial cancers (UCs). Though FGFR GAs are common in noninvasive UC, little is known about their role in the metastatic(m) setting and response to therapy. This study aimed to assess the impact of FGFR alterations on sensitivity to systemic treatments and survival and to validate Bajorin’s and Bellmunt’s prognostic scores in mUC patients according to their FGFR status. We retrospectively analyzed data from 98 patients with tumor-sequenced UC who received treatment between January 2010 and December 2020. Up to 77 developed metastatic disease and were deemed the study population. Twenty-six showed FGFR GAs. A trend toward a better response to cisplatin and checkpoint inhibitors was suggested favoring FGFR GA tumors. FGFR GA patients who received an FGFR inhibitor as first-line had poorer responses compared with other options (20% vs. 68.4%, p = 0.0065). Median PFS was 6 vs. 5 months in the FGFR GA vs. FGFR WT cohort (p = 0.71). Median OS was significantly worse in the FGFR GA vs. FGFR WT cohort (16.2 vs. 31.9 months, p = 0.045). Multivariate analyses deemed FGFR GAs as a factor independently associated with the outcome (HR 2.59 (95% CI 1.21–5.55)). Bajorin’s model correctly predicted clinical outcomes in the whole study population but not in FGFR GA cases. FGFR GAs are a relevant biomarker in mUC that could condition the response to systemic therapy. New prognostic models, including this molecular determination, should be designed and validated.
Collapse
Affiliation(s)
- Elena Sevillano Fernández
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
- Departamento de Oncología Médica, Hospital Sanchinarro, Universidad San Pablo-CEU, CEU Universities, 28003 Madrid, Spain
- Correspondence:
| | | | - Juan Francisco Rodriguez Moreno
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
| | - Arantzazu Barquín García
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
| | - Mónica Yagüe Fernández
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
| | - Paloma Navarro Alcaraz
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
| | - María Barba Llacer
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
| | - Miguel Quiralte Pulido
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
| | - Jesús García-Donás Jiménez
- HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain; (J.F.R.M.); (A.B.G.); (M.Y.F.); (P.N.A.); (M.B.L.); (M.Q.P.); (J.G.-D.J.)
- Departamento de Oncología Médica, Hospital Sanchinarro, Universidad San Pablo-CEU, CEU Universities, 28003 Madrid, Spain
| |
Collapse
|
10
|
Wang X, Chen H. Prognosis Prediction Through an Integrated Analysis of Single-Cell and Bulk RNA-Sequencing Data in Triple-Negative Breast Cancer. Front Genet 2022; 13:928175. [PMID: 35846145 PMCID: PMC9283578 DOI: 10.3389/fgene.2022.928175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Genomic and antigenic heterogeneity pose challenges in the precise assessment of outcomes of triple-negative breast cancer (TNBC) patients. Thus, this study was designed to investigate the cardinal genes related to cell differentiation and tumor malignant grade to advance the prognosis prediction in TNBC patients through an integrated analysis of single-cell and bulk RNA-sequencing (RNA-seq) data. Methods: We collected RNA-seq and microarray data of TNBC from two public datasets. Using single-cell pseudotime analysis, differentially expressed genes (DEGs) among trajectories from 1534 cells of 6 TNBC patients were identified as the potential genes crucial for cell differentiation. Furthermore, the grade- and tumor mutational burden (TMB)-related DEGs were explored via a weighted correlation network analysis using the Molecular Taxonomy of Breast Cancer International Consortium dataset. Subsequently, we utilized the DEGs to construct a prognostic signature, which was validated using another independent dataset. Moreover, as gene set variation analysis indicated the differences in immune-related pathways between different risk groups, we explored the immune differences between the two groups. Results: A signature including 10 genes related to grade and TMB was developed to assess the outcomes of TNBC patients, and its prognostic efficacy was prominent in two cohorts. The low-risk group generally harbored lower immune infiltration compared to the high-risk group. Conclusion: Cell differentiation and grade- and TMB-related DEGs were identified using single-cell and bulk RNA-seq data. A 10-gene signature for prognosis prediction in TNBC patients was constructed, and its performance was excellent. Interestingly, the signature was found to be closely related to tumor immune infiltration, which might provide evidence for the crucial roles of immune cells in malignant initiation and progression in TNBC.
Collapse
Affiliation(s)
- Xiangru Wang
- Department of General Surgery, The Affiliated Hospital Of Henan Medical College, Henan Medical College Hospital Workers, Zhengzhou, China
- *Correspondence: Xiangru Wang
| | | |
Collapse
|
11
|
Zhang S, Xiao X, Wang Y, Song T, Li C, Bao H, Liu Q, Sun G, Sun X, Su T, Fu T, Wang Y, Liang P. Developing an Immune-Related Signature for Predicting Survival Rate and the Response to Immune Checkpoint Inhibitors in Patients With Glioma. Front Genet 2022; 13:899125. [PMID: 35719378 PMCID: PMC9204856 DOI: 10.3389/fgene.2022.899125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Glioma is one of the most aggressive cancer types affecting the central nerve system, with poor overall survival (OS) rates. The present study aimed to construct a novel immune-related signature to predict prognosis and the efficiency of immunotherapy in patients with glioma.Methods: The mRNA expression data and other clinical information of patients with glioblastoma multiforme (GBM) and low grade glioma (LGG) were obtained from The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. The immune-related genes were obtained from the Immunology Database and Analysis Portal database. Subsequently, an immune-related signature was created following the results obtained from the Least Absolute Shrinkage and Selection Operator regression model. To validate the predictability of the signature, Kaplan-Meier survival curves and time-dependent receiver operating characteristic curves were created. Moreover, both univariate and multivariate analyses were carried out using the OS between this signature and other clinicopathologic factors, and a nomogram was constructed. In addition, the association between signature, immune cell infiltration, tumor mutation burden and immunophenoscore were determined.Results: Results of the present study using 118 GBM and LGG samples uncovered 15 immune-related genes that were also differently expressed in glioma samples. These were subsequently used to construct the immune-related signature. This signature exhibits the ability to predict prognosis, the infiltration of immune cells in the tumor microenvironment and the response of patients with glioma to immunotherapy.Conclusion: Results of the present study demonstrated that the aforementioned novel immune-related signature may accurately predict prognosis and the response of patients with glioma to immunotherapy.
Collapse
Affiliation(s)
- Sibin Zhang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xu Xiao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Wang
- Department of Esophageal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianjun Song
- Department of Medicine II, University Hospital LMU Munich, Munich, Germany
| | - Chenlong Li
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongbo Bao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qing Liu
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guiyin Sun
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaoyang Sun
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianqi Su
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianjiao Fu
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yujie Wang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Peng Liang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Peng Liang,
| |
Collapse
|
12
|
Zhuang J, Chen Z, Chen Z, Chen J, Liu M, Xu X, Liu Y, Yang S, Hu Z, He F. Construction of an immune-related lncRNA signature pair for predicting oncologic outcomes and the sensitivity of immunosuppressor in treatment of lung adenocarcinoma. Respir Res 2022; 23:123. [PMID: 35562727 PMCID: PMC9101821 DOI: 10.1186/s12931-022-02043-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 05/04/2022] [Indexed: 12/22/2022] Open
Abstract
Background Although immunotherapy has shown clinical activity in lung adenocarcinoma (LUAD), LUAD prognosis has been a perplexing problem. We aimed to construct an immune-related lncRNA pairs (IRLPs) score for LUAD and identify what immunosuppressor are appropriate for which group of people with LUAD. Methods Based on The Cancer Genome Atlas (TCGA)-LUAD cohort, IRLPs were identified to construct an IRLPs scoring system by Cox regression and validated in the Gene Expression Omnibus (GEO) dataset using log-rank test and the receiver operating characteristic curve (ROC). Next, we used spearman’s correlation analysis, t-test, signaling pathways analysis and gene mutation analysis to explore immune and molecular characteristics in different IRLP subgroups. The “pRRophetic” package was used to predict the sensitivity of immunosuppressant. Results The IRLPs score was constructed based on eight IRLPs calculated as 2.12 × (MIR31HG|RRN3P2) + 0.43 × (NKX2-1-AS1|AC083949.1) + 1.79 × (TMPO-AS1|LPP-AS2) + 1.60 × (TMPO-AS1|MGC32805) + 1.79 × (TMPO-AS1|PINK1-AS) + 0.65 × (SH3BP5-AS1|LINC01137) + 0.51 × (LINC01004|SH3PXD2A-AS1) + 0.62 × (LINC00339|AGAP2-AS1). Patients with a lower IRLPs risk score had a better overall survival (OS) (Log-rank test PTCGA train dataset < 0.001, PTCGA test dataset = 0.017, PGEO dataset = 0.027) and similar results were observed in the AUCs of TCGA dataset and GEO dataset (AUC TCGA train dataset = 0.777, AUC TCGA test dataset = 0.685, AUC TCGA total dataset = 0.733, AUC GEO dataset = 0.680). Immune score (Cor = -0.18893, P < 0.001), stoma score (Cor = -0.24804, P < 0.001), and microenvironment score (Cor = -0.22338, P < 0.001) were significantly decreased in the patients with the higher IRLP risk score. The gene set enrichment analysis found that high-risk group enriched in molecular changes in DNA and chromosomes signaling pathways, and in this group the tumor mutation burden (TMB) was higher than in the low-risk group (P = 0.0015). Immunosuppressor methotrexate sensitivity was higher in the high-risk group (P = 0.0052), whereas parthenolide (P < 0.001) and rapamycin (P = 0.013) sensitivity were lower in the high-risk group. Conclusions Our study established an IRLPs scoring system as a biomarker to help in the prognosis, the identification of molecular and immune characteristics, and the patient-tailored selection of the most suitable immunosuppressor for LUAD therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02043-4.
Collapse
Affiliation(s)
- Jinman Zhuang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Digital Tumor Data Research Center, Fuzhou, China
| | - Zhongwu Chen
- Department of Interventional Therapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zishan Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Digital Tumor Data Research Center, Fuzhou, China
| | - Jin Chen
- Department of Interventional Therapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Maolin Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Digital Tumor Data Research Center, Fuzhou, China
| | - Xinying Xu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Digital Tumor Data Research Center, Fuzhou, China
| | - Yuhang Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Digital Tumor Data Research Center, Fuzhou, China
| | - Shuyan Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Digital Tumor Data Research Center, Fuzhou, China
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China. .,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China. .,Fujian Digital Tumor Data Research Center, Fuzhou, China.
| | - Fei He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China. .,Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China. .,Fujian Digital Tumor Data Research Center, Fuzhou, China.
| |
Collapse
|
13
|
Powles T, Sridhar SS, Loriot Y, Bellmunt J, Mu XJ, Ching KA, Pu J, Sternberg CN, Petrylak DP, Tambaro R, Dourthe LM, Alvarez-Fernandez C, Aarts M, di Pietro A, Grivas P, Davis CB. Avelumab maintenance in advanced urothelial carcinoma: biomarker analysis of the phase 3 JAVELIN Bladder 100 trial. Nat Med 2021; 27:2200-2211. [PMID: 34893775 DOI: 10.1038/s41591-021-01579-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 10/13/2021] [Indexed: 12/20/2022]
Abstract
In a recent phase 3 randomized trial of 700 patients with advanced urothelial cancer (JAVELIN Bladder 100; NCT02603432 ), avelumab/best supportive care (BSC) significantly prolonged overall survival relative to BSC alone as maintenance therapy after first-line chemotherapy. Exploratory biomarker analyses were performed to identify biological pathways that might affect survival benefit. Tumor molecular profiling by immunohistochemistry, whole-exome sequencing and whole-transcriptome sequencing revealed that avelumab survival benefit was positively associated with PD-L1 expression by tumor cells, tumor mutational burden, APOBEC mutation signatures, expression of genes underlying innate and adaptive immune activity and the number of alleles encoding high-affinity variants of activating Fcγ receptors. Pathways connected to tissue growth and angiogenesis might have been associated with reduced survival benefit. Individual biomarkers did not comprehensively identify patients who could benefit from therapy; however, multi-parameter models incorporating genomic alteration, immune responses and tumor growth showed promising predictive utility. These results characterize the complex biologic pathways underlying survival benefit from immune checkpoint inhibition in advanced urothelial cancer and suggest that multiple biomarkers might be needed to identify patients who would benefit from treatment.
Collapse
Affiliation(s)
- Thomas Powles
- Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St. Bartholomew's Hospital, London, UK.
| | - Srikala S Sridhar
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yohann Loriot
- Gustave Roussy, INSERMU981, Université Paris-Saclay, Villejuif, France
| | - Joaquim Bellmunt
- Department of Medical Oncology, Beth Israel Deaconess Medical Center and IMIM-PSMAR Lab, Harvard Medical School, Boston, MA, USA
| | - Xinmeng Jasmine Mu
- Computational Biology, Oncology Research and Development, Pfizer, La Jolla, CA, USA
| | - Keith A Ching
- Computational Biology, Oncology Research and Development, Pfizer, La Jolla, CA, USA
| | - Jie Pu
- Statistics, Global Biometrics and Data Management, Pfizer, La Jolla, CA, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Hematology/Oncology, Meyer Cancer Center, New York, NY, USA
| | | | - Rosa Tambaro
- Istituto Nazionale per lo Studio e la Cura dei Tumori, IRCCS Fondazione Giovanni Pascale, Naples, Italy
| | - Louis M Dourthe
- Service d'Oncologie Médicale, Clinique St Anne, Strasbourg, France
| | - Carlos Alvarez-Fernandez
- Department of Medical Oncology, Hospital Universitario Central de Asturias. Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Maureen Aarts
- Department of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Petros Grivas
- Department of Medicine, Division of Medical Oncology, University of Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA
| | | |
Collapse
|
14
|
Ahluwalia P, Ahluwalia M, Mondal AK, Sahajpal NS, Kota V, Rojiani MV, Kolhe R. Natural Killer Cells and Dendritic Cells: Expanding Clinical Relevance in the Non-Small Cell Lung Cancer (NSCLC) Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13164037. [PMID: 34439191 PMCID: PMC8394984 DOI: 10.3390/cancers13164037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a major subtype of lung cancer that accounts for almost 85% of lung cancer cases worldwide. Although recent advances in chemotherapy, radiotherapy, and immunotherapy have helped in the clinical management of these patients, the survival rate in advanced stages remains dismal. Furthermore, there is a critical lack of accurate prognostic and stratification markers for emerging immunotherapies. To harness immune response modalities for therapeutic benefits, a detailed understanding of the immune cells in the complex tumor microenvironment (TME) is required. Among the diverse immune cells, natural killer (NK cells) and dendritic cells (DCs) have generated tremendous interest in the scientific community. NK cells play a critical role in tumor immunosurveillance by directly killing malignant cells. DCs link innate and adaptive immune systems by cross-presenting the antigens to T cells. The presence of an immunosuppressive milieu in tumors can lead to inactivation and poor functioning of NK cells and DCs, which results in an adverse outcome for many cancer patients, including those with NSCLC. Recently, clinical intervention using modified NK cells and DCs have shown encouraging response in advanced NSCLC patients. Herein, we will discuss prognostic and predictive aspects of NK cells and DC cells with an emphasis on NSCLC. Additionally, the discussion will extend to potential strategies that seek to enhance the anti-tumor functionality of NK cells and DCs.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (P.A.); (A.K.M.); (N.S.S.)
| | - Meenakshi Ahluwalia
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (P.A.); (A.K.M.); (N.S.S.)
| | - Nikhil S. Sahajpal
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (P.A.); (A.K.M.); (N.S.S.)
| | - Vamsi Kota
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Mumtaz V. Rojiani
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, USA;
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (P.A.); (A.K.M.); (N.S.S.)
- Correspondence: ; Tel.: +1-706-721-2771; Fax: +1-706-434-6053
| |
Collapse
|
15
|
Santopietro AL, Einstein D, Bellmunt J. Advances in the management of urothelial carcinoma: is immunotherapy the answer? Expert Opin Pharmacother 2021; 22:1743-1759. [PMID: 33905290 DOI: 10.1080/14656566.2021.1921149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Abstract
Introduction: Urothelial carcinoma (UC) is the second most common malignancy of the genitourinary system in the US, but mortality rate has not significantly improved despite advances in therapy. Over the past few years, the treatment landscape of non-muscle-invasive, muscle-invasive and metastatic UC (mUC) has evolved with the advent of immunotherapy.Areas covered: This paper summarizes current data and ongoing research into the use of immune checkpoint inhibitors (ICIs) in various settings of UC, including as maintenance therapy in chemotherapy-responsive mUC (with recent approval for avelumab in this setting) and as neoadjuvant and adjuvant therapies in localized and non-muscle-invasive disease. In addition, the authors review the combination of ICIs with chemotherapy, radiation and targeted therapies in an effort to increase response durability and efficacy.Expert opinion: While there has been a rapid expansion in clinical trials, platinum-based chemotherapy remains standard treatment in perioperative and first-line metastatic UC. The identification of biomarkers that can identify patients who will respond to ICIs has yielded conflicting results and has been largely non-generalizable to clinical practice. Further research into novel strategies and combinations with ICIs is needed to better characterize the role of immunotherapy in UC.
Collapse
Affiliation(s)
| | - David Einstein
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Joaquim Bellmunt
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
16
|
Adam T, Becker TM, Chua W, Bray V, Roberts TL. The Multiple Potential Biomarkers for Predicting Immunotherapy Response-Finding the Needle in the Haystack. Cancers (Basel) 2021; 13:cancers13020277. [PMID: 33451015 PMCID: PMC7828488 DOI: 10.3390/cancers13020277] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are being increasingly utilised in a variety of advanced malignancies. Despite promising outcomes in certain patients, the majority will not derive benefit and are at risk of potentially serious immune-related adverse events (irAEs). The development of predictive biomarkers is therefore critical to personalise treatments and improve outcomes. A number of biomarkers have shown promising results, including from tumour (programmed cell death ligand 1 (PD-L1), tumour mutational burden (TMB), stimulator of interferon genes (STING) and apoptosis-associated speck-like protein containing a CARD (ASC)), from blood (peripheral blood mononuclear cells (PBMCs), circulating tumour DNA (ctDNA), exosomes, cytokines and metal chelators) and finally the microbiome.
Collapse
Affiliation(s)
- Tamiem Adam
- Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW 2170, Australia; (T.M.B.); (W.C.)
- School of Medicine, Western Sydney University, Campbelltown, NSW 2170, Australia
- Liverpool Cancer Therapy Centre, Corner of Goulburn and Elizabeth Streets, Liverpool, NSW 2170, Australia;
- Correspondence: (T.A.); (T.L.R.)
| | - Therese M. Becker
- Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW 2170, Australia; (T.M.B.); (W.C.)
- University of New South Wales, Sydney, NSW 2170, Australia
| | - Wei Chua
- Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW 2170, Australia; (T.M.B.); (W.C.)
- School of Medicine, Western Sydney University, Campbelltown, NSW 2170, Australia
- Liverpool Cancer Therapy Centre, Corner of Goulburn and Elizabeth Streets, Liverpool, NSW 2170, Australia;
| | - Victoria Bray
- Liverpool Cancer Therapy Centre, Corner of Goulburn and Elizabeth Streets, Liverpool, NSW 2170, Australia;
| | - Tara L. Roberts
- Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW 2170, Australia; (T.M.B.); (W.C.)
- School of Medicine, Western Sydney University, Campbelltown, NSW 2170, Australia
- University of New South Wales, Sydney, NSW 2170, Australia
- Correspondence: (T.A.); (T.L.R.)
| |
Collapse
|