Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.001
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 26221, 2024 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-39482333

RESUMO

Immunotherapy has become a primary and secondary treatment for gastric cancer (GC) patients with mismatch repair deficiency (dMMR), and is used in both perioperative and advanced stages. The tumor immune microenvironment (TiME) is crucial for immunotherapy efficacy, yet the impact of MMR status on TiME remains understudied. We employed single-cell RNA sequencing (scRNA-seq) to analyze 33 fresh tissue samples from 25 patients, which included 10 normal tissues, 6 dMMR tumor tissues, and 17 pMMR tumor tissues, aiming to characterize the cellular and molecular components of the TiME. The proficient mismatch repair (pMMR) group displayed a significantly higher prevalence of a specific GC cell type, termed GC2, characterized by increased hypoxia, epithelial-mesenchymal transition (EMT), and angiogenic activities compared to the dMMR group. GC2 cells overexpressed BEX3 and GPC3, and they significantly correlated with poorer survival. The pMMR group also showed increased infiltration of SPP1 + macrophages and FAP + fibroblasts, exhibiting strong hypoxic and pro-angiogenic features. Furthermore, a higher proportion of E2 endothelial cells, involved in extracellular matrix (ECM) remodeling and showing heightened VEGF pathway, HIF pathway, and angiogenesis activity, were identified in pMMR patients. Intercellular communication analyses revealed that GC2 cells, SPP1 + macrophages, FAP + fibroblasts, and E2 endothelial cells interact through VEGF, SPP1, and MIF signals, forming a TiME characterized by hypoxia, pro-angiogenesis, and ECM remodeling. This study uncovered TiME heterogeneity among GC patients with different MMR states, highlighting that the pMMR TiME is distinguished by hypoxia, pro-angiogenesis, and ECM remodeling, driven by the presence of GC2 cells, SPP1 + macrophages, FAP + fibroblasts, and E2 endothelial cells. These findings are pivotal for developing targeted immunotherapies for GC patients with pMMR.


Assuntos
Progressão da Doença , Macrófagos , Neoplasias Gástricas , Microambiente Tumoral , Humanos , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/genética , Macrófagos/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Masculino , Fibroblastos/metabolismo , Fibroblastos/patologia , Reparo de Erro de Pareamento de DNA , Pessoa de Meia-Idade , Linhagem Celular Tumoral , Gelatinases/metabolismo , Gelatinases/genética , Proteínas de Membrana , Endopeptidases
3.
Front Immunol ; 15: 1466497, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39399495

RESUMO

Background: In recent years, there has been significant research interest in immunotherapy for colorectal cancer (CRC). Specifically, immunotherapy has emerged as the primary treatment for patients with mismatch repair gene defects (dMMR) or microsatellite highly unstable (MSI-H) who have colorectal cancer. Yet, there is currently no data to support the practicality and safety of neoadjuvant immunotherapy for colorectal cancer with dMMR or MSI-H. Therefore, a study was conducted to identify the postoperative pathology, safety profile, and imaging features of patients with dMMR or MSI-H CRC following neoadjuvant immunotherapy. Methods: The retrospective study was carried out on patients with locally advanced or metastatic CRC who received immunotherapy at Sichuan Cancer Hospital, with approval from the hospital's ethics committee. The study aimed to assess the short-term effectiveness of immunotherapy by focusing on pathological complete response (pCR) as the primary outcome, while also considering secondary endpoints such as objective response rate, disease-free survival, and safety profile. Results: Twenty patients with dMMR/MSI-H CRC who underwent neoadjuvant immunotherapy as part of the treatment were enrolled between May 2019 and February 2024 at Sichuan Cancer Hospital. Out of these patients, eight patients received PD-1 blockade monotherapy as neoadjuvant treatment, while 12 were administered a combined therapy of anti-CTLA-4 and anti-PD-1. 12 patients received Nivolumab plus Ipilimumab regimen and 8 patients received PD-1 blockades (2 patients were Pembrolizumab, 2 patients were Sintilimab, 4 patients were Tislelizumab) monotherapy. Additionally, 19 patients underwent surgery after immunotherapy and of these, 15 (75.0%) achieved complete pathological response (pCR), 8 (66.7%) achieved the same on Nivolumab plus Ipilimumab immunotherapy while 7 (87.5%) achieved on PD-1 antibody monotherapy. The overall response rate (ORR) was 75%, with 45.0% of patients experiencing grade I/II immunotherapy-related adverse events. The most frequent adverse event observed was increased ALT i.e. 20%. Notably, no postoperative complications were observed. Conclusion: Based on the findings, neoadjuvant immunotherapy for colorectal cancer may be both safe and effective in clinical practice. Furthermore, the study suggested that dual immunotherapy could potentially increase the immunotherapy cycle and contribute to a superior pCR rate. However, the conclusion emphasized the need for further prospective clinical trials to validate these results.


Assuntos
Neoplasias Colorretais , Reparo de Erro de Pareamento de DNA , Imunoterapia , Instabilidade de Microssatélites , Terapia Neoadjuvante , Humanos , Neoplasias Colorretais/terapia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/imunologia , Terapia Neoadjuvante/métodos , Feminino , Pessoa de Meia-Idade , Masculino , Estudos Retrospectivos , Idoso , Imunoterapia/métodos , Adulto , Resultado do Tratamento , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/efeitos adversos
4.
BMC Cancer ; 24(1): 1232, 2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39375615

RESUMO

There is limited research on the clinicopathological characteristics of combined hepatocellular-cholangiocarcinoma (cHCC-CCA) currently. The aim of this study is to summerize the clinicopathological factors and prognosis of cHCC-CCA, which could help us understand this disease. 72 cases of cHCC-CCA from West China Hospital of Sichuan University were collected. Tissue components were reviewed by pathologists. Immunohistochemistry was used to detect the status of mismatch repair (MMR) and human epidermal growth factor receptor 2 (HER2) in cHCC-CCA, as well as the quantity and distribution of CD3+ T cells and CD8+ T cells. Fluorescence in situ hybridization was used to detect fibroblast growth factor receptor 2 (FGFR2) gene alteration. COX univariate and multivariate analyses were used to evaluate risk factors, and survival curves were plotted. 49 cases were classified as classic type cHCC-CCA and 23 cases as intermediate cell carcinoma. The cut-off value for diagnosing classic type was determined to be ≥ 30% for the cholangiocarcinoma component based on prognostic calculations. All tumors were MMR proficient. The rate of strong HER2 protein expression (3+) was 8.3%, and the frequency of FGFR2 gene alteration was 26.4%. CD3+ T cells and CD8+ T cells were mainly distributed at the tumor margin, and were protective factors for patients with cHCC-CCA. The overall survival of the 72 patients was 18.9 months, with a median survival of 12 months. Tumor size, TNM stage, and serum AFP level were prognostic factors for cHCC-CCA. The proportion of cholangiocarcinoma component reaching the threshold of 30%, may provide a reference for future pathology diagnosis. FGFR2 gene alteration was 26.4%, providing a clue for anti-FGFR2 therapy. However, more data is needed for further verification.


Assuntos
Neoplasias dos Ductos Biliares , Carcinoma Hepatocelular , Colangiocarcinoma , Neoplasias Hepáticas , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Humanos , Colangiocarcinoma/patologia , Colangiocarcinoma/genética , Colangiocarcinoma/mortalidade , Colangiocarcinoma/diagnóstico , Masculino , Feminino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Idoso , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/mortalidade , Neoplasias dos Ductos Biliares/diagnóstico , Adulto , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Biomarcadores Tumorais/genética , Reparo de Erro de Pareamento de DNA , Imuno-Histoquímica
5.
Gan To Kagaku Ryoho ; 51(9): 873-878, 2024 Sep.
Artigo em Japonês | MEDLINE | ID: mdl-39462618

RESUMO

Overall assessment of KEYNOTE-164 study, KEYNOTE-158 study and CheckMate 142 study demonstrated the clinical benefits of immune checkpoint inhibitors(ICIs)among patients with mismatch repair deficient(dMMR)/high microsatellite instability(MSI-H)cancer. As a result, ICIs have been approved for treatment of MSI-H solid tumor regardless of the tumor type. However, the frequency of real-world diagnosed dMMR gastrointestinal cancer were rarely reported. Therefore, the results of immunohistochemical staining for DNA mismatch repair proteins was investigated. 175 samples of gastrointestinal cancers were examined between November 2019 and June 2023. Clinical and pathological characteristics were obtained from clinical and histopathological records. In real world populations with high proportion of elderly people, the frequency of diagnosed dMMR gastrointestinal cancer may be high compared with previous reports. Furthermore, based on the deficient pattern of mismatch repair protein and age, most cases classified as dMMR may be sporadic. Right side tumors and female may increase the likelihood of dMMR colorectal cancer. The current results justified immunohistochemical staining for DNA mismatch repair proteins, strongly involved in the appropriate patient selection for ICIs therapy, should be conducted for elderly patients newly diagnosed as gastrointestinal cancer. We believe that further clinical cancer immunology research, and then challenging insight targeting dMMR gastrointestinal cancer will result in future development of novel immunotherapy combination strategies well tolerated even in elderly patients.


Assuntos
Reparo de Erro de Pareamento de DNA , Neoplasias Gastrointestinais , Inibidores de Checkpoint Imunológico , Imuno-Histoquímica , Humanos , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/imunologia , Neoplasias Gastrointestinais/patologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Idoso , Feminino , Masculino , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Adulto
6.
ESMO Open ; 9(10): 103929, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39357124

RESUMO

BACKGROUND: Sustained clinical complete remissions were reported in all of 23 mismatch repair deficient/microsatellite instable (dMMR/MSI) locally advanced rectal cancer (LARC) patients treated with dostarlimab alone in a recent phase II study. These results led to off-label use of dostarlimab or other immune checkpoint inhibitors (ICIs) in dMMR/MSI-LARC even before regulatory approval. The present study [STAR(t)-IT-REDUCE] describes the outcome of dMMR/MSI-LARC patients treated with ICI in Italy. MATERIALS AND METHODS: Investigator-initiated, observational, retrospective-cohort, multicentric study of ICI treatment in dMMR/MSI-LARC. Patients were eligible if treated with ≥1 ICI dose from July 2022 to December 2023 (date of approval of dostarlimab for this indication in Italy). RESULTS: Seventeen dMMR/MSI-LARC patients (13 of 17 treatment-naïve) were eligible. Fourteen patients completed 6 months of treatment, two discontinued after four doses and one after five doses because of immune-related pneumonia, social constraints, or non-oncological bowel obstruction, respectively. Overall, 16 of 17 assessable patients [94.1%; 95% confidence interval (CI) 69.24% to 99.69%, 'ITT analysis'] achieved complete clinical response (cCR). Ten of 11 treatment-naïve patients completing 6 months of treatment had cCR (90.9%; 95% CI 57.12% to 99.52%, 'per-protocol analysis'). One patient with near-CR underwent rectal surgery and minimal residual intramucosal tumor was found. With a median follow-up of 9.5 months, no local relapse occurred. One patient developed unconfirmed lung metastases. Two grade 3 and no grade 4 adverse events were reported. CONCLUSION: The present STAR(t)-IT-REDUCE study documents the immunoablative and curative activity of ICI monotherapy in dMMR/MSI-LARC. Toxicity and compliance issues inherent to real-world practice are limited and do not affect achievement of initial complete tumor response but may limit response duration.


Assuntos
Reparo de Erro de Pareamento de DNA , Inibidores de Checkpoint Imunológico , Neoplasias Retais , Humanos , Neoplasias Retais/tratamento farmacológico , Neoplasias Retais/patologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Estudos Retrospectivos , Itália , Adulto , Resultado do Tratamento , Idoso de 80 Anos ou mais
7.
Asian Pac J Cancer Prev ; 25(10): 3667-3671, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39471035

RESUMO

BACKGROUND: Mismatch repair deficiency (dMMR) is a well-established characteristic of endometrial adenocarcinoma and is crucial in screening for Lynch syndrome, guiding adjuvant treatment decisions, and identifying candidates for immune checkpoint inhibitors. The traditional approach to dMMR screening involves a four-antibody panel, but a simplified two-antibody method utilizing PMS2 and MSH6 has shown promise. This study aims to compare the diagnostic performance of the simplified two-antibody method with the traditional four-antibody panel in endometrial cancer samples. METHODS: We conducted a retrospective cohort study on endometrial carcinoma cases diagnosed between 2013 and 2022. We compared the diagnostic performance of the two-antibody panel with the traditional four-antibody panel in detecting dMMR. Clinical data and immunohistochemistry results were collected, and agreement between the two methods was evaluated using Cohen's kappa coefficient. RESULTS: 304 endometrial cancer cases were included, with 27% demonstrating loss of at least one MMR protein using the four-antibody panel. The two-antibody method detected MMR deficiency in 26.6% of cases, with a high agreement rate of 98.8% between the two methods. Only one case showed discordant results, prompting further investigation. CONCLUSION: The simplified two-antibody MMR IHC screening approach using PMS2 and MSH6 showed high concordance with the traditional four-antibody panel. This suggests its potential as an alternative method for reflex MMR status testing in endometrial adenocarcinoma. The implementation of this approach could streamline the diagnostic process, reduce costs, and improve the detection of Lynch syndrome in affected individuals and their families. Further studies with larger cohorts and long-term follow-up are needed to validate these findings and assess the clinical implications of this approach in routine practice.


Assuntos
Adenocarcinoma , Biomarcadores Tumorais , Proteínas de Ligação a DNA , Neoplasias do Endométrio , Imuno-Histoquímica , Endonuclease PMS2 de Reparo de Erro de Pareamento , Humanos , Feminino , Neoplasias do Endométrio/patologia , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/diagnóstico , Endonuclease PMS2 de Reparo de Erro de Pareamento/metabolismo , Estudos Retrospectivos , Proteínas de Ligação a DNA/metabolismo , Pessoa de Meia-Idade , Imuno-Histoquímica/métodos , Biomarcadores Tumorais/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/diagnóstico , Adenocarcinoma/metabolismo , Seguimentos , Idoso , Reparo de Erro de Pareamento de DNA , Prognóstico , Adulto , Detecção Precoce de Câncer/métodos
8.
Fam Cancer ; 23(4): 569-577, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39387980

RESUMO

The discovery of "mismatch repair deficient (MMRd)-crypt foci" in non-neoplastic intestinal mucosa in Lynch syndrome (LS) has significantly enhanced our understanding of how tumors and tumor immunity form and evolve in LS. In this study, we report the frequent presence of "mismatch repair proficient (MMRp)-crypt foci" in both non-neoplastic and neoplastic intestinal mucosa in a patient with constitutional MMR deficiency (CMMRD), who carried a germline MSH6 pathogenic variant (c.3261dupC) in trans with an MSH6 likely pathogenic variant (c.3724_3726del) and whose tissues were otherwise deficient in MMR globally. The MMRp-crypts occurred at a rate of 1.1/100 crypts in non-neoplastic intestinal mucosa and were readily discernible in adenomas > 1 cm. Sequencing analysis revealed normalization of the MSH6c.3261dupC variant in MMRp-adenoma crypts, indicating reverse frameshifting of the exon 5 C8 microsatellite. Interestingly but not surprisingly, the MMRp-adenoma crypts remained microsatellite-instability-high (MSI-H), and shared oncogenic APC mutations with the background MMRd-adenoma. Contrasting with MSH6-CMMRD, no PMS2-CMMRD individuals (0/5) harbored MMRp-crypts. In conclusion, our study documents distinct MMRp-crypts in MSH6-CMMRD, a phenomenon in keeping with MSH6 being a frequent target of MSI-H due to its coding microsatellite and suggesting that MSH6-CMMRD can potentially serve as a unique model system to further our understanding of MSH6's role in MSI-H tumor formation and evolution. Our findings also bear diagnostic implications; when using MMR immunohistochemistry as an ancillary tool in detecting CMMRD, awareness of these MMRp crypts can help avoid diagnostic pitfalls.


Assuntos
Proteínas de Ligação a DNA , Humanos , Proteínas de Ligação a DNA/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/patologia , Instabilidade de Microssatélites , Mutação da Fase de Leitura , Reparo de Erro de Pareamento de DNA/genética , Feminino , Repetições de Microssatélites/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Mutação em Linhagem Germinativa , Masculino , Mucosa Intestinal/patologia , Pessoa de Meia-Idade , Focos de Criptas Aberrantes/genética , Focos de Criptas Aberrantes/patologia , Síndromes Neoplásicas Hereditárias/genética , Adenoma/genética , Adenoma/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia
9.
ESMO Open ; 9(9): 103698, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39241497

RESUMO

BACKGROUND: Immunotherapy is used routinely for treating deficient mismatch repair (dMMR) colon cancer (CC). This case series highlights an emerging safety issue, where patients develop bowel obstruction associated with immunotherapy response. PATIENTS AND METHODS: Patients with dMMR CC who developed bowel obstruction while responding to immunotherapy were retrospectively identified. Data on patient, disease, treatment, and response-specific factors were explored for potential risk factors. Overall treatment numbers were used to estimate incidence. RESULTS: Nine patients from eight European centres were included. Common features were hepatic flexure location (5/9), T4 radiological staging (6/9), annular shape (8/9), radiological stricturing (5/9), and endoscopic obstruction (6/9). All received pembrolizumab and obstructed between 45 and 652 days after starting treatment. Seven patients underwent surgical resection; one was managed with a defunctioning stoma; and one was managed conservatively. One patient died from obstruction. Radiological response was seen in eight patients, including two complete responses. Pathological response was seen in all seven who underwent resection, including four complete responses. The overall incidence of immunotherapy response-related obstruction in these centres was 1.51%. CONCLUSIONS: Bowel obstruction associated with immunotherapy response may represent a rare treatment-related complication in dMMR CC. Clinicians must recognise this safety signal and share experience to maintain patient safety.


Assuntos
Neoplasias do Colo , Imunoterapia , Obstrução Intestinal , Humanos , Obstrução Intestinal/etiologia , Masculino , Feminino , Neoplasias do Colo/complicações , Imunoterapia/métodos , Imunoterapia/efeitos adversos , Pessoa de Meia-Idade , Idoso , Estudos Retrospectivos , Anticorpos Monoclonais Humanizados/uso terapêutico , Fatores de Risco , Reparo de Erro de Pareamento de DNA , Adulto
10.
DNA Repair (Amst) ; 143: 103768, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39332392

RESUMO

We show that the rates of single base substitutions, additions, and deletions across the nuclear genome are strongly increased in a strain harboring a mutator variant of DNA polymerase α combined with a mutation that inactivates the 3´-5´ exonuclease activity of DNA polymerase δ. Moreover, tetrad dissections attempting to produce a haploid triple mutant lacking Msh6, which is essential for DNA mismatch repair (MMR) of base•base mismatches made during replication, result in tiny colonies that grow very slowly and appear to be aneuploid and/or defective in oxidative metabolism. These observations are consistent with the hypothesis that during initiation of nuclear DNA replication, single-base mismatches made by naturally exonuclease-deficient DNA polymerase α are extrinsically proofread by DNA polymerase δ, such that in the absence of this proofreading, the mutation rate is strongly elevated. Several implications of these data are discussed, including that the mutational signature of defective extrinsic proofreading in yeast could appear in human tumors.


Assuntos
DNA Polimerase III , DNA Polimerase I , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , DNA Polimerase III/metabolismo , DNA Polimerase III/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , DNA Polimerase I/metabolismo , DNA Polimerase I/genética , Genoma Fúngico , Reparo de Erro de Pareamento de DNA , Replicação do DNA , Mutação , Taxa de Mutação , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Pareamento Incorreto de Bases , DNA Fúngico/metabolismo
11.
Eur J Pharmacol ; 983: 176979, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-39241942

RESUMO

Colon cancer is a significant health problem worldwide as it is one of the most common and deadliest cancers. The standard approach for the treatment of colon cancer is 5-fluorouracil (5-FU) based chemotherapy, which is limited by the development of resistance to this drug. Therefore, our study aimed to establish 5-FU resistance in SW-480 and HT-29 colon cancer cells and to precisely determine the molecular mechanisms and biomarkers that contribute to its development, both after short-term exposure and in cells with already developed resistance (SW-480-5FUR and HT-29-5FUR). The expression of various molecules involved in the different mechanisms of resistance development was monitored at the gene (qPCR) and protein (immunocytochemistry) levels. Based on the obtained results, alterations in the 5-FU anabolic pathway, biotransformation, drug efflux, mismatch repair, and apoptosis process together contributed to the development of 5-FU resistance in SW-480 and HT-29 colon cancer cells. In addition, UMPS, ABCC1, ABCC5, and MLH1, as well as the disturbed ratio of pro-apoptotic BAX and anti-apoptotic BCL2, should be taken into consideration as potential targets for the discovery of 5-FU resistance-related biomarkers in colon cancer cells. We suggest that future investigations focus on further validation of these findings by additional in vitro and in vivo testing, which is a limitation of our study.


Assuntos
Neoplasias do Colo , Resistencia a Medicamentos Antineoplásicos , Fluoruracila , Fluoruracila/farmacologia , Humanos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linhagem Celular Tumoral , Células HT29 , Antimetabólitos Antineoplásicos/farmacologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Proteína 1 Homóloga a MutL/genética , Proteína 1 Homóloga a MutL/metabolismo
12.
Nat Commun ; 15(1): 7695, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227583

RESUMO

Neoadjuvant immune checkpoint blockade (ICB) has shown unprecedented activity in mismatch repair deficient (MMRd) colorectal cancers, but its effectiveness in MMRd endometrial cancer (EC) remains unknown. In this investigator-driven, phase I, feasibility study (NCT04262089), 10 women with MMRd EC of any grade, planned for primary surgery, received two cycles of neoadjuvant pembrolizumab (200 mg IV) every three weeks. A pathologic response (primary objective) was observed in 5/10 patients, with 2 patients showing a major pathologic response. No patient achieved a complete pathologic response. A partial radiologic response (secondary objective) was observed in 3/10 patients, 5/10 patients had stable disease and 2/10 patients were non-evaluable on magnetic resonance imaging. All patients completed treatment without severe toxicity (exploratory objective). At median duration of follow-up of 22.5 months, two non-responders experienced disease recurrence. In-depth analysis of the loco-regional and systemic immune response (predefined exploratory objective) showed that monoclonal T cell expansion significantly correlated with treatment response. Tumour-draining lymph nodes displayed clonal overlap with intra-tumoural T cell expansion. All pre-specified endpoints, efficacy in terms of pathologic response as primary endpoint, radiologic response as secondary outcome and safety and tolerability as exploratory endpoint, were reached. Neoadjuvant ICB with pembrolizumab proved safe and induced pathologic, radiologic, and immunologic responses in MMRd EC, warranting further exploration of extended neoadjuvant treatment.


Assuntos
Anticorpos Monoclonais Humanizados , Reparo de Erro de Pareamento de DNA , Neoplasias do Endométrio , Inibidores de Checkpoint Imunológico , Terapia Neoadjuvante , Humanos , Feminino , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Neoplasias do Endométrio/imunologia , Neoplasias do Endométrio/diagnóstico por imagem , Inibidores de Checkpoint Imunológico/uso terapêutico , Pessoa de Meia-Idade , Anticorpos Monoclonais Humanizados/uso terapêutico , Idoso , Adulto , Resultado do Tratamento
13.
Cell Rep Med ; 5(9): 101727, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39293403

RESUMO

Testing for DNA mismatch repair deficiency (MMRd) is recommended for all colorectal cancers (CRCs). Automating this would enable precision medicine, particularly if providing information on etiology not captured by deep learning (DL) methods. We present AIMMeR, an AI-based method for determination of mismatch repair (MMR) protein expression at a single-cell level in routine pathology samples. AIMMeR shows an area under the receiver-operator curve (AUROC) of 0.98, and specificity of ≥75% at 98% sensitivity against pathologist ground truth in stage II/III in two trial cohorts, with positive predictive value of ≥98% for the commonest pattern of somatic MMRd. Lower agreement with microsatellite instability (MSI) testing (AUROC 0.86) reflects discordance between MMR and MSI PCR rather than AIMMeR misclassification. Analysis of the SCOT trial confirms MMRd prognostic value in oxaliplatin-treated patients; while MMRd does not predict differential benefit of chemotherapy duration, it correlates with difference in relapse by regimen (PInteraction = 0.04). AIMMeR may help reduce pathologist workload and streamline diagnostics in CRC.


Assuntos
Neoplasias Colorretais , Reparo de Erro de Pareamento de DNA , Instabilidade de Microssatélites , Análise de Célula Única , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/diagnóstico , Reparo de Erro de Pareamento de DNA/genética , Prognóstico , Análise de Célula Única/métodos , Feminino , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Curva ROC , Idoso
14.
Immunotherapy ; 16(10): 649-657, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39259508

RESUMO

Clinical evidences of neoadjuvant immunotherapy in patients with mismatch repair deficient/microsatellite instability-high status (dMMR/MSI-H) colorectal cancer have not been well received. A 36-year-old man complained of recurrent right upper quadrant pain for more than 1 year, and the symptoms were not significantly relieved after 10 days of oral Changyanning tablet. The patient was finally diagnosed as dMMR/MSI-H colon cancer. Tumor regression was achieved after seven cycles of envafolimab treatment, and the patient obtained postoperative pathological complete response (pCR). Here, we report a case of MSI-H/dMMR transverse colon cancer, who obtained pCR after neoadjuvant envafolimab (a novel subcutaneous single-domain anti-PD-L1 antibody) with a favorable safety profile, aiming to enhance the experiences of comprehensive diagnosis and treatment of colon cancer.


Immune checkpoint inhibitors (ICIs) are a type of immunotherapy which can be used in the treatment of colorectal cancer. The authors here report the functions of envafolimab (a type of ICI) used before surgery to shrink tumor volume in colorectal cancer. A 36-year-old man suffered from repeated illness of right upper quadrant for over 1 year, and the illness were not recovered after 10 days of oral Changyanning tablet. The patient was finally diagnosed with colorectal cancer. After seven cycles of envafolimab treatment, tumor volume was significantly decreased, and the patient obtained favorable surgical outcomes with tolerable safety after surgery.


Assuntos
Neoplasias do Colo , Instabilidade de Microssatélites , Terapia Neoadjuvante , Humanos , Masculino , Adulto , Neoplasias do Colo/diagnóstico , Neoplasias do Colo/terapia , Reparo de Erro de Pareamento de DNA , Inibidores de Checkpoint Imunológico/uso terapêutico
15.
Sci Rep ; 14(1): 21870, 2024 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300198

RESUMO

Microsatellite instability (MSI) occurs across a number of cancers and is associated with different clinical characteristics when compared to microsatellite stable (MSS) cancers. As MSI cancers have different characteristics, routine MSI testing is now recommended for a number of cancer types including colorectal cancer (CRC). Using gene panels for sequencing of known cancer mutations is routinely performed to guide treatment decisions. By adding a number of MSI regions to a small gene panel, the efficacy of simultaneous MSI detection in a series of CRCs was tested. Tumour DNA from formalin-fixed, paraffin-embedded (FFPE) tumours was sequenced using a 23-gene panel kit (ATOM-Seq) provided by GeneFirst. The mismatch repair (MMR) status was obtained for each patient from their routine pathology reports, and compared to MSI predictions from the sequencing data. By testing 29 microsatellite regions in 335 samples the MSI status was correctly classified in 314/319 samples (98.4% concordance), with sixteen failures. By reducing the number of regions in silico, comparable performance could be reached with as few as eight MSI marker positions. This test represents a quick, and accurate means of determining MSI status in FFPE CRC samples, as part of a routine gene mutation assay, and can easily be incorporated into a research or diagnostic setting. This could replace separate mutation and MSI tests with no loss of accuracy, thus improving testing efficiency.


Assuntos
Neoplasias Colorretais , Formaldeído , Instabilidade de Microssatélites , Mutação , Fixação de Tecidos , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/diagnóstico , Formaldeído/química , Inclusão em Parafina , Feminino , Masculino , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Reparo de Erro de Pareamento de DNA/genética , Análise Mutacional de DNA/métodos , Idoso , Pessoa de Meia-Idade
16.
J Gastrointestin Liver Dis ; 33(3): 339-347, 2024 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-39255351

RESUMO

BACKGROUND AND AIMS: Lynch Syndrome, a hereditary disorder characterized by germline mutations in mismatch repair (MMR) genes, is a major contributor to colorectal cancers. It has also been identified in endometrial cancer. Despite the established role of MMR deficiency in tumorigenesis, the specific genomic alterations driving Lynch syndrome-associated endometrial cancer, and their overlap with colorectal cancer, remain incompletely understood. This study aims to fill this gap by performing a detailed comparative analysis of germline and somatic mutations in endometrial cancer within the context of Lynch syndrome. METHODS: We conducted whole exome sequencing on matched germline and somatic DNA from 13 patients diagnosed with Lynch syndrome-associated endometrial cancer. High-depth sequencing was performed, followed by rigorous bioinformatics analysis to identify and annotate variants, focusing on their potential pathogenicity and relevance to both endometrial and colorectal cancer. RESULTS: Our analysis revealed 1,118 germline and 14,051 somatic variants, with 493 variants common to both. Recurrent pathogenic mutations in MLH1, MSH2, and MSH6 were confirmed, highlighting their critical role in Lynch syndrome. Notably, frequent somatic mutations in the PIK3CA and PTEN genes were identified, implicating the PI3K/AKT/mTOR pathway as a key oncogenic driver in these cancers. Additionally, novel somatic mutations in genes related to the extracellular matrix such as FBN1 and SPARC were uncovered, suggesting a possible unique role in endometrial tumor progression. CONCLUSIONS: This study provides new insights into the molecular basis of Lynch syndrome-associated endometrial cancer, emphasizing the overlap in oncogenic pathways with colorectal cancer. The discovery of shared and unique genetic mutations highlights the importance of developing combined treatment strategies and suggests that targeting these specific mutations could improve therapy for patients with Lynch syndrome-associated cancers.


Assuntos
Neoplasias Colorretais Hereditárias sem Polipose , Neoplasias do Endométrio , Sequenciamento do Exoma , Mutação em Linhagem Germinativa , Humanos , Neoplasias Colorretais Hereditárias sem Polipose/genética , Feminino , Neoplasias do Endométrio/genética , Pessoa de Meia-Idade , Predisposição Genética para Doença , Idoso , Proteína 2 Homóloga a MutS/genética , Mutação , Proteína 1 Homóloga a MutL/genética , PTEN Fosfo-Hidrolase/genética , Reparo de Erro de Pareamento de DNA/genética , Classe I de Fosfatidilinositol 3-Quinases/genética , Adulto , Proteínas de Ligação a DNA
17.
Sci Rep ; 14(1): 20859, 2024 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-39242811

RESUMO

Mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) gastric cancer (GC) exhibits an immune-active tumor microenvironment (TME) compared to MMR proficient (pMMR)/microsatellite stable/Epstein-Barr virus-negative [EBV (-)] GC. The tumor cell-intrinsic cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has been considered a key regulator of immune cell activation in the TME. However, its significance in regulating the immune-active TME in dMMR/MSI-H GC remains unclear. Here, we demonstrated that tumor cell-intrinsic cGAS-STING was highly expressed in dMMR GC compared to pMMR/EBV (-) GC. The expression of tumor cell-intrinsic STING was significantly and positively associated with the number of CD8+ tumor-infiltrating lymphocytes in GC. Analysis of TCGA datasets revealed that the expression of interferon-stimulated genes and STING downstream T-cell attracting chemokines was significantly higher in MSI-H GC compared to other subtypes of GC with EBV (-). These results suggest that tumor cell-intrinsic STING signaling plays a key role in activating immune cells in the dMMR/MSI-H GC TME and might serve as a novel biomarker predicting the efficacy of immunotherapy for GC treatment.


Assuntos
Linfócitos T CD8-Positivos , Linfócitos do Interstício Tumoral , Proteínas de Membrana , Instabilidade de Microssatélites , Transdução de Sinais , Neoplasias Gástricas , Microambiente Tumoral , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Microambiente Tumoral/imunologia , Masculino , Feminino , Reparo de Erro de Pareamento de DNA/genética , Pessoa de Meia-Idade , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Regulação Neoplásica da Expressão Gênica , Idoso
18.
Turk J Gastroenterol ; 35(9): 718-725, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39344689

RESUMO

DNA mismatch repair (MMR) proteins are essential for repairing genetic mutations that occur during DNA replication. Deficiency of MMR proteins results in a phenotype called microsatellite instability (MSI), which occurs in Lynch syndrome as well as sporadic colorectal cancers (CRC), and it is associated with several clinicopathological features. We aimed to investigate the association of the loss of MMR proteins with clinicopathologic considerations in our CRC series. In this retrospective study, DNA MMR protein status in CRC is evaluated in a total of 200 colorectal resection specimens by immunohistochemistry (IHC) for MLH1, MSH2, MSH6 and PMS2 protein expression. The BRAF mutation was investigated by the real-time PCR in cases with loss of MLH1 protein expression. The relationship between MMR status and clinicopathological parameters was investigated statistically. Loss of MMR protein expression was detected in 26 of 200 CRC cases. The BRAFV600E mutation was detected in 2 of the cases with MLH1 loss and accepted as sporadic. The remaining 24 cases (12%) were identified as Lynch syndrome candidates. There were statistical differences observed regarding the presence of tumor-infiltrating lymphocytes (P < .001), Crohn's-like reaction (P = .001), expansile growth (P < .001), tumor heterogeneity (P < .001), mucinous differentiation (P < .001), and presence of metastatic lymph nodes (P = .045) between sporadic cases with preserved MMR and Lynch candidates. However, difference in the survival rates between sporadic cases and Lynch candidates was not significant. Immunohistochemical staining for MMR is a practical method for predicting MSI phenotype as well as Lynch candidates. MMR expression status was found to be associated with certain clinicopathological features some of which also have prognostic significance.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais Hereditárias sem Polipose , Reparo de Erro de Pareamento de DNA , Imuno-Histoquímica , Instabilidade de Microssatélites , Proteína 1 Homóloga a MutL , Mutação , Proteínas Proto-Oncogênicas B-raf , Humanos , Reparo de Erro de Pareamento de DNA/genética , Masculino , Feminino , Estudos Retrospectivos , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas B-raf/genética , Idoso , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Proteína 1 Homóloga a MutL/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/patologia , Adulto , Proteínas de Ligação a DNA/genética , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Idoso de 80 Anos ou mais , Endonuclease PMS2 de Reparo de Erro de Pareamento/genética
19.
Dis Colon Rectum ; 67(11): 1413-1422, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-39260435

RESUMO

BACKGROUND: The use of programmed death-1 blockade has a significant therapeutic effect in patients with mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. However, data on preoperative single-agent programmed death-1 blockade are rare. OBJECTIVE: This study aims to evaluate the effectiveness and safety of preoperative programmed death-1 blockade as a conversion strategy in patients with locally advanced and resectable metastatic mismatch repair-deficient/microsatellite instability-high colorectal cancer. DESIGN: This is a retrospective observational study. SETTINGS: This study was conducted at a high-volume tertiary referral cancer center in China. PATIENTS: Twenty-four patients of consecutive cases since 2020 to 2022 with mismatch repair-deficient/microsatellite instability-high colorectal cancer who received preoperative single-agent programmed death-1 blockade were retrospectively reviewed. These patients had either bulking tumors scheduled for multivisceral resection, a strong desire for organ preservation, or potentially resectable metastatic lesions. MAIN OUTCOME MEASURES: Pathological complete response, clinical complete response, toxicity, R0 resection rate, and complications were evaluated. RESULTS: Patients tolerated preoperative immunotherapy well. The R0 resection rate was 95.2%, and the pathological complete response rate was 47.6%. Three patients (12.5%) were evaluated as having a clinical complete response and then underwent "watch and wait." One-half of the patients with cT4b were spared multivisceral resection, whereas 60% (3/5) achieved pathological complete response. All 3 patients with liver metastases obtained complete response of all liver lesions after programmed death-1 blockade treatment. Grade III postoperative complications occurred in 2 patients. LIMITATIONS: The limitations of this study are as follows: retrospective study, small sample size, and short follow-up. CONCLUSIONS: Preoperative anti-programmed death-1 therapy alone as a conversion strategy in initially resected difficult mismatch repair-deficient/microsatellite instability-high colorectal cancer can achieve a high tumor complete response. The use of immunopreoperative therapy in patients with T4b colon cancer or low rectal cancer can reduce multivisceral resection and achieve high organ function preservation. See the Video Abstract . INMUNOTERAPIA NEOADYUVANTE SOLA PARA PACIENTES CON CNCER COLORRECTAL LOCALMENTE AVANZADO Y METASTSICO RESECABLE CON ESTADO DMMR/MSIH: ANTECEDENTES:El uso del bloqueo de muerte programada-1 tiene un efecto terapéutico significativo en pacientes con cáncer colorrectal metastásico deficiente en reparación de desajustes/inestabilidad de microsatélites-alta (dMMR/MSI-H). Sin embargo, los datos sobre el bloqueo preoperatorio de muerte programada-1 con un solo agente son escasos.OBJETIVO:Este estudio tiene como objetivo evaluar la eficacia y seguridad del bloqueo preoperatorio de muerte programada-1 como estrategia de conversión en pacientes con cáncer colorrectal localmente avanzado y metastásico resecable con dMMR/MSI-H.DISEÑO:Este es un estudio observacional retrospectivo.ESCENARIO:Este estudio se realizó en un centro oncológico terciario de referencia de gran volumen en China.PACIENTES:Se revisaron retrospectivamente veinticuatro pacientes de casos consecutivos desde 2020-2022 con cáncer colorrectal y dMMR/MSI-H que recibieron bloqueo preoperatorio de muerte programada-1 con un solo agente. Estos pacientes tenían un tumor voluminoso programado para resección multivisceral, un fuerte deseo de preservación del órgano o lesiones metastásicas potencialmente resecables.PRINCIPALES MEDIDAS DE RESULTADO:Se evaluaron la respuesta patológica completa, la respuesta clínica completa, la toxicidad, la tasa de resección R0 y las complicaciones.RESULTADOS:Los pacientes toleraron bien la inmunoterapia preoperatoria. La tasa de resección R0 fue del 95,2% y la tasa de respuesta patológica completa fue del 47,6%. Tres pacientes (12,5%) fueron evaluados como respuesta clínica completa y luego sometidos a "observar y esperar". La mitad de los pacientes cT4b se salvaron de la resección multivisceral, mientras que el 60% (3/5) lograron una respuesta patológica completa. Los tres pacientes con metástasis hepáticas obtuvieron respuesta completa de todas las lesiones hepáticas después del tratamiento de bloqueo de muerte programada-1. En dos pacientes se produjeron complicaciones postoperatorias de grado III.LIMITACIONES:Las limitaciones de este estudio son las siguientes: estudio retrospectivo, tamaño de muestra pequeño y seguimiento corto.CONCLUSIONES:La terapia preoperatoria anti muerte programada-1 sola como estrategia de conversión en el cáncer colorrectal inicialmente difícil de resecar con dMMR/MSI-H puede lograr una alta respuesta completa tumoral. El uso de terapia inmunopreoperatoria en pacientes con cáncer de colon T4b o cáncer de recto bajo puede reducir la resección multivisceral y lograr una alta preservación de la función del órgano. (Traducción-Dr. Felipe Bellolio ).


Assuntos
Neoplasias Colorretais , Instabilidade de Microssatélites , Terapia Neoadjuvante , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Estudos Retrospectivos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Idoso , Terapia Neoadjuvante/métodos , Reparo de Erro de Pareamento de DNA , Inibidores de Checkpoint Imunológico/uso terapêutico , Adulto , Imunoterapia/métodos
20.
J Clin Lab Anal ; 38(15-16): e25085, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39132875

RESUMO

BACKGROUND: This study evaluated the performance of the PCR-HRM assay by comparing it with immunohistochemistry (IHC) for mismatch repair (MMR) proteins and the PCR capillary electrophoresis (PCR-CE) methods. RESULTS: A total of 224 patients with colorectal cancer participated in the study, with nearly half having mismatch repair deficiency (dMMR) tissues and the remainder possessing pMMR tissues. There was a 97.77% concordance between the PCR-HRM assay and IHC, and a 97.56% concordance between PCR-HRM and the PCR-CE assay. In comparison with IHC for dMMR proteins, the PCR-HRM demonstrated a sensitivity of 96.36% and a specificity of 99.12%. When juxtaposed with the PCR-CE assay, its sensitivity was 98.96% and specificity stood at 96.33%. The mutations observed in the microsatellite loci were uniformly distributed across all eight loci. Discrepant outcomes were more frequent in instances of MLH1 and PMS2 deficiency. Furthermore, the germline mutation status of MLH1, MSH2, PMS2, and MSH6 in 62 patients was ascertained using next-generation sequencing. All patients displaying MMR gene pathogenic mutations (N = 14) were identified as MSI-H by PCR-HRM, whereas those with MSS tissues (N = 43) did not exhibit MMR gene pathogenic mutations. Thus, the PCR-HRM method proficiently pinpoints tumors with verified germline MMR mutations, indicative of Lynch syndrome. CONCLUSION: Conclusively, the PCR-HRM assay emerges as a swift and congruent diagnostic tool for microsatellite instability, boasting commendable sensitivity and specificity in colorectal cancer.


Assuntos
Neoplasias Colorretais , Instabilidade de Microssatélites , Reação em Cadeia da Polimerase , Sensibilidade e Especificidade , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/diagnóstico , Feminino , Pessoa de Meia-Idade , Masculino , Reação em Cadeia da Polimerase/métodos , Idoso , Adulto , Reparo de Erro de Pareamento de DNA/genética , Idoso de 80 Anos ou mais , Eletroforese Capilar/métodos , Imuno-Histoquímica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA