1
|
Andrade DAP, Bonatelli M, de Paula FE, Berardinelli GN, Teixeira GR, dos Reis MT, Barbin FF, Andrade CEMDC, Aguiar VP, Hermoza AD, Hirai WY, Schmidt RL, Reis RM, dos Reis R. Implementation of the ProMisE classifier and validation of its prognostic impact in Brazilian endometrial carcinomas. Front Oncol 2024; 14:1503901. [PMID: 39735598 PMCID: PMC11671357 DOI: 10.3389/fonc.2024.1503901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose Molecular classification of endometrial cancer (EC) has emerged as a key approach to individualize therapy and define prognostic outcomes. This study aimed to implement the traditional ProMisE classification in a Brazilian population, compared with a molecular setting of ProMisE biomarkers, and evaluate its impact on patients' prognosis. Patient and methods A prospective cohort of 114 patients with primary EC treated at Barretos Cancer Hospital (BCH) between October 2020 and December 2022 was conducted. Pathology diagnosis, staging, treatment, and follow-up data were collected. The traditional ProMisE methodology was carried out by POLE hotspot sequencing and immunohistochemistry (IHC) for p53 and mismatch repair (MMR) proteins. We further evaluate the MMR and TP53 status by molecular approach, namely microsatellite instability (MSI) by PCR-based and TP53 mutation analysis by next-generation sequencing (NGS). The results of the 4 molecular groups in both methodologies were compared regarding agreement accuracy and survival outcomes. Results Among the 114 cases, the traditional ProMisE groups were: POLEmut 15.8%, MMRd 28.1%, p53abn 27.2%, and no specific molecular profile (NSMP) 28.9%. Considering the molecular classification approach, we observed a POLEmut group of 15.8%, MSI group of 23.7%, TP53 mutation of 27.2%, and NSMP of 33.3%. The concordance rate of both approaches was 86.8% (99/114 cases) with an overall accuracy of 0.87. Importantly, both traditional and molecular ProMisE approaches were associated with significant distinct overall survival (OS) and progression-free survival (PFS) outcomes, with POLEmut patients exhibiting a better prognosis (93.8% OS, at 24 months), whereas the p53abn having a worse survival time (68.9% of OS, at 24 months). Conclusion We reported for the first time the Brazilian profile of the ProMisE classification of endometrial cancer and demonstrated the prognostic impact of the traditional and molecular ProMisE classification on patient outcomes.
Collapse
Affiliation(s)
| | - Murilo Bonatelli
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
| | | | | | - Gustavo Ramos Teixeira
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
- Pathology Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata, FACISB, São Paulo, Brazil
| | | | | | - Carlos Eduardo Mattos da Cunha Andrade
- Barretos School of Health Sciences Dr. Paulo Prata, FACISB, São Paulo, Brazil
- Gynecologic Oncology Department, Barretos Cancer Hospital, São Paulo, Brazil
| | | | | | - Welinton Yoshio Hirai
- Department of Epidemiology and Biostatistics, Barretos Cancer Hospital, São Paulo, Brazil
| | - Ronaldo Luís Schmidt
- Department of Surgical Oncology, Lagarto Unit, Barretos Cancer Hospital, Sergipe, Brazil
| | - Rui Manuel Reis
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Ricardos dos Reis
- Gynecologic Oncology Department, Barretos Cancer Hospital, São Paulo, Brazil
| |
Collapse
|
2
|
Chen W, Zheng K, Yuan W, Jia Z, Wu Y, Duan X, Yang W, Wen Z, Zhong L, Liu X. A CT-based deep learning for segmenting tumors and predicting microsatellite instability in patients with colorectal cancers: a multicenter cohort study. LA RADIOLOGIA MEDICA 2024:10.1007/s11547-024-01909-5. [PMID: 39586941 DOI: 10.1007/s11547-024-01909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE To develop and validate deep learning (DL) models using preoperative contrast-enhanced CT images for tumor auto-segmentation and microsatellite instability (MSI) prediction in colorectal cancer (CRC). MATERIALS AND METHODS Patients with CRC who underwent surgery or biopsy between January 2018 and April 2023 were retrospectively enrolled. Mismatch repair protein expression was determined via immunohistochemistry or fluorescence multiplex polymerase chain reaction-capillary electrophoresis. Manually delineated tumor contours using arterial and venous phase CT images by three abdominal radiologists are served as ground truth. Tumor auto-segmentation used nnU-Net. MSI prediction employed ViT or convolutional neural networks models, trained and validated with arterial and venous phase images (image model) or combined clinical-pathological factors (combined model). The segmentation model was evaluated using patch coverage ratio, Dice coefficient, recall, precision, and F1-score. The predictive models' efficacy was assessed using areas under the curves and decision curve analysis. RESULTS Overall, 2180 patients (median age: 61 years ± 17 [SD]; 1285 males) were divided into training (n = 1159), validation (n = 289), and independent external test (n = 732) groups. High-level MSI status was present in 435 patients (20%). In the external test set, the segmentation model performed well in the arterial phase, with patch coverage ratio, Dice coefficient, recall, precision, and F1-score values of 0.87, 0.71, 0.72, 0.74, and 0.71, respectively. For MSI prediction, the combined models outperformed the clinical model (AUC = 0.83 and 0.82 vs 0.67, p < 0.001) and two image models (AUC = 0.75 and 0.77, p < 0.001). Decision curve analysis confirmed the higher net benefit of the combined model compared to the other models across probability thresholds ranging from 0.1 to 0.45. CONCLUSION DL enhances tumor segmentation efficiency and, when integrated with contrast-enhanced CT and clinicopathological factors, exhibits good diagnostic performance in predicting MSI in CRC.
Collapse
Affiliation(s)
- Weicui Chen
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Kaiyi Zheng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wenjing Yuan
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Ziqi Jia
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Yuankui Wu
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaohui Duan
- Radiology Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Wei Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhibo Wen
- Radiology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Liming Zhong
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Xian Liu
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
3
|
Lin DI, Quintanilha JCF, Danziger N, Lang L, Levitan D, Hayne C, Hiemenz MC, Smith DL, Albacker LA, Leibowitz J, Mata DA, Decker B, Lakis S, Patel NR, Graf RP, Elvin JA, Ross JS, Pattani V, Huang RSP, Wehn AK. Pan-tumor validation of a NGS fraction-based MSI analysis as a predictor of response to Pembrolizumab. NPJ Precis Oncol 2024; 8:204. [PMID: 39277692 PMCID: PMC11401835 DOI: 10.1038/s41698-024-00679-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024] Open
Abstract
Microsatellite instability high (MSI-H) and mismatch repair deficient (dMMR) tumor status have been demonstrated to predict patient response to immunotherapies. We developed and validated a next-generation sequencing (NGS)-based companion diagnostic (CDx) to detect MSI-H solid tumors via a comprehensive genomic profiling (CGP) assay, FoundationOne®CDx (F1CDx). To determine MSI status, F1CDx calculates the fraction of unstable microsatellite loci across >2000 loci using a fraction-based (FB) analysis. Across solid tumor types, F1CDx demonstrated a high analytical concordance with both PCR (n = 264) and IHC (n = 279) with an overall percent agreement (OPA) of 97.7% and 97.8%, respectively. As part of a retrospective bridging clinical study from KEYNOTE-158 Cohort K and KEYNOTE-164, patients with MSI-H tumors as determined by F1CDx demonstrated an objective response rate (ORR) of 43.0% to pembrolizumab. In real-world cancer patients from a deidentified clinicogenomic database, F1CDx was at least equivalent in assessing clinical outcome following immunotherapy compared with MMR IHC. Demonstrated analytical and clinical performance of F1CDx led to the pan-tumor FDA approval in 2022 of F1CDx to identify MSI-H solid tumor patients for treatment with pembrolizumab. F1CDx is an accurate, reliable, and FDA-approved method for the identification of MSI-H tumors for treatment with pembrolizumab.
Collapse
Affiliation(s)
| | | | | | | | | | - Cynthia Hayne
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Zucca LER, Laus AC, Sorroche BP, Paro E, Sussuchi L, Marques RF, Teixeira GR, Berardinelli GN, Arantes LMRB, Reis RM, Cárcano FM. Immune-checkpoint gene expression and BCG response in non-muscle invasive bladder cancer. Transl Oncol 2024; 46:102003. [PMID: 38838438 PMCID: PMC11214516 DOI: 10.1016/j.tranon.2024.102003] [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: 12/22/2023] [Revised: 04/05/2024] [Accepted: 05/19/2024] [Indexed: 06/07/2024] Open
Abstract
METHODS One-hundred-six patients diagnosed with non-muscle invasive bladder cancer and treated with intravesical BCG were included and divided into two groups, BCG-responsive (n = 47) and -unresponsive (n = 59). Immunohistochemistry was used to evaluate PD-L1 expression and MSI was assessed by a commercial multiplex PCR kit. The mRNA expression profile of 15 immune checkpoints was performed using the nCounter technology. For in silico validation, two distinct cohorts sourced from the Gene Expression Omnibus (GEO) database were used. RESULTS Among the 106 patients, only one (<1 %) exhibited MSI instability. PD-L1 expression was present in 9.4 % of cases, and no association was found with BCG-responsive status. We found low gene expression of canonic actionable immune checkpoints PDCD1 (PD-1), CD274 (PD-L1), and CTLA4, while high expression was observed for CD276 (B7-H3), CD47, TNFRSF14, IDO1 and PVR (CD155) genes. High IDO1 expression levels was associated with worst overall survival. The PDCD1, CTLA4 and TNFRSF14 expression levels were associated with BCG responsiveness, whereas TIGIT and CD276 were associated with unresponsiveness. Finally, CD276 was validated in silico cohorts. CONCLUSION In NMIBC, MSI is rare and PD-L1 expression is present in a small subset of cases. Expression levels of PDCD1, CTLA4, TNFRSF14, TIGIT and CD276 could constitute predictive biomarkers of BCG responsiveness.
Collapse
Affiliation(s)
- Luis Eduardo Rosa Zucca
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Instituto do Câncer Brasil, Taubaté, Brazil
| | - Ana Carolina Laus
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | - Eduarda Paro
- Barretos School of Health Sciences Dr. Paulo Prata - FACISB, Barretos, Brazil
| | - Luciane Sussuchi
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Rui Ferreira Marques
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal
| | | | | | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal; 3ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Flavio Mavignier Cárcano
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Oncoclinicas & Co - Medica Scientia Innovation Research (MEDSIR), Sao Paulo, Brazil.
| |
Collapse
|
5
|
Li X, Che Y, Wang X, Zhu Y. A pan-cancer analysis of the core pre-mRNA 3' end processing factors, and their association with prognosis, tumor microenvironment, and potential targets. Sci Rep 2024; 14:17428. [PMID: 39075070 PMCID: PMC11286879 DOI: 10.1038/s41598-024-57402-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/18/2024] [Indexed: 07/31/2024] Open
Abstract
Alternative polyadenylation (APA) is a crucial mechanism for regulating gene expression during pre-mRNA 3' processing. Pre-mRNA 3' end processing factors is the main factor involved in this process. However, pre-mRNA 3' end processing factors in different cancer expression profiles and the relationship between pre-mRNA 3' end processing factors and tumor microenvironment and the prognosis of the same patient is still unclear. In this study, we conducted a comprehensive exploration of the core pre-mRNA 3' end processing factors across various cancer types by utilizing common cancer database, and revealing a robust correlation between the expression of these core factors and tumor characteristics. Leveraging advanced bioinformatics databases, we evaluated the expression levels and prognostic relevance of pre-mRNA 3' end processing factors across pan-cancer tissues. Our extensive pan-cancer analysis revealed unique expression patterns of pre-mRNA 3' end processing factors in both tumor and adjacent non-tumorous tissues. Notably, we found a significant correlation between the expression levels of pre-mRNA 3' end processing factors and patient prognosis. Furthermore, we identified strong associations between pre-mRNA 3' end processing factors expression and various factors, such as stromal, immune, RNA stemness, and DNA stemness scores across pan-cancer tissues. Our data also highlighted a link between the expression of pre-mRNA 3' end processing factors and sensitivity to specific drugs, including pyrazoloacndine, amonaflide, and chelerythrinede, among others. We found four key pre-mRNA 3' end processing factors that play a crucial role in mRNA preprocessing. Our study illuminates the potential promotion and inhibition role of pre-mRNA 3' end processing regulators in the progression of cancer, CPSF2, CPSF3, CSTF2, SYMPK offering valuable insights for future research investigations on these regulators as diagnostic markers and therapeutic targets across pan-cancer.
Collapse
Affiliation(s)
- Xiangyu Li
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yu Che
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoyan Wang
- Chongqing Medical and Pharmaceutical College, Chongqing, 401331, China.
| | - Yong Zhu
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
6
|
Laus AC, Gomes INF, da Silva ALV, da Silva LS, Milan MB, AparecidaTeixeira S, Martin ACBM, do Nascimento Braga Pereira L, de Carvalho CEB, Crovador CS, de Paula FE, Nascimento FC, de Freitas HT, de Lima Vazquez V, Reis RM, da Silva-Oliveira RJ. Establishment and molecular characterization of HCB-541, a novel and aggressive human cutaneous squamous cell carcinoma cell line. Hum Cell 2024; 37:1170-1183. [PMID: 38565739 PMCID: PMC11194207 DOI: 10.1007/s13577-024-01054-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a common type of skin cancer that can result in significant morbidity, although it is usually well-managed and rarely metastasizes. However, the lack of commercially available cSCC cell lines hinders our understanding of this disease. This study aims to establish and characterize a new metastatic cSCC cell line derived from a Brazilian patient. A tumor biopsy was taken from a metastatic cSCC patient, immortalized, and named HCB-541 after several passages. The cytokeratin expression profile, karyotypic alterations, mutational analysis, mRNA and protein differential expression, tumorigenic capacity in xenograft models, and drug sensitivity were analyzed. The HCB-541 cell line showed a doubling time between 20 and 30 h and high tumorigenic capacity in the xenograft mouse model. The HCB-541 cell line showed hypodiploid and hypotetraploidy populations. We found pathogenic mutations in TP53 p.(Arg248Leu), HRAS (Gln61His) and TERT promoter (C228T) and high-level microsatellite instability (MSI-H) in both tumor and cell line. We observed 37 cancer-related genes differentially expressed when compared with HACAT control cells. The HCB-541 cells exhibited high phosphorylated levels of EGFR, AXL, Tie, FGFR, and ROR2, and high sensitivity to cisplatin, carboplatin, and EGFR inhibitors. Our study successfully established HCB-541, a new cSCC cell line that could be useful as a valuable biological model for understanding the biology and therapy of metastatic skin cancer.
Collapse
Affiliation(s)
- Ana Carolina Laus
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Izabela Natalia Faria Gomes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Aline Larissa Virginio da Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Luciane Sussuchi da Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Mirella Baroni Milan
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Silvia AparecidaTeixeira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Ana Carolina Baptista Moreno Martin
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Letícia do Nascimento Braga Pereira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | | | - Camila Souza Crovador
- Department of Surgery of Melanoma and Sarcoma, Barretos Cancer Hospital, São Paulo, Brazil
| | - Flávia Escremin de Paula
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Flávia Caroline Nascimento
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Helder Teixeira de Freitas
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
| | - Vinicius de Lima Vazquez
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
- Department of Surgery of Melanoma and Sarcoma, Barretos Cancer Hospital, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil
- Life and Health Sciences Research Institute (ICVS) Medical School, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos, São Paulo, Brazil
| | - Renato José da Silva-Oliveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Antenor Duarte Villela, 1331, Barretos, São Paulo, Zip Code: 14784 400, Brazil.
- Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos, São Paulo, Brazil.
| |
Collapse
|
7
|
Vieira RADC, Sant'Anna D, Laus AC, Reis RM. Ancestry and self-reported race in Brazilian breast cancer women. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230767. [PMID: 37909531 PMCID: PMC10615220 DOI: 10.1590/1806-9282.20230767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE This study aimed to evaluate the association between self-reported race/color and ancestry in Brazilian patients with breast cancer. METHODS This was an observational, transversal, epidemiological study, evaluating race and ancestry in 1,127 patients with breast cancer. For genetic ancestry, a 46-AIM-INDEL panel was used. The ancestral profile was evaluated with the Structure v.2.3.3 software. Descriptive statistics were performed. To assess differences between race and ancestry, an analysis of variance with Bonferoni adjustment was used. RESULTS The race distribution was 77.7% white, 17.6% brown, 4.1% black, 0.4% yellow, and 0.3% cafuse. The African ancestry proportion was significantly (p<0.001) more evident in black [0.63±0.21 (0.17-0.96)], followed by brown [0.25±0.16 (0.02-0.70)], and less frequent in white skin color. The European ancestry proportion was significantly (p<0.001) higher in white [0.72±0.17 (0.02-0.97)], followed by brown [0.57±0.19 (0.12-0.92)], yellow [0.27±0.31 (0.12-0.620], and black [0.24±0.19 (0.02-0.72)]. The Asiatic ancestry proportion is significantly (p<0.001) higher in yellow [0.48±0.51 (0.04-0.93)]. The Amerindian ancestry proportion frequency was the least frequent in all groups, and cafuse patients did not express differences between all race groups. The brown race group presented differences in African and European ancestry. CONCLUSION Although we found many similarities between white European ancestry, black African ancestry, and yellow Asian ancestry, there is great miscegenation between patients. Although they can be labeled as having one race, they do present many ancestral genes that would allow their inclusion in another race group.
Collapse
Affiliation(s)
- René Aloisio da Costa Vieira
- Hospital de Câncer de Barretos, Molecular Oncology Research Center – Barretos (SP), Brazil
- Hospital de Câncer de Barretos, Postgraduate Program in Oncology – Barretos (SP), Brazil
- Faculdade de Medicina de Botucatu, Postgraduate Program in Tocogynecology – Botucatu (SP), Brazil
| | - Débora Sant'Anna
- Hospital de Câncer de Barretos, Molecular Oncology Research Center – Barretos (SP), Brazil
| | - Ana Carolina Laus
- Hospital de Câncer de Barretos, Molecular Oncology Research Center – Barretos (SP), Brazil
| | - Rui Manuel Reis
- Hospital de Câncer de Barretos, Molecular Oncology Research Center – Barretos (SP), Brazil
- Hospital de Câncer de Barretos, Postgraduate Program in Oncology – Barretos (SP), Brazil
- University of Minho, School of Medicine, Life and Health Sciences Research Institute – Braga, Portugal
- Life and Health Sciences Research Institute/3B's Research Group (Biomaterials, Biodegradables and Biomimetics)-PT Government Associate Laboratory – Braga/Guimarães, Portugal
| |
Collapse
|
8
|
Tostes K, Siqueira AP, Reis RM, Leal LF, Arantes LMRB. Biomarkers for Immune Checkpoint Inhibitor Response in NSCLC: Current Developments and Applicability. Int J Mol Sci 2023; 24:11887. [PMID: 37569262 PMCID: PMC10418476 DOI: 10.3390/ijms241511887] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer has the highest mortality rate among all cancer types, resulting in over 1.8 million deaths annually. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) has revolutionized the treatment of non-small cell lung cancer (NSCLC). ICIs, predominantly monoclonal antibodies, modulate co-stimulatory and co-inhibitory signals crucial for maintaining immune tolerance. Despite significant therapeutic advancements in NSCLC, patients still face challenges such as disease progression, recurrence, and high mortality rates. Therefore, there is a need for predictive biomarkers that can guide lung cancer treatment strategies. Currently, programmed death-ligand 1 (PD-L1) expression is the only established biomarker for predicting ICI response. However, its accuracy and robustness are not consistently reliable. This review provides an overview of potential biomarkers currently under development or in the validation stage that hold promise in improving the classification of responders and non-responders to ICI therapy in the near future.
Collapse
Affiliation(s)
- Katiane Tostes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
| | - Aléxia Polo Siqueira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4806-909 Guimarães, Portugal
| | - Leticia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
- Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos 14785-002, São Paulo, Brazil
| | | |
Collapse
|
9
|
Marques RF, Moreno DA, da Silva L, Leal LF, de Paula FE, Santana I, Teixeira G, Saggioro F, Neder L, Junior CA, Mançano B, Reis RM. Digital expression profile of immune checkpoint genes in medulloblastomas identifies CD24 and CD276 as putative immunotherapy targets. Front Immunol 2023; 14:1062856. [PMID: 36825029 PMCID: PMC9941636 DOI: 10.3389/fimmu.2023.1062856] [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: 10/06/2022] [Accepted: 01/24/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Medulloblastoma is the most common and lethal pediatric malignant brain tumor. It comprises four main molecular subgroups: WNT-activated, SHH-activated, Group 3, and Group 4. Medulloblastoma treatment is surgical resection, craniospinal radiation, and chemotherapy. However, many patients do not respond to therapy, and most suffer severe side effects. Cancer immunotherapy targeting immune checkpoints (IC) (PD-1, PD-L1, and CTLA4) has been getting disappointing outcomes in brain tumors. Nevertheless, other less explored immune checkpoints may be promising candidates for medulloblastoma therapy. Objectives In the present study, we aimed to characterize the expression profile of 19 immune checkpoints in medulloblastoma. Methods We analyzed 88 formalin-fixed paraffin-embedded medulloblastomas previously classified for each molecular subgroup and three non-tumoral brain tissue. mRNA levels of 19 immune checkpoint-related genes were quantified using the nCounter (PanCancer Immune Profiling Panel) assay. Further in silico analysis was performed in two larger public microarray datasets, one of which enabled comparisons between tumoral and non-tumoral tissues. Immunohistochemistry of PD-L1 was performed in a subset of cases. Microsatellite instability was also molecularly analyzed. Results We observed an absence of expression of the canonic ICs, namely PDCD1 (PD-1), CD274 (PD-L1), and CTLA4, as well as CD80, CD86, BTLA, IDO1, CD48, TNFSF14, CD160, CEACAM1, and CD244. PD-L1 protein expression was also practically absent. We found higher mRNA levels of CD24, CD47, CD276 (B7-H3), and PVR, and lower mRNA levels of HAVCR2, LAG3, and TIGIT genes, with significant differences across the four molecular subgroups. Compared to the non-tumor tissues, the expression levels of CD276 in all subgroups and CD24 in SHH, Group 3, and Group 4 subgroups are significantly higher. The in silico analysis confirmed the expression profile found in the Brazilian cohort, including the lower/absent expression of the canonic ICs. Moreover, it confirmed the overexpression of CD24 and CD276 in medulloblastomas compared with the non-tumor tissue. Additionally, CD276 and CD24 high levels were associated with worse survival. Conclusion These results highlight the low or absence of mRNA levels of the canonic targetable ICs in medulloblastomas. Importantly, the analysis revealed overexpression of CD24 and CD276, which can constitute prognostic biomarkers and attractive immunotherapy targets for medulloblastomas.
Collapse
Affiliation(s)
- Rui Ferreira Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | | | - Luciane da Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Leticia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Faculty of Health Sciences of Barretos Dr. Paulo Prata (FACISB), School of Medicine, Barretos, Brazil
| | | | - Iara Santana
- Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - Gustavo Teixeira
- Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - Fabiano Saggioro
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luciano Neder
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Bruna Mançano
- Barretos Children's Cancer Hospital, Barretos, Brazil
| | - Rui Manuel Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Braga, Guimarães, Portugal.,Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Laboratory of Molecular Diagnostic, Barretos Cancer Hospital, Barretos, Brazil
| |
Collapse
|
10
|
Corso G. Microsatellite instability in gastrointestinal cancers. Eur J Hum Genet 2022; 30:996-997. [PMID: 35726018 PMCID: PMC9437054 DOI: 10.1038/s41431-022-01134-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/18/2022] [Accepted: 06/09/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Giovanni Corso
- Division of Breast Surgery, European Institute of Oncology (IEO), IRCCS, Milan, 20141, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, 20122, Italy.
- European Cancer Prevention Organization (ECP), Milan, 20141, Italy.
| |
Collapse
|
11
|
Vicente ALSA, Novoloaca A, Cahais V, Awada Z, Cuenin C, Spitz N, Carvalho AL, Evangelista AF, Crovador CS, Reis RM, Herceg Z, de Lima Vazquez V, Ghantous A. Cutaneous and acral melanoma cross-OMICs reveals prognostic cancer drivers associated with pathobiology and ultraviolet exposure. Nat Commun 2022; 13:4115. [PMID: 35840550 PMCID: PMC9287446 DOI: 10.1038/s41467-022-31488-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 06/16/2022] [Indexed: 11/08/2022] Open
Abstract
Ultraviolet radiation (UV) is causally linked to cutaneous melanoma, yet the underlying epigenetic mechanisms, known as molecular sensors of exposure, have not been characterized in clinical biospecimens. Here, we integrate clinical, epigenome (DNA methylome), genome and transcriptome profiling of 112 cutaneous melanoma from two multi-ethnic cohorts. We identify UV-related alterations in regulatory regions and immunological pathways, with multi-OMICs cancer driver potential affecting patient survival. TAPBP, the top gene, is critically involved in immune function and encompasses several UV-altered methylation sites that were validated by targeted sequencing, providing cost-effective opportunities for clinical application. The DNA methylome also reveals non UV-related aberrations underlying pathological differences between the cutaneous and 17 acral melanomas. Unsupervised epigenomic mapping demonstrated that non UV-mutant cutaneous melanoma more closely resembles acral rather than UV-exposed cutaneous melanoma, with the latter showing better patient prognosis than the other two forms. These gene-environment interactions reveal translationally impactful mechanisms in melanomagenesis.
Collapse
Affiliation(s)
- Anna Luiza Silva Almeida Vicente
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France.
| | - Alexei Novoloaca
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - Vincent Cahais
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - Zainab Awada
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - Cyrille Cuenin
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - Natália Spitz
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - André Lopes Carvalho
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
- Early Detection Prevention and Infections Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | | | - Camila Souza Crovador
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - Vinicius de Lima Vazquez
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
- Department of Surgery-Melanoma and Sarcoma, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Akram Ghantous
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer (IARC), Lyon, France.
| |
Collapse
|
12
|
McNeill A. Clinical genomics testing: mainstreaming and globalising. Eur J Hum Genet 2022; 30:747-748. [PMID: 35794342 PMCID: PMC9259576 DOI: 10.1038/s41431-022-01131-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Alisdair McNeill
- grid.11835.3e0000 0004 1936 9262Department of Neuroscience, The University of Sheffield, Sheffield, UK ,grid.413991.70000 0004 0641 6082Sheffield Clinical Genetics Department, Sheffield Children’s Hospital NHS Foundation Trust, Sheffield, UK
| |
Collapse
|