1
|
Orhan A, Justesen TF, Raskov H, Qvortrup C, Gögenur I. Introducing Neoadjuvant Immunotherapy for Colorectal Cancer: Advancing the Frontier. Ann Surg 2025; 281:95-104. [PMID: 39005208 DOI: 10.1097/sla.0000000000006443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
OBJECTIVE To give surgeons a review of the current and future use of neoadjuvant immunotherapy in patients with localized colorectal cancer (CRC). BACKGROUND Immunotherapy has revolutionized the standard of care in oncology and improved survival outcomes in several cancers. However, the applicability of immunotherapy is still an ongoing challenge. Some cancer types are less responsive to immunotherapy, and the heterogeneity in responses within cancer types is poorly understood. Clinical characteristics of the patient, the timing of immunotherapy in relation to surgery, diversities in the immune responses, clonal heterogeneity, different features of the tumor microenvironment, and genetic alterations are some factors among many that may influence the efficacy of immunotherapy. RESULTS In this narrative review, we describe the major types of immunotherapy used to treat localized CRC. Furthermore, we discuss the prediction of response to immunotherapy in relation to biomarkers and radiologic assessment. Finally, we consider the future perspectives of clinical implications and response patterns, as well as the potential and challenges of neoadjuvant immunotherapy in localized CRC. CONCLUSIONS Establishing mismatch repair (MMR) status at the time of diagnosis is central to the potential use of neoadjuvant immunotherapy, in particular immune checkpoint inhibitors, in localized CRC. To date, efficacy is primarily seen in patients with deficient MMR status and polymerase epsilon mutations, although a small group of patients with proficient MMR does respond. In conclusion, neoadjuvant immunotherapy shows promising complete response rates, which may open a future avenue of an organ-sparing watch-and-wait approach for a group of patients.
Collapse
Affiliation(s)
- Adile Orhan
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Tobias F Justesen
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Hans Raskov
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Camilla Qvortrup
- Department of Clinical Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Ismail Gögenur
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Holla VR, Kahle MP, Kim SH, Ronaghy A, Yang RK, Patel KP, Routbort MJ, Overman MJ, Dumbrava EE, Shaw KRM, Karp DD, Meric-Bernstam F. Genomic Alterations in DNA Mismatch Repair Genes Across Different Cancer Types. JCO Precis Oncol 2024; 8:e2400419. [PMID: 39576951 PMCID: PMC11825140 DOI: 10.1200/po-24-00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/11/2024] [Accepted: 10/09/2024] [Indexed: 11/24/2024] Open
Abstract
PURPOSE PD-1 inhibition is effective in patients with mismatch repair deficient (dMMR) solid tumors in a tumor-agnostic fashion. However, dMMR testing by immunohistochemistry (IHC) is not routinely performed across tumor types. By contrast, next-generation sequencing (NGS) for somatic genomic alterations is frequently performed across tumor types. We hypothesized that NGS would identify patients with alterations in mismatch repair (MMR) genes and that these patients would have higher rates of MMR protein loss by IHC. This would support the utility of IHC reflex testing after NGS and potential matching to approved therapeutic options. METHODS From January 2016 to December 2021, 15,701 patients with solid tumors received NGS covering the MMR genes, and 4,994 patients had both IHC and NGS. Sequencing results were analyzed for mutations in MMR genes, tumor type distribution, and concordance with IHC results when available. RESULTS Six hundred and ninety-eight (4.4%) of 15,701 patients had mutations in one of the MMR genes. Mutations were found across tumor types. Three hundred and seventeen (6.3%) of 4,994 patients displayed IHC loss for at least one MMR protein. 33.8% patients (110/325) patients with MMR mutations had dMMR, compared with just 4.4% (207/4,669) patients without mutations (P < .001); dMMR rate varied by mutation type. CONCLUSION Mutations in MMR genes are found in multiple tumor types where IHC testing is not routine. Reflex IHC testing of patients carrying MMR gene mutations, especially those known or inferred to be inactivating, may identify more patients with dMMR and matched treatment options. However, dedicated IHC screening is needed to capture majority of the patients.
Collapse
Affiliation(s)
- Vijaykumar R. Holla
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Michael P Kahle
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Sun-Hee Kim
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Arash Ronaghy
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Richard K Yang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Keyur P Patel
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Mark J Routbort
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Kenna R Mills Shaw
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| |
Collapse
|
3
|
Hassan S, Mirza T, Khatoon A, Bukhari U, Shaikh F, Karim A. BRAF mutations and the association of V600E with CD133 and CDX2 expression in a Pakistani colorectal carcinoma cohort. BMC Cancer 2024; 24:1162. [PMID: 39300378 DOI: 10.1186/s12885-024-12925-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Despite a high incidence of colorectal carcinoma, data regarding genetic aberrations in colorectal carcinoma (CRC) patients in Pakistan is scarce. This study aimed to determine the frequency of BRAFV600E mutations in colorectal carcinoma tissue in the Pakistani population and to associate BRAFV600E expression with CD133, a marker of colorectal stem cells, and CDX2 marker of differentiation. METHODS Sanger Sequencing of exon 15 (426 bp) including the hotspot V600E was performed on formalin-fixed-paraffin-embedded (FFPE) CRC tissue samples of 115 patients. The samples were subjected to immunohistochemistry (IHC) to assess the expression of BRAFV600E, CDX2, and CD133. Additionally, homology modelling and docking were performed to investigate novel deletions revealed in sequencing. RESULTS Twenty-four (20.8%) BRAF variants were identified in the coding region, with V600E mutations detected in 14 (12.2% )cases (GenBank: PP003258.1; Pop Set: 2678087296). Moreover, a wide spectrum of novel non-V600E mutations (8.6%) were identified, including deletions and missense variations. In-silico analysis revealed that due to large deletions in the coding region of three samples, the affinity of the anti-BRAF drugs (Encorafenib and Vemurafenib) for the active site decreased in comparison to the wild type. The IHC analysis showed that BRAFV600E expression was significantly associated with CD133 expression (χ2(1, n=115) = 26.351; p = < 0.001) and with CDX2 expression (χ2(1, n=115) = 14.88; p = 0.001). Multivariate analysis using binary logistic regression revealed association of BRAFV600E mutations with age (OR = 1.123; CI = 1.024-1.232; p = 0.014), gender (OR = 0.071; CI = 0.006-0.831; p = 0.035), grade (0.007; CI = 0-0.644) and CD133 expression (OR = 65.649; CI = 2.153-2001.556; p = 0.016). CONCLUSION The present study demonstrates a notably high V600E frequency (12.2%) in comparison to global reported data, which ranges from 0.4 to 18%. This finding reflects the importance of upfront BRAF testing of the genetically distinct population of Pakistan. Previously unreported mutations identified in the sample may be of clinical significance and warrant further investigation. The concomitant high expression and significant association between CD133 and BRAFV600E represent vital actionable genes that may be targeted together to improve CRC patient management.
Collapse
Affiliation(s)
- Sobia Hassan
- Department of Pathology, Ziauddin Medical University, Karachi, 75000, Pakistan
| | - Talat Mirza
- Research Department, Ziauddin Medical University Karachi, Karachi, 75000, Pakistan
| | - Ambrina Khatoon
- Department of Molecular Medicine, Ziauddin Medical University Karachi, 4/B Shahrah-e-Ghalib Road, Block 6 Clifton, Karachi, 75000, Pakistan.
| | - Uzma Bukhari
- Department of Pathology, Dow University of Health Sciences Karachi, Karachi, 74200, Pakistan
| | - Fouzia Shaikh
- Department of Pathology, Ziauddin Medical University, Karachi, 75000, Pakistan
| | - Asad Karim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, Jamil-ur-Rahman Center for Genome Research, University of Karachi, Karachi, 75270, Pakistan
| |
Collapse
|
4
|
Chen H, Jiang RY, Hua Z, Wang XW, Shi XL, Wang Y, Feng QQ, Luo J, Ning W, Shi YF, Zhang DK, Wang B, Jie JZ, Zhong DR. Comprehensive analysis of gene mutations and mismatch repair in Chinese colorectal cancer patients. World J Gastrointest Oncol 2024; 16:2673-2682. [PMID: 38994136 PMCID: PMC11236251 DOI: 10.4251/wjgo.v16.i6.2673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/23/2024] [Accepted: 04/12/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND RAS, BRAF, and mismatch repair (MMR)/microsatellite instability (MSI) are crucial biomarkers recommended by clinical practice guidelines for colorectal cancer (CRC). However, their characteristics and influencing factors in Chinese patients have not been thoroughly described. AIM To analyze the clinicopathological features of KRAS, NRAS, BRAF, and PIK3CA mutations and the DNA MMR status in CRC. METHODS We enrolled 2271 Chinese CRC patients at the China-Japan Friendship Hospital. MMR proteins were tested using immunohistochemical analysis, and the KRAS/NRAS/BRAF/PIK3CA mutations were determined using quantitative polymerase chain reaction. Microsatellite status was determined using an MSI detection kit. Statistical analyses were conducted using SPSS software and logistic regression. RESULTS The KRAS, NRAS, BRAF, and PIK3CA mutations were detected in 44.6%, 3.4%, 3.7%, and 3.9% of CRC patients, respectively. KRAS mutations were more likely to occur in patients with moderate-to-high differentiation. BRAF mutations were more likely to occur in patients with right-sided CRC, poorly differentiated, or no perineural invasion. Deficient MMR (dMMR) was detected in 7.9% of all patients and 16.8% of those with mucinous adenocarcinomas. KRAS, NRAS, BRAF, and PIK3CA mutations were detected in 29.6%, 1.1%, 8.1%, and 22.3% of patients with dMMR, respectively. The dMMR was more likely to occur in patients with a family history of CRC, aged < 50 years, right-sided CRC, poorly differentiated histology, no perineural invasion, and with carcinoma in situ, stage I, or stage II tumors. CONCLUSION This study analyzed the molecular profiles of KRAS, NRAS, BRAF, PIK3CA, and MMR/MSI in CRC, identifying key influencing factors, with implications for clinical management of CRC.
Collapse
Affiliation(s)
- Huang Chen
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Rui-Ying Jiang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Zhan Hua
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiao-Wei Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiao-Li Shi
- Department of Scientific Research, Geneis, Beijing 100012, China
| | - Ye Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Qian-Qian Feng
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jie Luo
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wu Ning
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yan-Fen Shi
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Da-Kui Zhang
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Bei Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jian-Zheng Jie
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ding-Rong Zhong
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
5
|
Chen H, Jiang RY, Hua Z, Wang XW, Shi XL, Wang Y, Feng QQ, Luo J, Ning W, Shi YF, Zhang DK, Wang B, Jie JZ, Zhong DR. Comprehensive analysis of gene mutations and mismatch repair in Chinese colorectal cancer patients. World J Gastrointest Oncol 2024; 16:2661-2670. [DOI: 10.4251/wjgo.v16.i6.2661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/23/2024] [Accepted: 04/12/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND RAS, BRAF, and mismatch repair (MMR)/microsatellite instability (MSI) are crucial biomarkers recommended by clinical practice guidelines for colorectal cancer (CRC). However, their characteristics and influencing factors in Chinese patients have not been thoroughly described.
AIM To analyze the clinicopathological features of KRAS, NRAS, BRAF, and PIK3CA mutations and the DNA MMR status in CRC.
METHODS We enrolled 2271 Chinese CRC patients at the China-Japan Friendship Hospital. MMR proteins were tested using immunohistochemical analysis, and the KRAS/NRAS/BRAF/PIK3CA mutations were determined using quantitative polymerase chain reaction. Microsatellite status was determined using an MSI detection kit. Statistical analyses were conducted using SPSS software and logistic regression.
RESULTS The KRAS, NRAS, BRAF, and PIK3CA mutations were detected in 44.6%, 3.4%, 3.7%, and 3.9% of CRC patients, respectively. KRAS mutations were more likely to occur in patients with moderate-to-high differentiation. BRAF mutations were more likely to occur in patients with right-sided CRC, poorly differentiated, or no perineural invasion. Deficient MMR (dMMR) was detected in 7.9% of all patients and 16.8% of those with mucinous adenocarcinomas. KRAS, NRAS, BRAF, and PIK3CA mutations were detected in 29.6%, 1.1%, 8.1%, and 22.3% of patients with dMMR, respectively. The dMMR was more likely to occur in patients with a family history of CRC, aged < 50 years, right-sided CRC, poorly differentiated histology, no perineural invasion, and with carcinoma in situ, stage I, or stage II tumors.
CONCLUSION This study analyzed the molecular profiles of KRAS, NRAS, BRAF, PIK3CA, and MMR/MSI in CRC, identifying key influencing factors, with implications for clinical management of CRC.
Collapse
Affiliation(s)
- Huang Chen
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Rui-Ying Jiang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Zhan Hua
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiao-Wei Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiao-Li Shi
- Department of Scientific Research, Geneis, Beijing 100012, China
| | - Ye Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Qian-Qian Feng
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jie Luo
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wu Ning
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yan-Fen Shi
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Da-Kui Zhang
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Bei Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jian-Zheng Jie
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ding-Rong Zhong
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
6
|
El Zaitouni S, Laraqui A, Ghaouti M, Benzekri A, Kettani F, Bajjou T, Sekhsokh Y, Benmokhtar S, Jafari M, Baba W, Oukabli M, El Annaz H, Abi R, Tagajdid MR, El Kochri S, Lahlou IA, Ameziane El Hassani R, Ennibi K. KRAS, NRAS and BRAF Mutational Profile of Colorectal Cancer in a Series of Moroccan Patients. Cancer Control 2024; 31:10732748241262179. [PMID: 38875469 PMCID: PMC11179507 DOI: 10.1177/10732748241262179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024] Open
Abstract
OBJECTIVES The present study aimed to evaluate the frequencies of KRAS, NRAS, and BRAF mutations and their possible associations with clinicopathological features in 249 Moroccan patients with colorectal cancer (CRC). METHODS A retrospective investigation of a cohort of formalin-fixed paraffin-embedded tissues of 249 patients with CRC was screened for KRAS/NRAS/BRAF mutations using Idylla™ technology and pyrosequencing. RESULTS KRAS, NRAS, and BRAF mutations were revealed in 46.6% (116/249), 5.6% (14/249), and 2.4% (6/249) of patients. KRAS exon 2 mutations were identified in 87.9% of patients (102/116). KRAS G12D and G12 C were the most frequent, at 32.8% and 12.93%, respectively. Among the patients with KRAS exon 2 wild-type (wt), 27.6% (32/116) harbored additional KRAS mutations. Concurrent KRAS mutations were identified in 9.5% (11/116); including six in codon 146 (A146P/T/V), three in codon 61 (Q61H/L/R), one in codon 12 (G12 A and Q61H), and one in codon 13 (G13D and Q61 L). Among the NRAS exon 2 wt patients, 64.3% (9/14) harbored additional NRAS mutations. Concurrent NRAS mutations were identified in 28.6% (4/14) of NRAS-mutant patients. Since 3.2% wt KRAS were identified with NRAS mutations, concomitant KRAS and NRAS mutations were identified in 2.4% (6/249) of patients. KRAS mutations were higher in the >50-year-old age-group (P = .031), and the tumor location was revealed to be significantly associated with KRAS mutations (P = .028) predominantly in left colon (27.5%) and colon (42.2%) locations. NRAS mutations were most prevalent in the left colon (42.8%) and in well-differentiated tumors (64.2%). CONCLUSION Detection of KRAS mutations, particularly the G12 C subtype, may be significant for patients with CRC and has possible therapeutic implications. However, rare KRAS concomitant mutations in CRC patients suggest that each individual may present distinct therapeutic responses. KRAS testing alongside the identification of other affected genes in the same patient will make the treatments even more personalized by contributing more accurately to the clinical decision process. Overall, early diagnosis using novel molecular techniques may improve the management of CRC by providing the most efficient therapies for Moroccan patients.
Collapse
Affiliation(s)
- Sara El Zaitouni
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital of Rabat, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Abdelilah Laraqui
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Meriem Ghaouti
- Department of Pathology, Nations-Unites Pathology Center, Rabat, Morocco
| | - Asmae Benzekri
- Department of Pathology, Nations-Unites Pathology Center, Rabat, Morocco
| | - Fouad Kettani
- Department of Pathology, Nations-Unites Pathology Center, Rabat, Morocco
| | - Tahar Bajjou
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital of Rabat, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Yassine Sekhsokh
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital of Rabat, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Soukaina Benmokhtar
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital of Rabat, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Meryem Jafari
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital of Rabat, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Walid Baba
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
- Laboratory of Research and Biosafety P3, Mohammed V Military Teaching Hospital of Rabat, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Mohamed Oukabli
- Department of Pathology, Mohammed V Military Teaching Hospital of Rabat, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Hicham El Annaz
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Rachid Abi
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Mohamed Rida Tagajdid
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Safae El Kochri
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Idriss Amine Lahlou
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Rabii Ameziane El Hassani
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Khalid Ennibi
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious, and Tropical Diseases, Mohammed V Military Teaching Hospital, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
7
|
Zeng J, Fan W, Li J, Wu G, Wu H. KRAS/NRAS Mutations Associated with Distant Metastasis and BRAF/PIK3CA Mutations Associated with Poor Tumor Differentiation in Colorectal Cancer. Int J Gen Med 2023; 16:4109-4120. [PMID: 37720173 PMCID: PMC10503567 DOI: 10.2147/ijgm.s428580] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/30/2023] [Indexed: 09/19/2023] Open
Abstract
Background The occurrence, progression, and prognosis of colorectal cancer (CRC) are regulated by EGFR-mediated signaling pathways. However, the relationship between the core genes (KRAS/NRAS/BRAF/PIK3CA) status in the signaling pathways and clinicopathological characteristics of CRC patients in Hakka population remains controversial. Methods Patients were genotyped for KRAS (codons 12, 13, 61, 117, and 146), NRAS (codons 12, 61, 117, and 146), BRAF (codons 600), and PIK3CA (codons 542, 545 and 1047) mutations. Clinical records were collected, and clinicopathological characteristic associations were analyzed together with mutations of studied genes. Results Four hundred and eight patients (256 men and 152 women) were included in the analysis. At least one mutation in the four genes was detected in 216 (52.9%) patients, while none was detected in 192 (47.1%) patients. KRAS, NRAS, BRAF, and PIK3CA mutation status were detected in 190 (46.6%), 11 (2.7%), 10 (2.5%), 34 (8.3%) samples, respectively. KRAS exon 2 had the highest proportion (62.5%). Age, tumor site, tumor size, lymphovascular invasion, and perineural invasion were not associated with gene mutations. KRAS mutations (adjusted OR 1.675, 95% CI 1.017-2.760, P=0.043) and NRAS mutations (adjusted OR 5.183, 95% CI 1.239-21.687, P=0.024) appeared more frequently in patients with distant metastasis. BRAF mutations (adjusted OR 7.224, 95% CI 1.356-38.488, P=0.021) and PIK3CA mutations (adjusted OR 3.811, 95% CI 1.268-11.455, P=0.017) associated with poorly differentiated tumor. Conclusion KRAS/NRAS mutations are associated with distant metastasis and BRAF/PIK3CA mutations are associated with poor tumor differentiation in CRC. And the results provided a better understanding between clinicopathological characteristics and gene mutations in CRC patients.
Collapse
Affiliation(s)
- Juanzi Zeng
- Department of Medical Oncology, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
- Center for Precision Medicine, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Wenwei Fan
- Department of Gastroenterology, Dongguan Eighth People’s Hospital, Dongguan, People’s Republic of China
| | - Jiaquan Li
- Department of Medical Oncology, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
- Center for Precision Medicine, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Guowu Wu
- Department of Medical Oncology, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
- Center for Precision Medicine, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Heming Wu
- Center for Precision Medicine, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| |
Collapse
|
8
|
Afrăsânie VA, Marinca MV, Gafton B, Alexa-Stratulat T, Rusu A, Froicu EM, Sur D, Lungulescu CV, Popovici L, Lefter AV, Afrăsânie I, Ivanov AV, Miron L, Rusu C. Clinical, Pathological and Molecular Insights on KRAS, NRAS, BRAF, PIK3CA and TP53 Mutations in Metastatic Colorectal Cancer Patients from Northeastern Romania. Int J Mol Sci 2023; 24:12679. [PMID: 37628868 PMCID: PMC10454287 DOI: 10.3390/ijms241612679] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Mutations in RAS, BRAF, PIK3CA, and TP53 are well-established genetic abnormalities in metastatic colorectal cancer (mCRC). However, limited information is available for patients from Eastern Europe, including Romania. In this retrospective analysis, we investigated 104 mCRC patients from the Northeastern region of Romania to determine the frequency, distribution, coexistence, and clinicopathological and molecular correlations of these mutations. TP53 was the most frequently mutated gene (73.1%), followed by KRAS (45.2%) and PIK3CA (6.7%). Patients with KRAS mutant tumors and wild-type TP53 genotype were found to have no personal history of gastrointestinal cancer (p = 0.02, p = 0.007). KRAS mutations in exon 3 were associated with the female gender (p = 0.02) and the absence of lymph node invasion (p = 0.02). PIK3CA mutations were linked to the absence of lymph node invasion (p = 0.006). TP53 mutations were associated with KRAS mutations in exon 2 (p = 0.006), ulcerated histopathologic type (p = 0.04), and G2 differentiation (p = 0.01). It provides novel insights into genetic variations specific to the population from Northeastern Romania, which has been underrepresented in previous studies within Eastern Europe. Furthermore, our findings enable the development of genetic profiles in a developing country with limited access to specialized genetic tests and facilitate comparisons with other populations.
Collapse
Affiliation(s)
- Vlad-Adrian Afrăsânie
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
- Department of Oncology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihai-Vasile Marinca
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
- Department of Oncology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Gafton
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
- Department of Oncology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Teodora Alexa-Stratulat
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
- Department of Oncology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alexandra Rusu
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
| | - Eliza-Maria Froicu
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
- Department of Oncology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Daniel Sur
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
- 11th Department of Medical Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | | | - Larisa Popovici
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
| | - Andrei-Vlad Lefter
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
| | - Irina Afrăsânie
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, 700111 Iasi, Romania;
| | - Anca-Viorica Ivanov
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Lucian Miron
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania (A.R.); (A.-V.L.); (L.M.)
- Department of Oncology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristina Rusu
- Department of Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
9
|
Eefsen RL, Larsen JS, Klarskov LL, Altaf R, Høgdall E, Ingeholm P, Lykke J, Nielsen DL, Pfeiffer P, Poulsen LØ, Qvortrup C, Schou JV, Mau-Sørensen M, Østerlind K, Jensen BV. Therapy with pembrolizumab in treatment-naïve patients with nonmetastatic, mismatch repair deficient colorectal cancer. Int J Cancer 2023; 152:2145-2152. [PMID: 36594580 DOI: 10.1002/ijc.34420] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023]
Abstract
Therapy with immune checkpoint inhibitors (ICI) is effective in patients with metastatic mismatch-repair deficient (dMMR) colorectal cancer (CRC); however, data on treatment with neoadjuvant ICI in patients with locally advanced CRC are limited. From March 2019 to June 2020, five Danish oncological centers treated 10 patients with a treatment-naïve dMMR CRC with preoperative pembrolizumab, 9 with a nonmetastatic, unresectable colon cancer and 1 with a locally advanced rectum cancer. All 10 patients were evaluated regularly at a multidisciplinary team (MDT) meeting, and they all had a radical resection after a median of 8 cycles (range 2-13) of pembrolizumab. A microscopic evaluation of the resected tumors revealed no remaining tumor cells in five patients, while five still had tumor cells present. The patients were given no additional therapy. No recurrences were reported after a median follow-up of 26 months (range 23-38.5 months). Biopsies from Danish patients with CRC are routinely screened for dMMR proteins. In 2017, data from the Danish Colorectal Cancer Group showed that 19% (565/3000) of the patients with colon cancer and 1.5% (19/1279) of those with rectum cancer had an dMMR tumor. Among the patients with MMR determination, 26% (99/384) patients had a T4 dMMR colon cancer; thus, the 10 patients treated with neoadjuvant pembrolizumab comprised about 9% of the patients with a T4 dMMR colon cancer (9/99) and 5% of patients with dMMR rectal cancer (1/19). Therapy with pembrolizumab was feasible and effective. Larger prospective trials are needed to confirm our findings.
Collapse
Affiliation(s)
- Rikke Løvendahl Eefsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Jim S Larsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Louise L Klarskov
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Rahim Altaf
- Department of Oncology, Zealand University Hospital, Roskilde, Denmark
| | - Estrid Høgdall
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Peter Ingeholm
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Jakob Lykke
- Department of Surgery, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Dorte L Nielsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Laurids Ø Poulsen
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | - Camilla Qvortrup
- Department of Oncology, Copenhagen University Hospital-Rigshospitalet, København, Denmark
| | - Jakob V Schou
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Morten Mau-Sørensen
- Department of Oncology, Copenhagen University Hospital-Rigshospitalet, København, Denmark
| | - Kell Østerlind
- Department of Oncology, Northern Zealand University Hospital, Hillerød, Denmark
| | - Benny V Jensen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
10
|
Offermans K, Jenniskens JCA, Simons CCJM, Samarska I, Fazzi GE, van der Meer JRM, Smits KM, Schouten LJ, Weijenberg MP, Grabsch HI, van den Brandt PA. Association between mutational subgroups, Warburg-subtypes, and survival in patients with colorectal cancer. Cancer Med 2022; 12:1137-1156. [PMID: 35785488 PMCID: PMC9883416 DOI: 10.1002/cam4.4968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/24/2022] [Accepted: 06/11/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Previous research suggests that Warburg-subtypes are related to potentially important survival differences in colorectal cancer (CRC) patients. In the present study, we investigated whether mutational subgroups based on somatic mutations in RAS, BRAF, PIK3CA, and MET, which are known to promote the Warburg-effect, as well as mismatch repair (MMR) status, hold prognostic value in CRC. In addition, we investigated whether Warburg-subtypes provide additional prognostic information, independent of known prognostic factors like TNM stage. METHODS CRC patients (n = 2344) from the prospective Netherlands Cohort Study (NLCS) were classified into eight mutually exclusive mutational subgroups, based on observed mutations in RAS, BRAF, PIK3CA, and MET, and MMR status: All-wild-type + MMRproficient , KRASmut + MMRproficient , KRASmut + PIK3CAmut + MMRproficient , PIK3CAmut + MMRproficient , BRAFmut + MMRproficient , BRAFmut + MMRdeficient , other + MMRproficient , and other + MMRdeficient . Kaplan-Meier curves and Cox regression models were used to investigate associations between mutational subgroups and survival, as well as associations between our previously established Warburg-subtypes and survival within these mutational subgroups. RESULTS Compared to patients with all-wild-type + MMRproficient CRC, patients with KRASmut + MMRproficient , KRASmut + PIK3CAmut + MMRproficient , BRAFmut + MMRproficient , or other + MMRproficient CRC had a statistically significant worse survival (HRCRC-specific ranged from 1.29 to 1.88). In contrast, patients with other + MMRdeficient CRC had the most favorable survival (HRCRC-specific 0.48). No statistically significant survival differences were observed for the Warburg-subtypes within mutational subgroups. CONCLUSION Our results highlight the prognostic potential of mutational subgroups in CRC. Warburg-subtypes did not provide additional prognostic information within these mutational subgroups. Future larger-scale prospective studies are necessary to validate our findings and to examine the potential clinical utility of CRC subtyping based on mutational subgroups.
Collapse
Affiliation(s)
- Kelly Offermans
- Department of Epidemiology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Josien C. A. Jenniskens
- Department of Epidemiology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Colinda C. J. M. Simons
- Department of Epidemiology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Iryna Samarska
- Department of Pathology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Gregorio E. Fazzi
- Department of Pathology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Jaleesa R. M. van der Meer
- Department of Pathology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Kim M. Smits
- Department of Pathology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Leo J. Schouten
- Department of Epidemiology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Matty P. Weijenberg
- Department of Epidemiology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Heike I. Grabsch
- Department of Pathology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands,Pathology and Data Analytics, Leeds Institute of Medical Research at St James'sUniversity of LeedsLeedsUK
| | - Piet A. van den Brandt
- Department of Epidemiology, GROW School for Oncology and ReproductionMaastricht University Medical Center+MaastrichtThe Netherlands,Department of Epidemiology, Care and Public Health Research Institute (CAPHRI)Maastricht University Medical Center+MaastrichtThe Netherlands
| |
Collapse
|
11
|
Shetty O, Vengurlekar V, Kapoor A, Kamble V, Gurav M, Bhargava P, Srinivas S, Ramaswamy A, Ramadwar M, Saklani AP, Desouza A, Ostwal V. The Prevalence of BRAF, PIK3CA, and RAS Mutations in Indian Patients with Colorectal Cancer. South Asian J Cancer 2022; 11:190-194. [PMID: 36620501 PMCID: PMC9822779 DOI: 10.1055/s-0041-1740244] [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] [Indexed: 01/11/2023] Open
Abstract
Omshree ShettyVikas OstwalIntroduction The present study evaluates the mutation pattern and frequency of BRAF , PIK3CA and RAS in colorectal carcinoma observed in the tertiary cancer center in India. Materials and Methods Consecutive cases of colorectal adenocarcinoma ( n = 330) registered from January 2015 to December 2019 (5-year duration) were selected for the study. Molecular analysis for BRAF . PIK3CA (exon 9 and 20) and RAS ( KRAS & NRAS ) was performed on representative formalin-fixed paraffin-embedded tissues by Sanger sequencing. Results were correlated with clinicopathological features. Patient overall survival (OS) was obtained using Kaplan-Meier method. Results The study cohort was in the age range of 22 to 81 years (median age: 52 years) that included 202 males and 96 females (male: female ratio 2.1:1). BRAF V600E mutation was observed in three cases (1%), while 17 cases (5.7%) had mutations in the PIK3CA gene (exon 9 or exon 20). Mutation analysis for RAS gene ( KRAS & NRAS ) was observed among 42 (15.4%) cases with KRAS mutation and 11 (4%) cases were positive for NRAS mutations. Among RAS, KRAS G12D was the predominant mutation. Median OS with wild-type RAS was 46.6 months (95% confidence interval [CI]: 22.4-70.8), while for RAS mutated patients, it was 25.6 months (95% CI: 16.7-34.5), hazard ratio: 1.7 (95% CI: 1.1-2.7, p = 0.025). Conclusion This study evaluated the prevalence of BRAF, PIK3CA and RAS mutations in the Indian cohort and its impact on clinical behavior. There was lower incidence of BRAF mutations in this cohort and PIK3CA mutation (single) did not impact survival of the patients.
Collapse
Affiliation(s)
- Omshree Shetty
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vaibhavi Vengurlekar
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Akhil Kapoor
- Department of Medical Oncology, Mahamana Pandit Madan Mohan Malviya Cancer Center & Homi Bhabha Cancer Hospital, Tata Memorial Hospital, Varanasi, Uttar Pradesh, India
| | - Vishakha Kamble
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mamta Gurav
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Prabhat Bhargava
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sujay Srinivas
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mukta Ramadwar
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Avanish P. Saklani
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ashwin Desouza
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India,Address for correspondence Vikas Ostwal, MD DM Department of Medical Oncology, Tata Memorial HospitalDr. E Borges Road, Parel East, Mumbai, Maharashtra, 400012India
| |
Collapse
|
12
|
Wu W, Liu Y, Zeng S, Han Y, Shen H. Intratumor heterogeneity: the hidden barrier to immunotherapy against MSI tumors from the perspective of IFN-γ signaling and tumor-infiltrating lymphocytes. J Hematol Oncol 2021; 14:160. [PMID: 34620200 PMCID: PMC8499512 DOI: 10.1186/s13045-021-01166-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
In this era of precision medicine, with the help of biomarkers, immunotherapy has significantly improved prognosis of many patients with malignant tumor. Deficient mismatch repair (dMMR)/microsatellite instability (MSI) status is used as a biomarker in clinical practice to predict favorable response to immunotherapy and prognosis. MSI is an important characteristic which facilitates mutation and improves the likelihood of a favorable response to immunotherapy. However, many patients with dMMR/MSI still respond poorly to immunotherapies, which partly results from intratumor heterogeneity propelled by dMMR/MSI. In this review, we discuss how dMMR/MSI facilitates mutations in tumor cells and generates intratumor heterogeneity, especially through type II interferon (IFN-γ) signaling and tumor-infiltrating lymphocytes (TILs). We discuss the mechanism of immunotherapy from the perspective of dMMR/MSI, molecular pathways and TILs, and we discuss how intratumor heterogeneity hinders the therapeutic effect of immunotherapy. Finally, we summarize present techniques and strategies to look at the tumor as a whole to design personalized regimes and achieve favorable prognosis.
Collapse
Affiliation(s)
- Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
| | - Yihan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
13
|
Fan JZ, Wang GF, Cheng XB, Dong ZH, Chen X, Deng YJ, Song X. Relationship between mismatch repair protein, RAS, BRAF, PIK3CA gene expression and clinicopathological characteristics in elderly colorectal cancer patients. World J Clin Cases 2021; 9:2458-2468. [PMID: 33889611 PMCID: PMC8040173 DOI: 10.12998/wjcc.v9.i11.2458] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is common in elderly patients. Mismatch repair (MMR) protein deletion is one of the causes of CRC. The RAS (KRAS/NRAS), BRAF, and PIK3CA genes are important gene targets in CRC treatment and are closely related to the prognosis and survival of patients. However, little is known regarding the relationship between the expression of MMR, RAS, BRAF, PIK3CA and the clinicopathological features in CRC patients.
AIM To analyze the relationship between the expression of MMR, RAS, BRAF, PIK3CA and the clinicopathological features in CRC.
METHODS A total of 327 elderly patients with CRC were enrolled, and immuno-histochemistry was used to detect the MMR protein. Real-time quantitative polymerase chain reaction was used to detect the RAS (KRAS/NRAS), BRAF, and PIK3CA genes. The clinicopathological data of the patients were recorded and analyzed by SPSS 19.0 statistical software.
RESULTS In 327 elderly patients with CRC, the rate of MMR protein loss was 9.79% (32/327), and the deletion rate of four MMR proteins (MSH2, MSH6, MLH1, PMS2) was 1.83% (6/327), 3.06% (10/327), 7.65% (25/327), and 7.65% (25/327), respectively. There were no significant differences between MMR protein deletion and sex, pathological type, tumor morphology, differentiation degree or lymph node metastasis (P > 0.05), but there was a significant difference between MMR protein deletion and tumor diameter and tumor location (P = 0.048/P = 0.000). The mutation rates of the KRAS, NRAS, BRAF and PIK3CA genes in elderly CRC patients were 44.95% (147/327), 2.45% (8/327), 3.36% (11/327) and 2.75% (9/327), respectively; the KRAS gene mutation was closely related to tumor morphology (P = 0.002) but not to other clinicopathological features (P > 0.05), and there were no significant differences between NRAS gene mutation and clinicopathological features (P > 0.05). The BRAF gene mutation showed a significant difference in pathological type, tumor location, differentiation degree and lymph node metastasis (P < 0.05), but was not correlated with sex, tumor size and tumor morphology (P > 0.05). The PIK3CA gene mutation showed no significant differences in the above clinicopathological characteristics (P > 0.05). Significant differences were observed between MMR protein deletion and KRAS, BRAF, and PIK3CA gene mutations in elderly CRC patients (P = 0.044, P = 0.000, P = 0.003, respectively), but there was no significant difference between MMR protein deletion and NRAS mutation (P > 0.05).
CONCLUSION In elderly CRC patients, the tumor is mainly located in the right colon, and the deletion rate of MMR protein is higher when the tumor diameter is greater than or equal to 5 cm; the deletion rate of MLH1 and PMS2 is more common; the mutation rate of KRAS gene is higher than that of the NRAS, BRAF and PIK3CA genes, the BRAF gene mutation has different degrees of correlation with clinicopathological characteristics; when the MMR protein is deleted, the BRAF and PIK3CA gene mutations are often present, and the KRAS gene mutation rate is low.
Collapse
Affiliation(s)
- Jun-Zhen Fan
- Department of Pathology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Gao-Fei Wang
- Department of Pathology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Xue-Bin Cheng
- Department of Pathology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Zhou-Huan Dong
- Department of Pathology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Xin Chen
- Department of Pathology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Yu-Jiao Deng
- Department of Ultrasound, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Xin Song
- Department of Pathology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| |
Collapse
|