1
|
Boonsriroj H, Tangtrongsup S, Arunorat J, Mamom T, Chantawong P. Unveiling HER2 immunoexpression in canine hepatoid gland neoplasms: clinicopathological and morphological associations. Int J Vet Sci Med 2025; 13:1-12. [PMID: 40290666 PMCID: PMC12024500 DOI: 10.1080/23144599.2025.2495522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/18/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Canine hepatoid gland neoplasms (HGNs) are significant clinical concerns due to their high prevalence and diverse biological behaviour. Human epidermal growth factor receptor 2 (HER2), a tyrosine kinase receptor implicated in various aspects of tumorigenesis, has been extensively studied in human and animal neoplasms but remains unexplored in HGNs. This study aimed to assess HER2 immunoexpression in canine HGNs and its association with clinicopathological and morphological features. A total of 61 formalin-fixed paraffin-embedded samples, including normal hepatoid glands (n = 10), hepatoid gland adenomas (HGAs, n = 20), hepatoid gland epitheliomas (HGEs, n = 16), and hepatoid gland carcinomas (HGCs, n = 15), were analysed using immunohistochemistry. HER2 expression was scored based on percentage positivity and staining intensity. HER2-positive expression was detected in 50% of HGEs (score 2 + ) and 73.3% of HGCs, with 36.4% of cases scoring 3 + . In contrast, all HGAs and normal hepatoid tissues were HER2-immunonegative. Statistical analysis revealed significant differences in HER2 expression among normal and neoplastic hepatoid glands (p < 0.001). Only in HGCs, HER2 expression was significantly associated with tissue invasion (p = 0.007), mitotic count (p = 0.033), and nuclear pleomorphism (p = 0.007). These findings suggest that HER2 may play a role in the progression of malignant HGNs, particularly HGCs. This preliminary study highlights the potential of HER2 as a diagnostic marker and emphasizes the need for further investigation into its prognostic value and role in HER2-targeted therapy for canine HGCs.
Collapse
Affiliation(s)
- Hassadin Boonsriroj
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
- Animal Diagnostic Laboratory Center, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
| | - Sahatchai Tangtrongsup
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center of Producing and Development of Products and Innovations for Animal Health and Production, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jirapat Arunorat
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center for Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanongsak Mamom
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
- Animal Diagnostic Laboratory Center, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, Thailand
| | - Pinkarn Chantawong
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center of Producing and Development of Products and Innovations for Animal Health and Production, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center for Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
2
|
Liang W, Zhang Y, Li J, Ji C, Jiang X. Her-2 nanobody modified cisplatin nanoparticles for precise chemotherapy of colon cancer. J Microencapsul 2025:1-9. [PMID: 39976420 DOI: 10.1080/02652048.2025.2467060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
Construct a Her-2 nanobody modified nanoplatform as a versatile carrier of cisplatin and evaluate its anti-tumour effects. Size, morphology, cellular uptake, in vitro release, cell viability, bio-distribution and antitumor efficacy were respectively measured by dynamic light scattering, transmission electron microscopy, confocal microscopy, HPLC, MTT assay, ICP-Mass and tumour volume. Nb-CDDP NPs was prepared with average diameter 60.4 ± 8.4 nm, PDI 0.2 ± 0.02, Zeta potential -35.74 mV, entrapment efficiency 89.5%±0.8% and drug loading 28.7%±1.3% (w/w). From which cisplatin could release more rapidly in acidic solution. NPs could be easily phagocytised and exhibited stronger cytotoxic effect in HCT-116 cells with IC50 1.46 ± 0.019 μg/mL. The concentration of Nb-CDDP NPs in tumour and its inhibition ratio on tumour volume were both higher than without Nb modification, with hardly any influence on body weight. This cisplatin nanoplatform exhibits exceptional properties and high targeting anti-tumour efficacy in colon cancer cells and mice, which maybe provide a promising strategy for precise chemotherapy.
Collapse
Affiliation(s)
- Weina Liang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Yan Zhang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Jianpeng Li
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Chenglin Ji
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Xuexin Jiang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| |
Collapse
|
3
|
Dey S, Ghosh M, Dev A. Signalling and molecular pathways, overexpressed receptors of colorectal cancer and effective therapeutic targeting using biogenic silver nanoparticles. Gene 2025; 936:149099. [PMID: 39557372 DOI: 10.1016/j.gene.2024.149099] [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: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Increasing morbidity and mortality in CRC is a potential threat to human health. The major challenges for better treatment outcomes are the heterogeneity of CRC cases, complicated molecular pathway cross-talks, the influence of gut dysbiosis in CRC, and the lack of multimodal target-specific drug delivery. The overexpression of many receptors in CRC cells may pave the path for targeting them with multiple ligands. The design of a more target-specific drug-delivery device with multiple ligand-functionalized, green-synthesized silver nanoparticles is highly promising and may also deliver other approved chemotherapeutic agents. This review presents the various aspects of colorectal cancer and over-expressed receptors that can be targeted with appropriate ligands to enhance the specific drug delivery potency of green synthesised silver nanoparticles. This review aims to broaden further research into this multi-ligand functionalised, safer and effective silver nano drug delivery system.
Collapse
Affiliation(s)
- Sandip Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India.
| |
Collapse
|
4
|
Huang P, Wen F, Wang X. Case report: Pyrotinib and tegafur combined with radiotherapy achieved notable response in HER2-amplified rectal cancer with multiple metastases after multiline treatments. Front Pharmacol 2024; 15:1431542. [PMID: 39193330 PMCID: PMC11347432 DOI: 10.3389/fphar.2024.1431542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Metastatic colorectal cancer (mCRC) is characterized by significant phenotypic heterogeneity at the molecular level and presents a poor prognosis. Chemotherapy is commonly employed as the primary treatment option. Nevertheless, the advantages of chemotherapy are constrained, underscoring the critical necessity for novel treatment protocols aimed at enhancing patient outcomes. Human epidermal growth factor receptor 2 (HER2) has been recognized as a promising therapeutic target in mCRC. Pyrotinib, an innovative irreversible dual tyrosine kinase inhibitor targeting HER2, effectively inhibits cancer progression in various types of human cancers. Here, we present a case of a 39-year-old female with metastatic rectal cancer showing amplification of HER2. Despite resistance to multiple therapies, including trastuzumab and pertuzumab, the patient exhibited a remarkable therapeutic response to pyrotinib, tegafur combined with radiotherapy. This case provides evidence for the feasibility and potential efficacy of deploying pyrotinib in the salvage treatment of mCRC patients with HER2 amplification even though resistant to other anti-HER2 drugs.
Collapse
Affiliation(s)
- Peng Huang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Wen
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Wang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Amrell L, Bär E, Glasow A, Kortmann RD, Seidel C, Patties I. Enhanced anti-tumor effects by combination of tucatinib and radiation in HER2-overexpressing human cancer cell lines. Cancer Cell Int 2024; 24:277. [PMID: 39107782 PMCID: PMC11302197 DOI: 10.1186/s12935-024-03458-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Tucatinib (TUC), a HER2-directed tyrosine kinase inhibitor, is the first targeted drug demonstrating intracranial efficacy and significantly prolonged survival in metastatic HER2-positive breast cancer (BC) patients with brain metastases. Current treatments for brain metastases often include radiotherapy, but little is known about the effects of combination treatment with TUC. Therefore, we examined the combined effects of irradiation and TUC in human HER2-overexpressing BC, non-small cell lung cancer (NSCLC), and colorectal cancer (CRC) cell lines. For the latter two, a standard therapy successfully targeting HER2 is yet to be established. METHODS Nine HER2-overexpressing (BC: BT474, ZR7530, HCC1954; CRC: LS411N, DLD1, COLO201; NSCLC: DV90, NCI-H1781) and three control cell lines (BC: MCF7, HCC38; NSCLC: NCI-H2030) were examined. WST-1 assay (metabolic activity), BrdU ELISA (proliferation), γH2AX assay (DNA double-strand breaks (DSB), Annexin V assay (apoptosis), and clonogenic assay (clonogenicity) were performed after treatment with TUC and/or irradiation (IR). The relevance of the treatment sequence was analyzed exemplarily. RESULTS In BC, combinatorial treatment with TUC and IR significantly decreased metabolic activity, cell proliferation, clonogenicity and enhanced apoptotis compared to IR alone, whereby cell line-specific differences occurred. In the PI3KCA-mutated HCC1954 cell line, addition of alpelisib (ALP) further decreased clonogenicity. TUC delayed the repair of IR-induced DNA damage but did not induce DSB itself. Investigation of treatment sequence indicated a benefit of IR before TUC versus IR after TUC. Also in CRC and NSCLC, the combination led to a stronger inhibition of metabolic activity, proliferation, and clonogenic survival (only in NSCLC) than IR alone, whereby about 10-fold higher concentrations of TUC had to be applied than in BC to induce significant changes. CONCLUSION Our data indicate that combination of TUC and IR could be more effective than single treatment strategies for BC. Thereby, treatment sequence seems to be an important factor. The lower sensitivity to TUC in NSCLC and particularly in CRC (compared to BC) implicates, that tumor promotion there might be less HER2-related. Combination with inhibitors of other driver mutations may aid in overcoming partial TUC resistance. These findings are of high relevance to improve long-time prognosis especially in brain-metastasized situations given the intracranial activity of TUC.
Collapse
Affiliation(s)
- Lukas Amrell
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103, Leipzig, Germany
| | - Eric Bär
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103, Leipzig, Germany
| | - Annegret Glasow
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103, Leipzig, Germany
- Comprehensive Cancer Center Central Germany (CCCG), Leipzig, Germany
| | - Rolf-Dieter Kortmann
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103, Leipzig, Germany
| | - Clemens Seidel
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103, Leipzig, Germany
- Comprehensive Cancer Center Central Germany (CCCG), Leipzig, Germany
| | - Ina Patties
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103, Leipzig, Germany.
- Comprehensive Cancer Center Central Germany (CCCG), Leipzig, Germany.
| |
Collapse
|
6
|
Singh M, Morris VK, Bandey IN, Hong DS, Kopetz S. Advancements in combining targeted therapy and immunotherapy for colorectal cancer. Trends Cancer 2024; 10:598-609. [PMID: 38821852 DOI: 10.1016/j.trecan.2024.05.001] [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: 01/11/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/02/2024]
Abstract
Colorectal cancer (CRC) is a prevalent gastrointestinal cancer posing significant clinical challenges. CRC management traditionally involves surgery, often coupled with chemotherapy. However, unresectable or metastatic CRC (mCRC) presents a complex challenge necessitating innovative treatment strategies. Targeted therapies have emerged as the cornerstone of treatment in such cases, with interventions tailored to specific molecular attributes. Concurrently, immunotherapies have revolutionized cancer treatment by harnessing the immune system to combat malignant cells. This review explores the evolving landscape of CRC treatment, focusing on the synergy between immunotherapies and targeted therapies, thereby offering new avenues for enhancing the effectiveness of therapy for CRC.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Van Karlyle Morris
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irfan N Bandey
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
Suwatthanarak T, Tanjak P, Chaiboonchoe A, Acharayothin O, Thanormjit K, Chanthercrob J, Suwatthanarak T, Niyomchan A, Tanaka M, Okochi M, Pongpaibul A, Chalermwai WV, Trakarnsanga A, Methasate A, Pithukpakorn M, Chinswangwatanakul V. Overexpression of TSPAN8 in consensus molecular subtype 3 colorectal cancer. Exp Mol Pathol 2024; 137:104911. [PMID: 38861838 DOI: 10.1016/j.yexmp.2024.104911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/21/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Recently, consensus molecular subtypes (CMSs) have been proposed as a robust transcriptome-based classification system for colorectal cancer (CRC). Tetraspanins (TSPANs) are transmembrane proteins. They have been associated with the development of numerous malignancies, including CRC, through their role as "master organizers" for multi-molecular membrane complexes. No previous study has investigated the correlation between TSPANs and CMS classification. Herein, we investigated the expression of TSPANs in patient-derived primary CRC tissues and their CMS classifications. METHODS RNA samples were derived from primary CRC tissues (n = 100 patients diagnosed with colorectal adenocarcinoma) and subjected to RNA sequencing for transcriptome-based CMS classification and TSPAN-relevant analyses. Immunohistochemistry (IHC) and immunofluorescence (IF) stains were conducted to observe the protein expression level. To evaluate the relative biological pathways, gene-set enrichment analysis was performed. RESULTS Of the highly expressed TSPAN genes in CRC tissues (TSPAN8, TSPAN29, and TSPAN30), TSPAN8 was notably overexpressed in CMS3-classified primary tissues. The overexpression of TSPAN8 protein in CMS3 CRC was also observed by IHC and IF staining. As a result of gene-set enrichment analysis, TSPAN8 may potentially play a role in organizing signaling complexes for kinase-based metabolic deregulation in CMS3 CRC. CONCLUSIONS The present study reports the overexpression of TSPAN8 in CMS3 CRC. This study proposes TSPAN8 as a subtype-specific biomarker for CMS3 CRC. This finding provides a foundation for future CMS-based studies of CRC, a complex disease and the second leading cause of cancer mortality worldwide.
Collapse
Affiliation(s)
- Thanawat Suwatthanarak
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pariyada Tanjak
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Amphun Chaiboonchoe
- Siriraj Center of Systems Pharmacy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Onchira Acharayothin
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kullanist Thanormjit
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jantappapa Chanthercrob
- Siriraj Center of Systems Pharmacy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tharathorn Suwatthanarak
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Apichaya Niyomchan
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Masayoshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Kanagawa, Japan
| | - Mina Okochi
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Tokyo, Japan
| | - Ananya Pongpaibul
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wipapat Vicki Chalermwai
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Atthaphorn Trakarnsanga
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Asada Methasate
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Manop Pithukpakorn
- Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vitoon Chinswangwatanakul
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
8
|
Ramya S, Manivannan HP, Veeraraghavan VP, Francis AP. In Silico Analysis of Selective Bioactive Compounds from Acronychia Pedunculata as a Potential Inhibitor of HER2 in Colorectal Cancer. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S1281-S1286. [PMID: 38882725 PMCID: PMC11174219 DOI: 10.4103/jpbs.jpbs_570_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 06/18/2024] Open
Abstract
Colorectal cancer (CRC) is a pervasive malignancy that stands as a prominent contributor to global cancer-related mortality. Among the numerous causative factors, the overexpression of human epidermal growth factor receptor 2 (HER2) is notably linked to CRC progression. Acronychia (A.) pedunculata has a longstanding history in folk medicine due to its multifaceted medicinal attributes. This study aimed to assess the potential of specific bioactive compounds derived from A. pedunculata for their inhibition of HER2 in CRC, utilizing in silico analysis. The compounds were systematically evaluated through a series of computational analyses. Drug-likeness assessment, pharmacokinetic evaluation, and toxicity analysis were conducted. Molecular docking studies were performed to investigate binding affinities with the HER2 target. Additionally, bioavailability radar analysis was employed to predict oral bioavailability, while molecular target prediction provided insights into potential protein interactions. All 12 compounds demonstrated favorable drug-likeness properties and adherence to Lipinski's rule of five, indicative of the potential for good oral bioavailability. Four compounds were found to have no toxicological endpoints. Molecular docking revealed two compounds, namely caryophylla-4 (14), 8 (15)-dien-5alpha-ol and (-)-globulol, which showed promising binding affinities between several compounds and HER2. From this study, two leads were identified from A. pedunculata. Further experimental studies are required to validate the action of leads.
Collapse
Affiliation(s)
- S Ramya
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Hema P Manivannan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Vishnu P Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Arul P Francis
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
9
|
Connolly RM, Wang V, Hyman DM, Grivas P, Mitchell EP, Wright JJ, Sharon E, Gray RJ, McShane LM, Rubinstein LV, Patton DR, Williams PM, Hamilton SR, Wang J, Wisinski KB, Tricoli JV, Conley BA, Harris LN, Arteaga CL, O'Dwyer PJ, Chen AP, Flaherty KT. Trastuzumab and Pertuzumab in Patients with Non-Breast/Gastroesophageal HER2-Amplified Tumors: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol J. Clin Cancer Res 2024; 30:1273-1280. [PMID: 38433347 PMCID: PMC10984755 DOI: 10.1158/1078-0432.ccr-23-0633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/05/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE NCI-MATCH assigned patients with advanced cancer and progression on prior treatment, based on genomic alterations in pretreatment tumor tissue. Arm J (EAY131-J) evaluated the combination of trastuzumab/pertuzumab (HP) across HER2-amplified tumors. PATIENTS AND METHODS Eligible patients had high levels of HER2 amplification [copy number (CN) ≥7] detected by central next-generation sequencing (NGS) or through NCI-designated laboratories. Patients with breast/gastroesophageal adenocarcinoma and those who received prior HER2-directed therapy were excluded. Enrollment of patients with colorectal cancer was capped at 4 based on emerging data. Patients received HP IV Q3 weeks until progression or unacceptable toxicity. Primary endpoint was objective response rate (ORR); secondary endpoints included progression-free survival (PFS) and overall survival (OS). RESULTS Thirty-five patients were enrolled, with 25 included in the primary efficacy analysis (CN ≥7 confirmed by a central lab, median CN = 28). Median age was 66 (range, 31-80), and half of all patients had ≥3 prior therapies (range, 1-11). The confirmed ORR was 12% [3/25 partial responses (colorectal, cholangiocarcinoma, urothelial cancers), 90% confidence interval (CI) 3.4%-28.2%]. There was one additional partial response (urothelial cancer) in a patient with an unconfirmed ERBB2 copy number. Median PFS was 3.3 months (90% CI 2.0-4.1), and median OS 9.4 months (90% CI 5.0-18.9). Treatment-emergent adverse events were consistent with prior studies. There was no association between HER2 CN and response. CONCLUSIONS HP was active in a selection of HER2-amplified tumors (non-breast/gastroesophageal) but did not meet the predefined efficacy benchmark. Additional strategies targeting HER2 and potential resistance pathways are warranted, especially in rare tumors.
Collapse
Affiliation(s)
- Roisin M Connolly
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, Maryland
- Cancer Research @UCC, College of Medicine and Health, University College Cork, Ireland
| | - Victoria Wang
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Petros Grivas
- University of Washington, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Edith P Mitchell
- Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - John J Wright
- Investigational Drug Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Elad Sharon
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Robert J Gray
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - Lisa M McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Larry V Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - David R Patton
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, Maryland
| | - P Mickey Williams
- Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | - Jue Wang
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, Texas
| | - Kari B Wisinski
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - James V Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Barbara A Conley
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Lyndsay N Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Carlos L Arteaga
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, Texas
| | | | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| |
Collapse
|
10
|
Shebbo S, Binothman N, Darwaish M, Niaz HA, Abdulal RH, Borjac J, Hashem AM, Mahmoud AB. Redefining the battle against colorectal cancer: a comprehensive review of emerging immunotherapies and their clinical efficacy. Front Immunol 2024; 15:1350208. [PMID: 38533510 PMCID: PMC10963412 DOI: 10.3389/fimmu.2024.1350208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer globally and presents a significant challenge owing to its high mortality rate and the limitations of traditional treatment options such as surgery, radiotherapy, and chemotherapy. While these treatments are foundational, they are often poorly effective owing to tumor resistance. Immunotherapy is a groundbreaking alternative that has recently emerged and offers new hope for success by exploiting the body's own immune system. This article aims to provide an extensive review of clinical trials evaluating the efficacy of various immunotherapies, including CRC vaccines, chimeric antigen receptor T-cell therapies, and immune checkpoint inhibitors. We also discuss combining CRC vaccines with monoclonal antibodies, delve into preclinical studies of novel cancer vaccines, and assess the impact of these treatment methods on patient outcomes. This review seeks to provide a deeper understanding of the current state of CRC treatment by evaluating innovative treatments and their potential to redefine the prognosis of patients with CRC.
Collapse
Affiliation(s)
- Salima Shebbo
- Strategic Research and Innovation Laboratories, Taibah University, Madinah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, Beirut Arab University, Debbieh, Lebanon
| | - Najat Binothman
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Chemistry, College of Sciences and Arts, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Manar Darwaish
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Research Program, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Hanan A. Niaz
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Rwaa H. Abdulal
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jamilah Borjac
- Department of Biological Sciences, Beirut Arab University, Debbieh, Lebanon
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- Strategic Research and Innovation Laboratories, Taibah University, Madinah, Saudi Arabia
- College of Applied Medical Sciences, Taibah University, Almadinah Almunawarah, Saudi Arabia
| |
Collapse
|
11
|
Morgan J, Manickavel S, Sorace A, Hartman Y, Eli A, Massicano A, Gonzalez ML, Warram JM, Walsh E. Utility of Targeted Positron Emission Tomography Imaging to Predict Schwannoma Growth in a Murine Tumor Model. Laryngoscope 2024; 134:1372-1380. [PMID: 37578272 DOI: 10.1002/lary.30943] [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: 01/24/2023] [Revised: 05/31/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023]
Abstract
OBJECTIVE To identify if targeted positron emission tomography (PET) imaging with radiolabeled antibodies can predict tumor growth rate and ultimate tumor size in a murine flank schwannoma model. STUDY DESIGN Animal research study. METHODS Rat schwannoma cells were cultured and implanted into 40 athymic nude mice. Once tumors reached 5 mm in diameter, 30 mice were injected with zirconium-89 labeled antibodies (HER2/Neu, vascular endothelial growth factor receptor 2 (VEGFR2), or IgG isotype). PET/CT was performed, and standardized uptake values (SUV) were recorded. Tumors were serially measured until mice were sacrificed per IACUC protocol. Statistical analysis was performed to measure correlations between SUV values, tumor size, and growth. RESULTS Mean tumor sizes in mm3 on Day 0 were 144 ± 162 for anti-HER2/Neu, 212 ± 247 for anti-VEGFR2, and 172 ± 204 for IgG isotype groups respectively. Mean growth rates in mm3 /day were 531 ± 250 for HER2, 584 ± 188 for VEGFR2, and 416 ± 163 for the IgG isotype group. For both initial tumor size and growth rates, there was no significant difference between groups. There were significant correlations between maximum tumor volume and both the SUV max in the HER2 group (p = 0.0218, R2 = 0.5020), and we observed significant correlations between growth rate, and SUV values (p = 0.0156, R2 = 0.5394). Respectively, in the anti-VEGFR2 group, there were no significant correlations. CONCLUSION In a murine schwannoma model, immunotargeted PET imaging with anti-HER2/Neu antibodies predicted tumor growth rate and final tumor size. Laryngoscope, 134:1372-1380, 2024.
Collapse
Affiliation(s)
- Jake Morgan
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sudhir Manickavel
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anna Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yolanda Hartman
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abbigael Eli
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Adriana Massicano
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manuel Lora Gonzalez
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jason M Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Erika Walsh
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
12
|
Huang J, Chen G, Li H. An Update on the Role of Immunohistochemistry in the Evaluation of Pancreatic/Liver/Gastrointestinal Luminal Tract Disorders. Arch Pathol Lab Med 2023; 147:1374-1382. [PMID: 37134268 DOI: 10.5858/arpa.2022-0462-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2023] [Indexed: 05/05/2023]
Abstract
CONTEXT.— Immunohistochemistry serves as an ancillary diagnostic tool for a wide variety of neoplastic and nonneoplastic disorders, including infections, workup of inflammatory conditions, and subtyping neoplasms of the pancreas/liver/gastrointestinal luminal tract. In addition, immunohistochemistry is also used to detect a variety of prognostic and predictive molecular biomarkers for carcinomas of the pancreas, liver, and gastrointestinal luminal tract. OBJECTIVE.— To highlight an update on the role of immunohistochemistry in the evaluation of pancreatic/liver/gastrointestinal luminal tract disorders. DATA SOURCES.— Literature review and authors' research data and personal practice experience were used. CONCLUSIONS.— Immunohistochemistry is a valuable tool, assisting in the diagnosis of problematic tumors and benign lesions of the pancreas, liver, and gastrointestinal luminal tract, and also in the prediction of prognosis and therapeutic response for carcinomas of the pancreas, liver, and gastrointestinal luminal tract.
Collapse
Affiliation(s)
- Jialing Huang
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania (Huang, Li)
| | - Guoli Chen
- The Department of Pathology, PennState Health, Hershey, Pennsylvania (Chen)
| | - Hongjie Li
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania (Huang, Li)
| |
Collapse
|
13
|
Ullah A, Khan BM, Khan NU, Muntaha ST, Khan S, Khan AU, Almutairi MH, Ali I. Assessment of HER1 (rs11543848) and HER2 (rs1136201) polymorphism and their association with colorectal cancer susceptibility in Khyber Pakhtunkhwa, Pakistan. Mol Biol Rep 2023; 51:1. [PMID: 38040921 DOI: 10.1007/s11033-023-08943-y] [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: 08/15/2023] [Accepted: 10/30/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a widespread malignancy characterized by uncontrolled growth in the colon or rectum and remains a leading cause of cancer-related mortality globally. Various genes polymorphisms have been linked with the risk of CRC, but our study aimed to investigate the association between HER1 (rs11543848) and HER2 (rs1136201) polymorphisms with the risk of CRC in the Khyber Pakhtunkhwa (KPK) population of Pakistan. The association of the selected polymorphisms (rs11543848 and rs1136201) with CRC risk has been investigated in various ethnic groups, but their impact remains unexplored in Pakistan, particularly within the KPK population, highlighting the need of the study in this region. METHODS In this study 120 CRC patients and 120 healthy controls were enrolled. The DNA was extracted from the blood by salting-out method and genotyping was done using ARMS-PCR. RESULTS Our investigations provided convincing evidence of a strong association between HER1 (rs11543848) and the risk of CRC. Both the genotypes heterozygous GA (OR = 2.07, CI = 1.18 to 3.64, P = 0.01) and homozygous AA (OR = 6.22, CI = 2.56 to 15.08, P = 0.0001) showed higher risk and significant association with the CRC risk. Similarly, heterozygous genotype AG of HER2 (rs1136201) was significantly associated (OR = 3.16, 95% CI = 1.78 to 5.58, P = 0.0001) while mutant genotype GG showed higher risk but non-significant association (OR = 3.23, 95% CI = 0.84 to 12.43, P = 0.08) with CRC patients. HER1 (rs11543848) demonstrated a significant association (P = 0.003) with the age at diagnosis in CRC patients, while HER2 (rs1136201) showed a non-significant association (P = 0.434). Both the SNPs were non-significantly associated with gender (P = 0.793 and 0.117), metastasis (P = 0.582 and 0.129), location of the tumor (P = 0.555 and 0.993), tumor grade (P = 0.290 and 0.920), tumor size (P = 0.535 and 0.289) and stages of cancer (P = 0.892 and 0.352). CONCLUSION In conclusion, both the polymorphisms rs11543848 and rs1136201 displayed susceptibility with CRC in the KPK population. However, further investigations are recommended while using whole exome sequencing on a larger sample size for more precise results.
Collapse
Affiliation(s)
- Asad Ullah
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, Pakistan
| | | | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, Pakistan.
| | - Sidra Tul Muntaha
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Samiullah Khan
- Institute of Radiotherapy and Nuclear Medicine (IRNUM), Peshawar, Pakistan
| | - Aakif Ullah Khan
- Institute of Radiotherapy and Nuclear Medicine (IRNUM), Peshawar, Pakistan
| | - Mikhlid H Almutairi
- Zoology Department, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Hawally, Kuwait
| |
Collapse
|
14
|
Wu YJ, Lei J, Zhao J, Cao XW, Wang FJ. Design and characterization of a novel tumor-homing cell-penetrating peptide for drug delivery in TGFBR3 high-expressing tumors. Chem Biol Drug Des 2023; 102:1421-1434. [PMID: 37620132 DOI: 10.1111/cbdd.14333] [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: 02/22/2023] [Revised: 06/03/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
Targeted therapy has attracted more and more attention in cancer treatment in recent years. However, due to the diversity of tumor types and the mutation of target sites on the tumor surface, some existing targets are no longer suitable for tumor therapy. In addition, the long-term administration of a single targeted drug can also lead to drug resistance and attenuate drug potency, so it is important to develop new targets for tumor therapy. The expression of Type III transforming growth factor β receptor (TGFBR3) is upregulated in colon, breast, and prostate cancer cells, and plays an important role in the occurrence and development of these cancers, so TGFBR3 may be developed as a novel target for tumor therapy, but so far there is no report on this research. In this study, the structure of bone morphogenetic protein 4 (BMP4), one of the ligands of TGFBR3 was analyzed through the docking analysis with TGFBR3 and sequence charge characteristic analysis, and a functional tumor-targeting penetrating peptide T3BP was identified. The results of fluorescent labeling experiments showed that T3BP could target and efficiently enter tumor cells with high expression of TGFBR3, especially A549 cells. When the expression of TGFBR3 on the surface of tumor cells (HeLa) was knocked down by RNA interference, the high delivery efficiency of T3BP was correspondingly reduced by 40%, indicating that the delivery was TGFBR3-dependent. Trichosanthin (TCS, a plant-derived ribosome inactivating protein) fused with T3BP can enhance the inhibitory activity of the fusion protein on A549 cells by more than 200 times that of TCS alone. These results indicated that T3BP, as a novel targeting peptide that can efficiently bind TGFBR3 and be used for targeted therapy of tumors with high expression of TGFBR3. This study enriches the supply of tumor-targeting peptides and provides a new potential application option for the treatment of tumors with high expression of TGFBR3.
Collapse
Affiliation(s)
- Yi-Jie Wu
- Department of Applied Biology, East China University of Science and Technology, Shanghai, China
| | - Jin Lei
- Department of Applied Biology, East China University of Science and Technology, Shanghai, China
| | - Jian Zhao
- Department of Applied Biology, East China University of Science and Technology, Shanghai, China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, Shanghai, China
| | - Xue-Wei Cao
- Department of Applied Biology, East China University of Science and Technology, Shanghai, China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, Shanghai, China
| | - Fu-Jun Wang
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, Shanghai, China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd, Dongyang, China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Chitkara A, Bakhtiar M, Sahin IH, Hsu D, Zhang J, Anamika FNU, Mahnoor M, Ahmed R, Gholami S, Saeed A. A Meta-Analysis to Assess the Efficacy of HER2-Targeted Treatment Regimens in HER2-Positive Metastatic Colorectal Cancer (mCRC). Curr Oncol 2023; 30:8266-8277. [PMID: 37754515 PMCID: PMC10528053 DOI: 10.3390/curroncol30090600] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
Recent trials provide evidence that HER2 is a potential new target for patients with colorectal cancer. While HER2-positive tumors do not show a very encouraging response to anti-HER2-positive agents like trastuzumab alone, promising results have been observed when combined with other synergistically acting tyrosine kinase inhibitors (TKIs). Our meta-analysis was conducted following the Cochrane Handbook and written following the PRISMA guidelines. The protocol was registered on PROSPERO with the registration number CRD42022338935. After a comprehensive search for relevant articles, 14 CTs were identified and uploaded to Rayyan, and six trials were ultimately selected for inclusion. The meta-analysis revealed that a median of three prior lines of therapy was used before enrolling in the six trials comprising 238 patients with HER2-positive metastatic colorectal cancer (mCRC). The pooled objective response rate (ORR) and disease control rate (DCR) were 31.33% (95% confidence interval [CI] 24.27-38.39) and 74.37% (95% CI 64.57-84.17), respectively. The pooled weighted progression-free survival (PFS) was 6.2 months. The pooled ORR and DCR meta-analysis indicate a significant response to HER2-targeted therapy in this patient in HER2-positive mCRC. Additionally, a pooled PFS of 6.2 months suggests that HER2-targeted treatment regimens are associated with a meaningful improvement in survival outcomes in this population.
Collapse
Affiliation(s)
- Akshit Chitkara
- Department of Internal Medicine, University of California, Riverside, CA 92521, USA;
| | - Muhammad Bakhtiar
- School of Medicine, King Edward Medical University/Mayo Hospital, Lahore 54000, Pakistan; (M.B.); (R.A.)
| | - Ibrahim Halil Sahin
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA; (I.H.S.); (D.H.); (J.Z.)
| | - Dennis Hsu
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA; (I.H.S.); (D.H.); (J.Z.)
| | - Janie Zhang
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA; (I.H.S.); (D.H.); (J.Z.)
| | - FNU Anamika
- Department of Internal Medicine, Hackensack Meridian Ocean University Medical Center, Brick, NJ 08724, USA;
| | - Mahnoor Mahnoor
- School of Medicine, Mohtarma Benazir Bhutto Shaheed Medical College, Mirpur 10230, Pakistan;
| | - Rabeea Ahmed
- School of Medicine, King Edward Medical University/Mayo Hospital, Lahore 54000, Pakistan; (M.B.); (R.A.)
| | - Sepideh Gholami
- Department of Hematology-Oncology, Northwell Health Cancer Institute, New Hyde Park, NY 11042, USA;
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA; (I.H.S.); (D.H.); (J.Z.)
| |
Collapse
|
16
|
Ham-Karim H, Negm O, Ahmad N, Ilyas M. Investigating genomic, proteomic, and post-transcriptional regulation profiles in colorectal cancer: a comparative study between primary tumors and associated metastases. Cancer Cell Int 2023; 23:192. [PMID: 37670299 PMCID: PMC10478430 DOI: 10.1186/s12935-023-03020-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/06/2023] [Indexed: 09/07/2023] Open
Abstract
INTRODUCTION Approximately 50% of patients with primary colorectal carcinoma develop liver metastases. This study investigates the possible molecular discrepancies between primary colorectal cancer (pCRC) and their respective metastases. METHODS A total of 22 pairs of pCRC and metastases were tested. Mutation profiling of 26 cancer-associated genes was undertaken in 22/22primary-metastasis tumour pairs using next-generation sequencing, whilst the expression of a panel of six microRNAs (miRNAs) was investigated using qPCRin 21/22 pairs and 22 protein biomarkers was tested using Reverse Phase Protein Array (RPPA)in 20/22 patients' tumour pairs. RESULTS Among the primary and metastatic tumours the mutation rates for the individual genes are as follows:TP53 (86%), APC (44%), KRAS (36%), PIK3CA (9%), SMAD4 (9%), NRAS (9%) and 4% for FBXW7, BRAF, GNAS and CDH1. The primary-metastasis tumour mutation status was identical in 54/60 (90%) loci. However, there was discordance in heterogeneity status in 40/58 genetic loci (z-score = 6.246, difference = 0.3793, P < 0.0001). Furthermore, there was loss of concordance in miRNA expression status between primary and metastatic tumours, and 57.14-80.95% of the primary-metastases tumour pairs showed altered primary-metastasis relative expression in all the miRNAs tested. Moreover, 16 of 20 (80%) tumour pairs showed alteration in at least 3 of 6 (50%) of the protein biomarker pathways analysed. CONCLUSION The molecular alterations of primary colorectal tumours differ significantly from those of their matched metastases. These differences have profound implications for patients' prognoses and response to therapy.
Collapse
Affiliation(s)
- Hersh Ham-Karim
- Department of Pharmacy, College of Medicine, Komar University of Science and Technology, Chaq-Chaq-Qualaraisi, Sulaimani, Iraq.
| | - Ola Negm
- Division of Medical Sciences and Graduate Entry Medicine, Faculty of Medicine and Health Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Narmeen Ahmad
- Kurdistan Institution for Strategic Studies and Scientific Research, Qirga, Sulaimani, KRG, Iraq
| | - Mohammad Ilyas
- Department of Pharmacy, College of Medicine, Komar University of Science and Technology, Chaq-Chaq-Qualaraisi, Sulaimani, Iraq
- Nottingham Molecular Pathology Node, University of Nottingham, Nottingham, UK
| |
Collapse
|
17
|
Bartolini M, Seeber A, Puccini A. Editorial: New therapeutic approaches in microsatellite stable colorectal cancer patients. Front Oncol 2023; 13:1240963. [PMID: 37546398 PMCID: PMC10403228 DOI: 10.3389/fonc.2023.1240963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Affiliation(s)
- Michela Bartolini
- Scientific Institute for Research, Hospitalization and Healthcare (Istituto di Ricovero e Cura a Carattere Scientifico) (IRCCS) Humanitas Research Hospital, Humanitas Cancer Center, Medical Oncology and Haematology Unit, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Alberto Puccini
- Scientific Institute for Research, Hospitalization and Healthcare (Istituto di Ricovero e Cura a Carattere Scientifico) (IRCCS) Humanitas Research Hospital, Humanitas Cancer Center, Medical Oncology and Haematology Unit, Rozzano, Milan, Italy
| |
Collapse
|
18
|
Fraga T, de Sousa MJ, Magalhães J, Basto R, Paulo J, Bonito N, Magalhães JP, Figueiredo P, Sousa GM. HER2 Status in RAS and BRAF Wild-Type Metastatic Colorectal Cancer: A Portuguese Study. Cureus 2023; 15:e42536. [PMID: 37637599 PMCID: PMC10460123 DOI: 10.7759/cureus.42536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) is the second-most deadly cancer worldwide. However, there remains a scarcity of precision treatments available for this type of cancer. Amplification or overexpression of human epidermal growth factor receptor 2 (HER2+) is a well-established therapeutic target in gastric and breast cancer. HER2 is positive in approximately 5% of CRC cases and has been implicated in resistance to therapy with anti-epidermal growth factor receptor antibodies. The aim of this study was to evaluate HER2 status in RAS and BRAF wild-type metastatic CRC (mCRC) and its correlation with survival outcomes. MATERIALS AND METHODS A single-center retrospective analysis of RAS and BRAF wild-type mCRC patients undergoing systemic treatment was conducted from July 2014 to September 2020. Tissue HER2 status was determined by immunohistochemistry (IHC) and/or fluorescence in situ hybridization (FISH) and/or chromogenic in situ hybridization (CISH). HER2+ was defined as IHC3 (+) or IHC2 (+) through FISH or CISH (+). RESULTS Fifty-nine patients were included. The median age of all the included patients was 64 years (33-82). Four patients had HER2+ tumors (7%). Four patients had HER2+ tumors (7%). The majority of HER2+ mCRC cases were males (n=3) and left-sided CRC (n=3). All patients received FOLFIRI plus cetuximab as first-line treatment. At the median follow-up of 24.0 months, patients with HER2-negative mCRC presented with a median overall survival (mOS) of 39.4 months (95% confidence interval (CI) 32.7-46.0) and the four patients with HER2+ mCRC had a mOS of 20.4 months (95% CI; 9.5-31.3; p=0.07). In HER2-negative patients, the median PFS (mPFS) was 11.3 months (95% CI; 9.2-13.4) vsHER2-positive patients with a mPFS of 10.9 months (95% CI; 1.3-20.4; p=0.47). CONCLUSIONS To our knowledge, this is the first study reporting HER2+ in mCRC patients in a Portuguese population and the HER2+ rate was consistent with previous studies. Our study suggests that HER2+ may potentially be a marker that is able to predict poor prognosis in RAS and BRAF wild-type mCRC.
Collapse
Affiliation(s)
- Teresa Fraga
- Medical Oncology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, PRT
| | | | - Joana Magalhães
- Medical Oncology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, PRT
| | - Raquel Basto
- Medical Oncology, Centro Hospitalar Vila Nova de Gaia/Espinho, Gaia, PRT
| | - Judy Paulo
- Medical Oncology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, PRT
| | - Nuno Bonito
- Medical Oncology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, PRT
| | - José Paulo Magalhães
- Pathology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, PRT
| | - Paulo Figueiredo
- Pathology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, PRT
| | - Gabriela M Sousa
- Medical Oncology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, PRT
| |
Collapse
|
19
|
Manzi J, Hoff CO, Ferreira R, Pimentel A, Datta J, Livingstone AS, Vianna R, Abreu P. Targeted Therapies in Colorectal Cancer: Recent Advances in Biomarkers, Landmark Trials, and Future Perspectives. Cancers (Basel) 2023; 15:cancers15113023. [PMID: 37296986 DOI: 10.3390/cancers15113023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
In 2022, approximately 600,000 cancer deaths were expected; more than 50,000 of those deaths would be from colorectal cancer (CRC). The CRC mortality rate in the US has decreased in recent decades, with a 51% drop between 1976 and 2014. This drop is attributed, in part, to the tremendous therapeutic improvements, especially after the 2000s, in addition to increased social awareness regarding risk factors and diagnostic improvement. Five-fluorouracil, irinotecan, capecitabine, and later oxaliplatin were the mainstays of mCRC treatment from the 1960s to 2002. Since then, more than a dozen drugs have been approved for the disease, betting on a new chapter in medicine, precision oncology, which uses patient and tumor characteristics to guide the therapeutic choice. Thus, this review will summarize the current literature on targeted therapies, highlighting the molecular biomarkers involved and their pathways.
Collapse
Affiliation(s)
- Joao Manzi
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Camilla O Hoff
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Raphaella Ferreira
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Agustin Pimentel
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Jashodeep Datta
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Alan S Livingstone
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Rodrigo Vianna
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Phillipe Abreu
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
20
|
de Jong E, Kocer A. Current Methods for Identifying Plasma Membrane Proteins as Cancer Biomarkers. MEMBRANES 2023; 13:409. [PMID: 37103836 PMCID: PMC10142483 DOI: 10.3390/membranes13040409] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 06/19/2023]
Abstract
Plasma membrane proteins are a special class of biomolecules present on the cellular membrane. They provide the transport of ions, small molecules, and water in response to internal and external signals, define a cell's immunological identity, and facilitate intra- and intercellular communication. Since they are vital to almost all cellular functions, their mutants, or aberrant expression is linked to many diseases, including cancer, where they are a part of cancer cell-specific molecular signatures and phenotypes. In addition, their surface-exposed domains make them exciting biomarkers for targeting by imaging agents and drugs. This review looks at the challenges in identifying cancer-related cell membrane proteins and the current methodologies that solve most of the challenges. We classified the methodologies as biased, i.e., search cells for the presence of already known membrane proteins. Second, we discuss the unbiased methods that can identify proteins without prior knowledge of what they are. Finally, we discuss the potential impact of membrane proteins on the early detection and treatment of cancer.
Collapse
|
21
|
Chen PC, Yeh YM, Chu CT, Su PF, Chiu PH, Lin BW, Chen SH, Lin PC, Lee CT, Chen HHW, Chen CC. HER2 amplification in colorectal cancer with brain metastasis: A propensity score matching study. Eur J Cancer 2023; 181:62-69. [PMID: 36640475 DOI: 10.1016/j.ejca.2022.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND The association between human epidermal growth factor receptor-2 (HER2) amplification and brain metastasis (BM) in patients having colorectal cancer (CRC) has been suggested but not yet established. This study investigated the expression patterns of HER2, its association with BM, and its prognostic value in patients having CRC. METHODS We retrospectively identified 99 patients having metastatic CRC (mCRC) and BM (the BM cohort) and compared them with a cohort of 249 patients having mCRC and without BM (the stage IV cohort) by propensity score matching. Immunohistochemical studies of HER2 on all available paraffin-embedded tumour samples, either from the primary tumour, the metastasis (brain and/or extracranial sites) or both, were performed and analysed. HER2 fluorescent in situ hybridisation was applied when necessary. The expression of HER2 was compared and correlated with survival. RESULTS HER2 amplifications were detected in 16 (18.4%) of 87 and 9 (3.6%) of 249 patients who had specimens available in the BM and stage IV cohorts, respectively (P < .001). After propensity score matching, HER2 amplification was significantly associated with BM (odds ratio: 5.38, P = .003). HER2 heterogeneity was frequently observed not only at the single tumour level but also in paired tumour samples. A marginally significant longer survival since BM was found in patients having HER2-amplified mCRC than in those without (P = .07). CONCLUSIONS HER2 amplification was significantly associated with BM in patients having mCRC and might have prognostic value for survival since BM. Given the heterogeneity of HER2 expression, the testing of HER2 status on available tissues from both primary and metastatic tumours should be encouraged.
Collapse
Affiliation(s)
- Po-Chuan Chen
- Division of Colorectal Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Yu-Min Yeh
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Chun-Ting Chu
- Division of Colorectal Surgery, Department of Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Pei-Fang Su
- Department of Statistics, National Cheng Kung University, Tainan 701, Taiwan
| | - Pin-Hsuan Chiu
- The Center of Quantitative Sciences, Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Bo-Wen Lin
- Division of Colorectal Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Shang-Hung Chen
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan; National Institute of Cancer Research, National Health Research Institute, Tainan 704, Taiwan
| | - Peng-Chan Lin
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan; Department of Genomic Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan; Department of Computer Science and Information Engineering, College of Electrical Engineering and Computer Science, National Cheng Kung University, Tainan 704, Taiwan
| | - Chung-Ta Lee
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Helen H W Chen
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan.
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan; Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan; Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan; Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
22
|
Tang YL, Li DD, Duan JY, Sheng LM, Wang X. Resistance to targeted therapy in metastatic colorectal cancer: Current status and new developments. World J Gastroenterol 2023; 29:926-948. [PMID: 36844139 PMCID: PMC9950860 DOI: 10.3748/wjg.v29.i6.926] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/24/2022] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal and common malignancies in the world. Chemotherapy has been the conventional treatment for metastatic CRC (mCRC) patients. However, the effects of chemotherapy have been unsatisfactory. With the advent of targeted therapy, the survival of patients with CRC have been prolonged. Over the past 20 years, targeted therapy for CRC has achieved substantial progress. However, targeted therapy has the same challenge of drug resistance as chemotherapy. Consequently, exploring the resistance mechanism and finding strategies to address the resistance to targeted therapy, along with searching for novel effective regimens, is a constant challenge in the mCRC treatment, and it is also a hot research topic. In this review, we focus on the current status on resistance to existing targeted therapies in mCRC and discuss future developments.
Collapse
Affiliation(s)
- Yuan-Ling Tang
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Dan-Dan Li
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jia-Yu Duan
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lei-Ming Sheng
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin Wang
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
23
|
CAR-T cells for cancer immunotherapy. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
|
24
|
Vtorushin SV, Krakhmal NV, Zavalishina LE, Kuznetsova OA, Moskvina LV, Frank GA. [Assessment of HER2 status of carcinomas of various localizations]. Arkh Patol 2023; 85:31-46. [PMID: 38010637 DOI: 10.17116/patol20238506131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
A detailed description of the methodological aspects of the evaluation of HER2-status in carcinomas of such localizations as the mammary gland, pancreas, salivary glands, stomach, colon, endometrium, bladder, lungs is presented. Approaches and criteria for assessing HER2 status from methodological and clinical points of view are analyzed. The data are systematized in tables for use in practical diagnostic work.
Collapse
Affiliation(s)
- S V Vtorushin
- Siberian State Medical University, Tomsk, Russia
- Cancer Research Institute of Tomsk National Research Medical Center, Tomsk, Russia
| | - N V Krakhmal
- Siberian State Medical University, Tomsk, Russia
- Cancer Research Institute of Tomsk National Research Medical Center, Tomsk, Russia
| | - L E Zavalishina
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - O A Kuznetsova
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - L V Moskvina
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - G A Frank
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|
25
|
Failmezger H, Hessel H, Kapil A, Schmidt G, Harder N. Spatial heterogeneity of cancer associated protein expression in immunohistochemically stained images as an improved prognostic biomarker. Front Oncol 2022; 12:964716. [PMID: 36601480 PMCID: PMC9806230 DOI: 10.3389/fonc.2022.964716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
The identification of new tumor biomarkers for patient stratification before therapy, for monitoring of disease progression, and for characterization of tumor biology plays a crucial role in cancer research. The status of these biomarkers is mostly scored manually by a pathologist and such scores typically, do not consider the spatial heterogeneity of the protein's expression in the tissue. Using advanced image analysis methods, marker expression can be determined quantitatively with high accuracy and reproducibility on a per-cell level. To aggregate such per-cell marker expressions on a patient level, the expression values for single cells are usually averaged for the whole tissue. However, averaging neglects the spatial heterogeneity of the marker expression in the tissue. We present two novel approaches for quantitative scoring of spatial marker expression heterogeneity. The first approach is based on a co-occurrence analysis of the marker expression in neighboring cells. The second approach accounts for the local variability of the protein's expression by tiling the tissue with a regular grid and assigning local spatial heterogeneity phenotypes per tile. We apply our novel scores to quantify the spatial expression of four different membrane markers, i.e., HER2, CMET, CD44, and EGFR in immunohistochemically (IHC) stained tissue sections of colorectal cancer patients. We evaluate the prognostic relevance of our spatial scores in this cohort and show that the spatial heterogeneity scores clearly outperform the marker expression average as a prognostic factor (CMET: p-value=0.01 vs. p-value=0.3).
Collapse
|
26
|
Ivanova M, Venetis K, Guerini-Rocco E, Bottiglieri L, Mastropasqua MG, Garrone O, Fusco N, Ghidini M. HER2 in Metastatic Colorectal Cancer: Pathology, Somatic Alterations, and Perspectives for Novel Therapeutic Schemes. Life (Basel) 2022; 12:1403. [PMID: 36143438 PMCID: PMC9502498 DOI: 10.3390/life12091403] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 12/03/2022] Open
Abstract
HER2 is an emerging biomarker in colorectal cancer (CRC). This oncogene plays an essential role in regulating cell proliferation, differentiation, migration, and, more in general, tumorigenesis and tumor progression. The most frequent types of HER2 alterations in CRC include gene amplification and missense mutations in 7-8% of CRC, often being mirrored by HER2 protein overexpression, representing founder events in solid tumors, including CRC. There are currently no approved HER2-targeted therapy guidelines for CRC; however, several studies have shown that HER2 can be effectively targeted in meta-static CRC settings. In this review, we discuss the current knowledge of HER2 testing in CRC and the immediate future perspectives for HER2 targeting in the metastatic setting.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Luca Bottiglieri
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Mauro Giuseppe Mastropasqua
- Department of Emergency and Organ Transplantation, School of Medicine, University of Bari “Aldo Moro”, Piazza G Cesare, 11, 70124 Bari, Italy
| | - Ornella Garrone
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
27
|
Wang M, Wang X, Li Y, Li Q, Cai S, Li X, Ma M. HER2 status is positively associated with vessel invasion of colorectal cancer: a retrospective large cohort study. Int J Colorectal Dis 2022; 37:2061-2067. [PMID: 36006442 PMCID: PMC9436852 DOI: 10.1007/s00384-022-04243-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/15/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE HER2-positive colorectal cancer was drawn increasing attention in recent years. Accumulating evidence showed HER2-positive metastatic colorectal cancer could benefit from HER2-targeted therapy. While HER2 expression and the relationship between HER2 status and clinicopathological characteristics of overall colorectal cancer remains largely unknown. The aim of this study was to evaluate HER2 expression in colorectal cancer and compare the clinicopathological features between HER2-positive and HER2-negative colorectal cancer. METHODS We retrospectively analyzed 3910 primary colorectal cancer patients treated in our institution from January 2016 to December 2019. Medical records and pathology reports after surgery were collected to provide information about HER2 status and other clinicopathological characteristics. RESULTS We identified 3347 HER2-negative and 79 HER2-positive colorectal cancer patients in our cohort. The chi-square test showed that vessel invasion was significantly more common in HER2-positive colorectal cancer patients. Crude analysis showed HER2 positive was associated with vessel invasion in colorectal cancer [OR and 95% CI 0.534 (0.341, 0.835), p = 0.006]. After adjusting for N stage, a significant association was still observed between HER2 status and vessel invasion in colorectal cancer [OR and 95% CI 0.550 (0.322, 0.941), p = 0.029]. Survival analysis showed that there was no significant difference in 3-year overall survival rate between HER2 positive and HER2 negative group (p = 0.603). CONCLUSION Our findings indicate that the rate of HER2 positivity in colorectal cancer was relatively low, and HER2 status was strongly associated with vessel invasion while having no significant influence on the 3-year overall survival rate in colorectal cancer patients.
Collapse
Affiliation(s)
- Mingdian Wang
- Department of Pathology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Xiang Wang
- Department of General Surgery, the Second People’s Hospital of Hefei, Hefei Hospital Anhui Medical University, Hefei, Anhui Province, China
| | - Yiwei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Maoguang Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
28
|
Alharbi KS, Javed Shaikh MA, Afzal O, Alfawaz Altamimi AS, Almalki WH, Alzarea SI, Kazmi I, Al-Abbasi FA, Singh SK, Dua K, Gupta G. An overview of epithelial growth factor receptor (EGFR) inhibitors in cancer therapy. Chem Biol Interact 2022; 366:110108. [PMID: 36027944 DOI: 10.1016/j.cbi.2022.110108] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 12/28/2022]
Abstract
Epithelial growth factor receptor (EGFR), a transmembrane receptor on the cell surface, carries extracellular messages into the cell and alters the activity of the nucleus through tyrosine signalling. EGFR-targeted treatments have influenced the new era of precision oncology throughout the last few decades. Despite significant progress, long-term remission from solid tumours is still a distant goal for many oncologists. There are several methods by which tumour cells alter the activity of this protein in solid tumours. EGFR-related oncogenic pathways, resistance mechanisms, and novel avenues to suppress tumour development and metastatic spread were discovered in clinical specimens using preclinical models (cell cultures, xenografts, mouse models), which were then validated in those specimens. EGFR has been implicated in the onset and advancement of a variety of cancers, according to research. An overview of EGFR's structural anatomy and physiology, its role in cancers, and clinical studies that target EGFR in various tumours are included in this review.
Collapse
Affiliation(s)
- Khalid Saad Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | | | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | | | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia.
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
29
|
Torres-Jiménez J, Esteban-Villarrubia J, Ferreiro-Monteagudo R. Precision Medicine in Metastatic Colorectal Cancer: Targeting ERBB2 (HER-2) Oncogene. Cancers (Basel) 2022; 14:3718. [PMID: 35954382 PMCID: PMC9367374 DOI: 10.3390/cancers14153718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in terms of incidence rate in adults and the second most common cause of cancer-related death in Europe. The treatment of metastatic CRC (mCRC) is based on the use of chemotherapy, anti-vascular endothelial growth factor (VEGF), and anti-epidermal growth factor receptor (EGFR) for RAS wild-type tumors. Precision medicine tries to identify molecular alterations that could be treated with targeted therapies. ERBB2 amplification (also known as HER-2) has been identified in 2-3% of patients with mCRC, but there are currently no approved ERBB2-targeted therapies for mCRC. The purpose of this review is to describe the molecular structure of ERBB2, clinical features of these patients, diagnosis of ERBB2 alterations, and the most relevant clinical trials with ERBB2-targeted therapies in mCRC.
Collapse
Affiliation(s)
- Javier Torres-Jiménez
- Medical Oncology Department, MD Anderson Cancer Center Madrid, 28033 Madrid, Spain
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
| | - Jorge Esteban-Villarrubia
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
- Medical Oncology Department, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Reyes Ferreiro-Monteagudo
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
| |
Collapse
|
30
|
EGFR, HER2, and HER3 protein expression in paired primary tumor and lymph node metastasis of colorectal cancer. Sci Rep 2022; 12:12894. [PMID: 35902718 PMCID: PMC9334602 DOI: 10.1038/s41598-022-17210-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Due to the difficulty in sampling of metastatic tumors, patient selection is commonly based on results of primary tumor samples when metastatic samples are not available. However, due to tumor heterogeneity, metastatic tumors may be different from primary tumors in their phenotypes. The aim of this study was to investigate the expression of EGFR, HER2, and HER3 between primary and lymph node metastatic lesions of colorectal cancer. Paired primary tumors and lymph node metastases from 79 patients with colorectal cancer were retrospectively collected and analyzed for EGFR, HER2, and HER3 expression. High EGFR, HER2, and HER3 expression (2+ and 3+) was found in 64.2%, 66.0%, and 85.0% of primary tumors, and 56.8%, 46.0%, and 76.0% of lymph node metastases, respectively. Correlation rates between primary and metastatic lesions were 67.1%, 63.3%, and 74.7% for EGFR, HER2, and HER3, respectively. Stage IV tumors (with distant metastasis) had higher correlation rates of HER2 expression compared to stage III tumors (without distant metastasis) (P = 0.050). Moderate correlation rates in EGFR, HER2, and HER3 expression were observed between primary and metastatic lesions of colorectal cancer. Tumor stage or existence of distant metastasis could serve as potential predictive markers for the correlation of HER2 expression between primary tumors and lymph node metastases of colorectal cancer.
Collapse
|
31
|
Della Chiesa M, Setti C, Giordano C, Obino V, Greppi M, Pesce S, Marcenaro E, Rutigliani M, Provinciali N, Paleari L, DeCensi A, Sivori S, Carlomagno S. NK Cell-Based Immunotherapy in Colorectal Cancer. Vaccines (Basel) 2022; 10:1033. [PMID: 35891197 PMCID: PMC9323201 DOI: 10.3390/vaccines10071033] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
Human Natural Killer (NK) cells are all round players in immunity thanks to their powerful and immediate response against transformed cells and the ability to modulate the subsequent adaptive immune response. The potential of immunotherapies based on NK cell involvement has been initially revealed in the hematological setting but has inspired the design of different immune tools to also be applied against solid tumors, including colorectal cancer (CRC). Indeed, despite cancer prevention screening plans, surgery, and chemotherapy strategies, CRC is one of the most widespread cancers and with the highest mortality rate. Therefore, further efficient and complementary immune-based therapies are in urgent need. In this review, we gathered the most recent advances in NK cell-based immunotherapies aimed at fighting CRC, in particular, the use of monoclonal antibodies targeting tumor-associated antigens (TAAs), immune checkpoint blockade, and adoptive NK cell therapy, including NK cells modified with chimeric antigen receptor (CAR-NK).
Collapse
Affiliation(s)
- Mariella Della Chiesa
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Chiara Giordano
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Valentina Obino
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | | | | | - Laura Paleari
- A.Li.Sa., Liguria Region Health Authority, 16121 Genoa, Italy;
| | - Andrea DeCensi
- Medical Oncology, Galliera Hospital, 16128 Genoa, Italy; (N.P.); (A.D.)
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Simona Carlomagno
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| |
Collapse
|
32
|
Li C, Lin Q, Hu F, Bao R, Cai H, Gu Y. Based on lapatinib innovative near-infrared fluorescent probes targeting HER1/HER2 for in vivo tumors imaging. Biosens Bioelectron 2022; 214:114503. [DOI: 10.1016/j.bios.2022.114503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/05/2022] [Accepted: 06/21/2022] [Indexed: 11/02/2022]
|
33
|
Bischoff NS, Proquin H, Jetten MJ, Schrooders Y, Jonkhout MCM, Briedé JJ, van Breda SG, Jennen DGJ, Medina-Reyes EI, Delgado-Buenrostro NL, Chirino YI, van Loveren H, de Kok TM. The Effects of the Food Additive Titanium Dioxide (E171) on Tumor Formation and Gene Expression in the Colon of a Transgenic Mouse Model for Colorectal Cancer. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1256. [PMID: 35457963 PMCID: PMC9027218 DOI: 10.3390/nano12081256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022]
Abstract
Titanium dioxide (TiO2) is present in many different food products as the food additive E171, which is currently scrutinized due to its potential adverse effects, including the stimulation of tumor formation in the gastrointestinal tract. We developed a transgenic mouse model to examine the effects of E171 on colorectal cancer (CRC), using the Cre-LoxP system to create an Apc-gene-knockout model which spontaneously develops colorectal tumors. A pilot study showed that E171 exposed mice developed colorectal adenocarcinomas, which were accompanied by enhanced hyperplasia in epithelial cells, lymphatic nodules at the base of the polyps, and increased tumor size. In the main study, tumor formation was studied following the exposure to 5 mg/kgbw/day of E171 for 9 weeks (Phase I). E171 exposure showed a statistically nonsignificant increase in the number of colorectal tumors in these transgenic mice, as well as a statistically nonsignificant increase in the average number of mice with tumors. Gene expression changes in the colon were analyzed after exposure to 1, 2, and 5 mg/kgbw/day of E171 for 2, 7, 14, and 21 days (Phase II). Whole-genome mRNA analysis revealed the modulation of genes in pathways involved in the regulation of gene expression, cell cycle, post-translational modification, nuclear receptor signaling, and circadian rhythm. The processes associated with these genes might be involved in the enhanced tumor formation and suggest that E171 may contribute to tumor formation and progression by modulation of events related to inflammation, activation of immune responses, cell cycle, and cancer signaling.
Collapse
Affiliation(s)
- Nicolaj S. Bischoff
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Héloïse Proquin
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- National Institute for Public Health and Environment (RIVM), Bilthoven, 3721 MA De Bilt, The Netherlands
| | - Marlon J. Jetten
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- Faculty of Health, Medicine and Life Science, Maastricht University Medical Center, 6229 ES Maastricht, The Netherlands
| | - Yannick Schrooders
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- Laboratory of Biosignaling & Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Marloes C. M. Jonkhout
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- Laboratory of Biosignaling & Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Jacco J. Briedé
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Simone G. van Breda
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Danyel G. J. Jennen
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Estefany I. Medina-Reyes
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (E.I.M.-R.); (N.L.D.-B.); (Y.I.C.)
| | - Norma L. Delgado-Buenrostro
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (E.I.M.-R.); (N.L.D.-B.); (Y.I.C.)
| | - Yolanda I. Chirino
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (E.I.M.-R.); (N.L.D.-B.); (Y.I.C.)
| | - Henk van Loveren
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Theo M. de Kok
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| |
Collapse
|
34
|
Huang W, Chen Y, Chang W, Ren L, Tang W, Zheng P, Wu Q, Liu T, Liu Y, Wei Y, Xu J. HER2 positivity as a biomarker for poor prognosis and unresponsiveness to anti-EGFR therapy in colorectal cancer. J Cancer Res Clin Oncol 2022; 148:993-1002. [PMID: 34156520 DOI: 10.1007/s00432-021-03655-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/27/2021] [Indexed: 01/27/2023]
Abstract
PURPOSE To investigate the role of HER2 positivity in prognosis and unresponsiveness to anti-EGFR therapy for colorectal cancer. METHODS Patients who underwent primary CRC tumor resection were included. HER2 status of CRC was confirmed by immunohistochemistry and fluorescence in situ hybridization tests. Comparison of survival analysis between HER2 positivity and negativity was evaluated by a stratified log-rank test and summarized with the use of Kaplan-Meier and Cox proportional hazards methods. The treatment effects of cetuximab were further compared in full subgroup analyses. RESULT 1240 patients were enrolled, including 763 with stage I-III CRC and 477 with stage IV CRC. 57 (4.6%) CRC patients presented HER2 positivity in the entire cohort. The survival analysis showed that patients with HER2 positivity had significantly worse disease-free survival and overall survival in stage III and IV CRC. The multivariable analysis also confirmed that HER2 positivity was a significantly independent risk factor in stage III and IV CRC. Univariate and multivariable survival analysis showed no prognostic significance of HER2 positivity in stage I-II CRC patients. Prespecified subgroup analysis showed no favorable trends in progression-free survival and overall survival for cetuximab in the patients with HER2 positivity, KRAS/NRAS/BRAF wild-type metastatic CRC (interaction P values = 0.005 and 0.014). CONCLUSION For stage III and IV CRC patients, HER2 positivity was confirmed as an independent prognostic risk factor. It could help predict the unresponsiveness to anti-EGFR therapy for metastatic CRC.
Collapse
Affiliation(s)
- Wenbai Huang
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yijiao Chen
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenju Chang
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China
| | - Li Ren
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wentao Tang
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Zheng
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Wu
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianyu Liu
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Liu
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Wei
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianmin Xu
- Department of General Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, People's Republic of China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Schröder C, Gower-Page C, Reyes-Rivera I, Patel A, Price R, Sen S, Davies J, Castellanos E, Snider J, Bai X, Fisher V, Mhatre SK. Natural History of Human Epidermal Growth Factor Receptor 2-Amplified and Human Epidermal Growth Factor Receptor 2 Wild-Type Refractory Metastatic Colorectal Cancer in US Clinical Practice. JCO Clin Cancer Inform 2022; 6:e2100133. [PMID: 35297649 PMCID: PMC8955081 DOI: 10.1200/cci.21.00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The molecular heterogeneity of metastatic colorectal cancer (mCRC) presents a therapeutic challenge, with few trials focused on patients with human epidermal growth factor receptor 2 amplification (HER2-Amp). Our limited understanding of real-world patterns and outcomes by HER2 status of treatment-refractory patients leaves treatment decisions with little contextual information. We conducted a retrospective cohort study to describe the natural disease history of patients with refractory mCRC using an electronic health record-derived database with oncogenomic information. METHODS We included patients with stage IV or recurrent mCRC diagnosed from January 2011 through December 2019 from a deidentified clinicogenomic database. Patients with ≥ 2 documented clinic visits, ≥ 2 lines of therapy (LOT) after mCRC diagnosis, and comprehensive genomic profiling were eligible. Patient records defined by treatment-refractory LOT were allocated to the HER2-Amp or HER2 wild-type (WT) cohort on the basis of comprehensive genomic profiling. Index date was defined as the start of any treatment-refractory LOT (≥ 2 LOT; patients could contribute multiple records). Descriptive statistics included demographic and clinical characteristics, treatments, laboratory values, and biomarkers. Overall survival (OS) was calculated as time (in months) from the index date until death from any cause and analyzed using Kaplan-Meier methodology. Sensitivity analyses were conducted to test the robustness of the primary findings. RESULTS A total of 576 patients were included (1,339 records); 63 (158 records) were HER2-Amp, and 513 (1,181 records) were HER2-WT. Demographics, clinical characteristics, biomarkers, and laboratory values were comparable between HER2 cohorts. OS was similar, with an unadjusted median OS of 11.2 months (95% CI, 8.6 to 15.1) and 9.9 months (95% CI, 8.3 to 10.9) across LOT for HER2-Amp and HER2-WT cohorts, respectively. CONCLUSION This study showed considerable treatment heterogeneity and poor outcomes among patients with treatment-refractory mCRC, emphasizing a substantial unmet therapeutic need.
Collapse
Affiliation(s)
| | | | | | | | | | - Shiraj Sen
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
36
|
Kaur S, Gill KS, Manjari M, Kumar S, Nauhria S, Nath R, Patel C, Hamdan K, Jeong Y, Nayak NP, Maity S, Hilgers R, Nauhria S. Human Epidermal Growth Factor Receptor 2 (HER2) Expression in Colorectal Carcinoma: A Potential Area of Focus for Future Diagnostics. Cureus 2022; 14:e22811. [PMID: 35382213 PMCID: PMC8976506 DOI: 10.7759/cureus.22811] [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] [Accepted: 03/03/2022] [Indexed: 11/05/2022] Open
Abstract
Objective In this study, we aimed to explore the potential diagnostic utility of human epidermal growth factor receptor 2 (HER2) expression in colorectal carcinoma. We investigated the association of HER2 expression with the type and grade of the tumor along with the pattern, staining intensity, and the percentage of cells stained. Methods This was an observational study involving 50 cases of colorectal carcinoma that underwent immunohistochemistry to analyze the HER2 expression. Results The positive expression of HER2 was seen in 16 (32%) cases. The majority of the study population was between the fifth-seventh decades of life. The most commonly diagnosed tumor was conventional adenocarcinoma with grade II, cytoplasmic pattern, +2 positivity, and moderate intensity. The maximum positivity for HER2 was seen in tumors of the rectum in eight (16%) cases. Conclusion A substantial rate of HER2 overexpression paves the way for it to become a potential future target in cancer therapeutics.
Collapse
Affiliation(s)
- Simrandeep Kaur
- Department of Pathology, Sri Guru Ram Das Institute of Medical Sciences & Research, Amritsar, IND
| | - Karamjit S Gill
- Department of Pathology, Sri Guru Ram Das Institute of Medical Sciences & Research, Amritsar, IND
| | - Mridu Manjari
- Department of Pathology, Sri Guru Ram Das Institute of Medical Sciences & Research, Amritsar, IND
| | - Surinder Kumar
- Department of Community Medicine, Armed Forces Medical College, Pune, IND
| | - Shreya Nauhria
- Department of Psychology, University of Leicester, Leicester, GBR
| | - Reetuparna Nath
- Department of Educational Services, St. George's University, St. George's, GRD
| | - Chandni Patel
- Medical Student Research Institute, St. Matthew's University, Georgetown, CYM
| | - Kamal Hamdan
- Medical Student Research Institute, St. Matthew's University, Georgetown, CYM
| | - Yujin Jeong
- Internal Medicine, American University of Antigua, St. John's, ATG
| | - Narendra P Nayak
- Department of Microbiology, St. Matthew's University, Georgetown, CYM
| | - Sabyasachi Maity
- Department of Physiology, St. George's University School of Medicine, St. George's, GRD
| | - Rob Hilgers
- Department of Pharmacology, St. Matthew's University, Georgetown, CYM
| | - Samal Nauhria
- Department of Pathology, St. Matthew's University, Georgetown, CYM
| |
Collapse
|
37
|
Mir-4746 inhibits the proliferation of colorectal cancer cells in vitro and in vivo by targeting CCND1. Biochem Biophys Res Commun 2022; 594:153-160. [DOI: 10.1016/j.bbrc.2022.01.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/15/2022] [Indexed: 12/19/2022]
|
38
|
Riedesser JE, Ebert MP, Betge J. Precision medicine for metastatic colorectal cancer in clinical practice. Ther Adv Med Oncol 2022; 14:17588359211072703. [PMID: 35237350 PMCID: PMC8882813 DOI: 10.1177/17588359211072703] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022] Open
Abstract
Globally, metastatic colorectal cancer is one of the leading causes for cancer-related death. Treatment limited to conventional chemotherapeutics extended life for only a few months. However, advances in surgical approaches and medical treatment regimens have greatly increased survival, even leading to long-term remission in selected patients. Advances in multiomics analysis of tumors have built a foundation for molecular-targeted therapies. Furthermore, immunotherapies are on the edge of revolutionizing oncological practice. This review summarizes recent advances in the growing toolbox of personalized treatment for patients with metastatic colorectal cancer. We provide an overview of current multimodal therapy and explain novel immunotherapy and targeted therapy approaches in detail. We emphasize clinically relevant therapies, such as inhibitors of MAPK signaling, and give recommendations for clinical practice. Finally, we describe the potential predictive impact of molecular subtypes and provide an outlook on novel concepts, such as functional precision medicine.
Collapse
Affiliation(s)
- Julian E. Riedesser
- Junior Clinical Cooperation Unit Translational
Gastrointestinal Oncology and Preclinical Models, German Cancer Research
Center (DKFZ), Heidelberg, Germany
| | - Matthias P. Ebert
- Department of Medicine II, University Medical
Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim,
GermanyMannheim Cancer Center, University Medical Center Mannheim, Medical
Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Betge
- Junior Clinical Cooperation Unit Translational
Gastrointestinal Oncology and Preclinical Models, German Cancer Research
Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, GermanyDKFZ-Hector
Cancer Institute at University Medical Center Mannheim, Mannheim,
Germany.Department of Medicine II, University Medical Center Mannheim,
Medical Faculty Mannheim, Heidelberg University, Mannheim, GermanyMannheim
Cancer Center, University Medical Center Mannheim, Medical Faculty Mannheim,
Heidelberg University, Mannheim, Germany
| |
Collapse
|
39
|
Abstract
Peritoneal surface malignancies comprise a heterogeneous group of primary tumours, including peritoneal mesothelioma, and peritoneal metastases of other tumours, including ovarian, gastric, colorectal, appendicular or pancreatic cancers. The pathophysiology of peritoneal malignancy is complex and not fully understood. The two main hypotheses are the transformation of mesothelial cells (peritoneal primary tumour) and shedding of cells from a primary tumour with implantation of cells in the peritoneal cavity (peritoneal metastasis). Diagnosis is challenging and often requires modern imaging and interventional techniques, including surgical exploration. In the past decade, new treatments and multimodal strategies helped to improve patient survival and quality of life and the premise that peritoneal malignancies are fatal diseases has been dismissed as management strategies, including complete cytoreductive surgery embedded in perioperative systemic chemotherapy, can provide cure in selected patients. Furthermore, intraperitoneal chemotherapy has become an important part of combination treatments. Improving locoregional treatment delivery to enhance penetration to tumour nodules and reduce systemic uptake is one of the most active research areas. The current main challenges involve not only offering the best treatment option and developing intraperitoneal therapies that are equivalent to current systemic therapies but also defining the optimal treatment sequence according to primary tumour, disease extent and patient preferences. New imaging modalities, less invasive surgery, nanomedicines and targeted therapies are the basis for a new era of intraperitoneal therapy and are beginning to show encouraging outcomes.
Collapse
|
40
|
Eefsen RL, Simonsen KS, Grundtvig P, Klarskov L, Chen IM, Høgdall D, Jensen BV, Lorentzen T, Poulsen TS, Theile S, Nielsen D, Høgdall E. Genomic landscape of treatment refractory metastatic colorectal cancer. Acta Oncol 2021; 60:1621-1628. [PMID: 34606390 DOI: 10.1080/0284186x.2021.1984575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Metastatic colorectal cancer (mCRC) is a complex and heterogeneous disease with few standard and targeted treatment options. Next-generation sequencing of tumor tissue was performed to identify cancer driver mutations to discover possible personalized treatment options, as targeted treatment possibilities are limited for this patient population. Results of genomic sequencing in patients with treatment-refractory mCRC are described in this retrospective analysis. MATERIAL AND METHODS Clinico-pathological characteristics and genomic sequence results of consecutive patients with refractory mCRC, referred to the Experimental Cancer Therapy Unit (ECTU) at Department of Oncology, Herlev & Gentofte Hospital in the period from 1 October 2015 to 14 December 2018 were reviewed in this retrospective analysis. Tumor tissue from the patients was analyzed by next-generation sequencing using the Oncomine Comprehensive primer panel to detect actionable variants of cancer driver mutations and microsatellite instability status. From August 2018 tumor mutational burden was also analyzed. RESULTS A total of 80 patients with treatment-refractory mCRC and in a fairly good performance were referred to the ECTU during this period. Genomic sequencing of tumor tissue was performed for all 80 patients and a cancer driver mutation was identified in 90% (n = 72) of the patients. A total of 31.3% (n = 25) of the patients received therapy either as targetable therapy outside an available trial (n = 2), FDA approved therapy (n = 2), or treatment in phase 1 or 2 trials, independent of the genomic signature 26.3% (n = 21). CONCLUSION Most mCRC patients refractory to standard anti-neoplastic therapies, presented with a cancer driver mutation, however, only a few of these mutations gave rise to matched therapies as only 2.5% of the patients from this period received targeted therapy.
Collapse
Affiliation(s)
- R. L. Eefsen
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - K. S. Simonsen
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - P. Grundtvig
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - L. Klarskov
- Department of Pathology, Herlev Gentofte Hospital, Herlev, Denmark
| | - I. M. Chen
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - D. Høgdall
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - B. V. Jensen
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - T. Lorentzen
- Department of Surgery, Herlev Gentofte Hospital, Herlev, Denmark
| | - T. S. Poulsen
- Department of Pathology, Herlev Gentofte Hospital, Herlev, Denmark
| | - S. Theile
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - D. Nielsen
- Department of Oncology, Herlev Gentofte Hospital, Herlev, Denmark
| | - E. Høgdall
- Department of Pathology, Herlev Gentofte Hospital, Herlev, Denmark
| |
Collapse
|
41
|
Großerueschkamp F, Jütte H, Gerwert K, Tannapfel A. Advances in Digital Pathology: From Artificial Intelligence to Label-Free Imaging. Visc Med 2021; 37:482-490. [PMID: 35087898 DOI: 10.1159/000518494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Digital pathology, in its primary meaning, describes the utilization of computer screens to view scanned histology slides. Digitized tissue sections can be easily shared for a second opinion. In addition, it allows tissue image analysis using specialized software to identify and measure events previously observed by a human observer. These tissue-based readouts were highly reproducible and precise. Digital pathology has developed over the years through new technologies. Currently, the most discussed development is the application of artificial intelligence to automatically analyze tissue images. However, even new label-free imaging technologies are being developed to allow imaging of tissues by means of their molecular composition. SUMMARY This review provides a summary of the current state-of-the-art and future digital pathologies. Developments in the last few years have been presented and discussed. In particular, the review provides an outlook on interesting new technologies (e.g., infrared imaging), which would allow for deeper understanding and analysis of tissue thin sections beyond conventional histopathology. KEY MESSAGES In digital pathology, mathematical methods are used to analyze images and draw conclusions about diseases and their progression. New innovative methods and techniques (e.g., label-free infrared imaging) will bring significant changes in the field in the coming years.
Collapse
Affiliation(s)
- Frederik Großerueschkamp
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Department of Biophysics, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Hendrik Jütte
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Institute of Pathology, Ruhr University Bochum, Bochum, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Department of Biophysics, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Andrea Tannapfel
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Institute of Pathology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
42
|
Xu J, Meng Q, Sun H, Zhang X, Yun J, Li B, Wu S, Li X, Yang H, Zhu H, Aschner M, Relucenti M, Familiari G, Chen R. HER2-specific chimeric antigen receptor-T cells for targeted therapy of metastatic colorectal cancer. Cell Death Dis 2021; 12:1109. [PMID: 34839348 PMCID: PMC8627513 DOI: 10.1038/s41419-021-04100-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/13/2021] [Accepted: 08/16/2021] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR) - T cell therapy is a new class of cellular immunotherapies, which has made great achievements in the treatment of malignant tumors. Despite improvements in colorectal cancer (CRC) therapy, treatment of many patients fails because of metastasis and recurrence. The human epidermal growth factor receptor 2 (HER2) is a substantiated target for CAR-T therapy, and has been reported recently to be over-expressed in CRC, which may provide a potential therapeutic target for CRC treatment. Herein, HER2 was a promising target of metastatic colorectal cancer (mCRC) in CAR-T therapy as assessed by flow cytometry and tissue microarray (TMA) with 9-year survival follow-up data. Furthermore, HER2-specific CAR-T cells exhibited strong cytotoxicity and cytokine-secreting ability against CRC cells in vitro. Moreover, through the tumor-bearing model of the NOD-Prkdcem26cd52Il2rgem26Cd22/Nju (NCG) mice, HER2 CAR-T cells showed signs of effectively preventing CRC progression in three different xenograft models. Notably, HER2 CAR-T cells displayed greater aggressiveness in HER2+ CRC in the patient-derived tumor xenograft (PDX) models and had potent immunotherapeutic capacity for mCRC in the metastatic xenograft mouse models. In conclusion, our studies provide scientific evidence that HER2 CAR-T cells represent an emerging immunotherapy for the treatment of mCRC.
Collapse
Affiliation(s)
- Jie Xu
- School of Public Health, Kunming Medical University, Kunming, 650500, China.,Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Qingtao Meng
- School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Hao Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xinwei Zhang
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, 210003, China
| | - Jun Yun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Bin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Shenshen Wu
- School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.,School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Hongbao Yang
- Center for Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Haitao Zhu
- Colorectal Cancer Center, Department of General Surgery, Jiangsu Cancer Hospital, Cancer Research Institute, Cancer hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Michela Relucenti
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Science, Sapienza University of Rome, Roma, 5000161, Italia
| | - Giuseppe Familiari
- Department of Anatomical, Histological, Medical and Legal locomotive Apparatus, Section of Human Anatomy Via Alfonso Borelli, Sapienza University of Rome, Roma, 5000161, Italia
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China. .,School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China. .,Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
43
|
Guo W, Zhang C, Wang X, Dou D, Chen D, Li J. Resolving the difference between left-sided and right-sided colorectal cancer by single-cell sequencing. JCI Insight 2021; 7:152616. [PMID: 34793335 PMCID: PMC8765049 DOI: 10.1172/jci.insight.152616] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Colorectal cancers (CRCs) exhibit differences in incidence, pathogenesis, molecular pathways, and outcome depending on the location of the tumor. The transcriptomes of 27,927 single human CRC cells from 3 left-sided and 3 right-sided CRC patients were profiled by single-cell RNA-Seq (scRNA-Seq). Right-sided CRC harbors a significant proportion of exhausted CD8+ T cells of a highly migratory nature. One cluster of cells from left-sided CRC exhibiting states preceding exhaustion and a high ratio of preexhausted/exhausted T cells were favorable prognostic markers. Notably, we identified a potentially novel RBP4+NTS+ subpopulation of cancer cells that exclusively expands in left-sided CRC. Tregs from left-sided CRC showed higher levels of immunotherapy-related genes than those from right-sided CRC, indicating that left-sided CRC may have increased responsiveness to immunotherapy. Antibody-dependent cellular phagocytosis (ADCP) and antibody-dependent cellular cytotoxicity (ADCC) induced by M2-like macrophages were more pronounced in left-sided CRC and correlated with a good prognosis in CRC.
Collapse
Affiliation(s)
- Wei Guo
- Department of Colorectal Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Cuiyu Zhang
- Department of Physiology, Shandong University, Jinan, China
| | - Xia Wang
- Department of Physiology, Shandong University, Jinan, China
| | - Dandan Dou
- Department of Physiology, Shandong University, Jinan, China
| | - Dawei Chen
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA) Stem Cells, Université de Liège, Liège, Belgium
| | - Jingxin Li
- Department of Physiology, Shandong University, Jinan, China
| |
Collapse
|
44
|
Toffoli EC, Sheikhi A, Lameris R, King LA, van Vliet A, Walcheck B, Verheul HMW, Spanholtz J, Tuynman J, de Gruijl TD, van der Vliet HJ. Enhancement of NK Cell Antitumor Effector Functions Using a Bispecific Single Domain Antibody Targeting CD16 and the Epidermal Growth Factor Receptor. Cancers (Basel) 2021; 13:cancers13215446. [PMID: 34771609 PMCID: PMC8582566 DOI: 10.3390/cancers13215446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Strategies to enhance the preferential accumulation and activation of Natural Killer (NK) cells in the tumor microenvironment can be expected to increase the efficacy of NK cell-based cancer immunotherapy. In this study, we report that a bispecific single domain antibody (VHH) that targets CD16 (FcRγIII) on NK cells and the epidermal growth factor receptor (EGFR) on tumor cells can be used to target and enhance cytolysis of cancer cells. The bispecific VHH enhanced NK cell activation and cytotoxicity in an EGFR- and CD16-dependent and KRAS-independent manner. Moreover, the bispecific VHH induced stronger activity of cancer patient-derived NK cells and resulted in tumor control in a co-culture of metastatic colorectal cancer cells and either autologous peripheral blood mononuclear cells or allogeneic CD16+ NK cells. We believe that this novel approach could represent a valid therapeutic strategy either alone or in combination with other NK cell-based therapies. Abstract The ability to kill tumor cells while maintaining an acceptable safety profile makes Natural Killer (NK) cells promising assets for cancer therapy. Strategies to enhance the preferential accumulation and activation of NK cells in the tumor microenvironment can be expected to increase the efficacy of NK cell-based therapies. In this study, we show binding of a novel bispecific single domain antibody (VHH) to both CD16 (FcRγIII) on NK cells and the epidermal growth factor receptor (EGFR) on tumor cells of epithelial origin. The bispecific VHH triggered CD16- and EGFR-dependent activation of NK cells and subsequent lysis of tumor cells, regardless of the KRAS mutational status of the tumor. Enhancement of NK cell activation by the bispecific VHH was also observed when NK cells of colorectal cancer (CRC) patients were co-cultured with EGFR expressing tumor cells. Finally, higher levels of cytotoxicity were found against patient-derived metastatic CRC cells in the presence of the bispecific VHH and autologous peripheral blood mononuclear cells or allogeneic CD16 expressing NK cells. The anticancer activity of CD16-EGFR bispecific VHHs reported here merits further exploration to assess its potential therapeutic activity either alone or in combination with adoptive NK cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Elisa C. Toffoli
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Abdolkarim Sheikhi
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
- School of Medicine, Dezful University of Medical Sciences, Department of Immunology, Dezful 64616-43993, Iran
| | - Roeland Lameris
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Lisa A. King
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Amanda van Vliet
- Glycostem Therapeutics, 5349 AB Oss, The Netherlands; (A.v.V.); (J.S.)
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN 55108, USA;
| | - Henk M. W. Verheul
- Radboud Institute for Health Sciences, Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Jan Spanholtz
- Glycostem Therapeutics, 5349 AB Oss, The Netherlands; (A.v.V.); (J.S.)
| | - Jurriaan Tuynman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, 1081 HV Amsterdam, The Netherlands;
| | - Tanja D. de Gruijl
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Hans J. van der Vliet
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
- Lava Therapeutics, 3584 CM Utrecht, The Netherlands
- Correspondence:
| |
Collapse
|
45
|
Lee H, Lee HW, Park EJ, Kang J, Baik SH. Clinicopathologic Characteristics and Survival of Patients With Double Primary Malignancies: Breast and Colorectal Cancer. Ann Coloproctol 2021; 38:197-206. [PMID: 34657410 PMCID: PMC9263303 DOI: 10.3393/ac.2021.00640.0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/26/2021] [Indexed: 11/15/2022] Open
Abstract
Purpose This study aimed to investigate the clinicopathologic features and survival in patients with both breast cancer (BrC) and colorectal cancer (CRC). Methods Between 1996 and 2019, patients who were diagnosed with both BrC and CRC were evaluated retrospectively. Patients with distant metastasis, palliative resection, and previous cancer histories except for BrCs or CRCs were excluded. Altogether, 105 patients were divided into the B=C group (n=21), B-first group (n=40), and C-first group (n=44) according to the definition of synchronous and metachronous cancers. The clinicopathologic features and overall survival were evaluated. Results TNM stages and histologic types were comparable among the 3 groups (P=0.434). The interval of diagnosis was 67.1±40.4 and 59.3±47.2 months in the B- and C-first groups, respectively. The incidence of adjuvant chemotherapy in the B-first group was 57.5%, which was higher than the B=C and C-first groups (P<0.001). The estrogen receptor, progesterone receptor, Ki-67, and HER-2 molecular markers were not significantly different among the groups. The overall survival of the B-first group showed lower survival rates than the C-first group (P=0.039). In the logistic regression, HER-2 status (hazard ratio [HR], 11.9; P=0.032) and lymph node metastasis of CRC (HR, 5.8; P=0.036) were prognostic factors affecting overall survival. Conclusion B-first group had poorer survival outcomes than the C-first group in patients with the metachronous BrC and CRC. HER2 positivity and CRC lymph node metastasis may be prognostic factors that affect overall survival in these patients. The findings support that a colorectal cancer screening program should be included during BrC surveillance.
Collapse
Affiliation(s)
- Hyundo Lee
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Hae Won Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Jung Park
- Division of Colon and Rectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeonghyun Kang
- Division of Colon and Rectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Hyuk Baik
- Division of Colon and Rectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
da Cunha IW, de Almeida Coudry R, de Macedo MP, de Assis EACP, Stefani S, Soares FA. A call to action: molecular pathology in Brazil. SURGICAL AND EXPERIMENTAL PATHOLOGY 2021. [DOI: 10.1186/s42047-021-00096-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Abstract
Background
Adoption of molecular pathology in Brazil is currently very limited. Of note, there are no programs for training new molecular pathologists in the country; thus, documents compiling nationally applicable information on molecular pathology are few.
Methods
A selected panel of Brazilian experts in fields related to molecular pathology were provided with a series of relevant questions to address prior to the multi-day conference. Within this conference, each narrative was discussed and edited by the entire group, through numerous drafts and rounds of discussion until a consensus was achieved.
Results
The panel proposes specific and realistic recommendations for implementing molecular pathology in cancer care in Brazil. In creating these recommendations, the authors strived to address all barriers to the widespread use and impediments to access mentioned previously within this manuscript.
Conclusion
This manuscript provides a review of molecular pathology principles as well as the current state of molecular pathology in Brazil. Additionally, the panel proposes practical and actionable recommendations for the implementation of molecular pathology throughout the country in order to increase awareness of the importance molecular pathology in Brazil.
Collapse
|
47
|
Stefani C, Miricescu D, Stanescu-Spinu II, Nica RI, Greabu M, Totan AR, Jinga M. Growth Factors, PI3K/AKT/mTOR and MAPK Signaling Pathways in Colorectal Cancer Pathogenesis: Where Are We Now? Int J Mol Sci 2021; 22:10260. [PMID: 34638601 PMCID: PMC8508474 DOI: 10.3390/ijms221910260] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a predominant malignancy worldwide, being the fourth most common cause of mortality and morbidity. The CRC incidence in adolescents, young adults, and adult populations is increasing every year. In the pathogenesis of CRC, various factors are involved including diet, sedentary life, smoking, excessive alcohol consumption, obesity, gut microbiota, diabetes, and genetic mutations. The CRC tumor microenvironment (TME) involves the complex cooperation between tumoral cells with stroma, immune, and endothelial cells. Cytokines and several growth factors (GFs) will sustain CRC cell proliferation, survival, motility, and invasion. Epidermal growth factor receptor (EGFR), Insulin-like growth factor -1 receptor (IGF-1R), and Vascular Endothelial Growth Factor -A (VEGF-A) are overexpressed in various human cancers including CRC. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) and all the three major subfamilies of the mitogen-activated protein kinase (MAPK) signaling pathways may be activated by GFs and will further play key roles in CRC development. The main aim of this review is to present the CRC incidence, risk factors, pathogenesis, and the impact of GFs during its development. Moreover, the article describes the relationship between EGF, IGF, VEGF, GFs inhibitors, PI3K/AKT/mTOR-MAPK signaling pathways, and CRC.
Collapse
Affiliation(s)
- Constantin Stefani
- Department of Family Medicine and Clinical Base, ‘‘Dr. Carol Davila’ Central Military Emergency University Hospital, 051075 Bucharest, Romania;
| | - Daniela Miricescu
- Department of Biochemistry, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (I.-I.S.-S.); (A.R.T.)
| | - Iulia-Ioana Stanescu-Spinu
- Department of Biochemistry, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (I.-I.S.-S.); (A.R.T.)
| | - Remus Iulian Nica
- Surgery 2, ‘Dr. Carol Davila’ Central Military Emergency University Hospital, 051075 Bucharest, Romania;
| | - Maria Greabu
- Department of Biochemistry, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (I.-I.S.-S.); (A.R.T.)
| | - Alexandra Ripszky Totan
- Department of Biochemistry, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (I.-I.S.-S.); (A.R.T.)
| | - Mariana Jinga
- Department of Gastroenterology, ‘Dr. Carol Davila’ Central Military Emergency University Hospital, 051075 Bucharest, Romania;
| |
Collapse
|
48
|
Plavec TV, Mitrović A, Perišić Nanut M, Štrukelj B, Kos J, Berlec A. Targeting of fluorescent Lactococcus lactis to colorectal cancer cells through surface display of tumour-antigen binding proteins. Microb Biotechnol 2021; 14:2227-2240. [PMID: 34347360 PMCID: PMC8449671 DOI: 10.1111/1751-7915.13907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
Development of targeted treatment for colorectal cancer is crucial to avoid side effects. To harness the possibilities offered by microbiome engineering, we prepared safe multifunctional cancer cell-targeting bacteria Lactococcus lactis. They displayed, on their surface, binding proteins for cancer-associated transmembrane receptors epithelial cell adhesion molecule (EpCAM) and human epidermal growth factor receptor 2 (HER2) and co-expressed an infrared fluorescent protein for imaging. Binding of engineered L. lactis to tumour antigens EpCAM and HER2 was confirmed and characterised in vitro using soluble receptors. The proof-of-principle of targeting was demonstrated on human cell lines HEK293, HT-29 and Caco-2 with fluorescent microscopy and flow cytometry. The highest L. lactis adhesion was seen for the HEK293 cells with the overexpressed tumour antigens, where colocalisation with their tumour antigens was seen for 39% and 67% of EpCAM-targeting and HER2-targeting bacteria, respectively. On the other hand, no binding was observed to HEK293 cells without tumour antigens, confirming the selectivity of the engineered L. lactis. Apart from cell targeting in static conditions, targeting ability of engineered L. lactis was also shown in conditions of constant flow of bacterial suspension over the HEK293 cells. Successful targeting by engineered L. lactis support the future use of these bacteria in biopharmaceutical delivery for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Tina Vida Plavec
- Department of BiotechnologyJožef Stefan InstituteJamova 39LjubljanaSlovenia
- Faculty of PharmacyUniversity of LjubljanaAškerčeva 7LjubljanaSlovenia
| | - Ana Mitrović
- Department of BiotechnologyJožef Stefan InstituteJamova 39LjubljanaSlovenia
| | | | - Borut Štrukelj
- Department of BiotechnologyJožef Stefan InstituteJamova 39LjubljanaSlovenia
- Faculty of PharmacyUniversity of LjubljanaAškerčeva 7LjubljanaSlovenia
| | - Janko Kos
- Department of BiotechnologyJožef Stefan InstituteJamova 39LjubljanaSlovenia
- Faculty of PharmacyUniversity of LjubljanaAškerčeva 7LjubljanaSlovenia
| | - Aleš Berlec
- Department of BiotechnologyJožef Stefan InstituteJamova 39LjubljanaSlovenia
- Faculty of PharmacyUniversity of LjubljanaAškerčeva 7LjubljanaSlovenia
| |
Collapse
|
49
|
Huang L, Luo S, Zhang X, Cai Y, Xue F, Hu H, Zeng Z, Lin T, Wang F, Wang W, Zhang S, Kang L. Distinct Genomic Landscape of Colorectal Mucinous Carcinoma Determined via Comprehensive Genomic Profiling: Steps to a New Treatment Strategy. Front Oncol 2021; 11:603564. [PMID: 34026601 PMCID: PMC8139246 DOI: 10.3389/fonc.2021.603564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 04/08/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal mucinous carcinoma (MC) is associated with inferior prognosis and response to treatment compared to adenocarcinoma (AC). The molecular landscapes of MC and adenocarcinoma with mucous composition (AMC) are not well-defined. We aimed to describe the genomic landscape of MC and AMC in a large colorectal cancer cohort. Tumor samples from patients with MC, AMC, or AC were analyzed using next-generation sequencing. MC had a molecular signature distinct from that of AC; genomic features were similar between AMC and MC but not between AMC and AC. HER2 amplification and TP53 and APC mutation rates were lower, whereas SMAD4, PIK3CA, ACVR2A, KMT2D, LRP1, TGFBR2, GRIN2A, BRAF V600E, PTEN, and BRCA2 mutation rates were higher in MC than in AC. The mutation frequencies in MAPK, PI3K, and TGF- pathways were higher, whereas those of cell cycle proteins and Wnt were lower in MC and AMC than in AC. The proportion of hypermutated tumors was significantly higher in MC and AMC than in AC. As MC has a distinct molecular signature from AC, immunotherapy can be potentially applied in treating MC. Similar molecular profiles of AMC and MC suggest that treatment strategies for MC, but not AC, can be used for AMC treatment.
Collapse
Affiliation(s)
- Liang Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuanglin Luo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xingwei Zhang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yonghua Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fangqin Xue
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Huanxin Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ziwei Zeng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tengjiao Lin
- Department of Research and Development, OrigiMed, Shanghai, China
| | - Fei Wang
- Department of Research and Development, OrigiMed, Shanghai, China
| | - Weifeng Wang
- Department of Research and Development, OrigiMed, Shanghai, China
| | - Sen Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liang Kang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
50
|
Rottmann D, Assem H, Matsumoto N, Wong S, Hui P, Buza N. Does Specimen Type Have an Impact on HER2 Status in Endometrial Serous Carcinoma? Discordant HER2 Status of Paired Endometrial Biopsy and Hysterectomy Specimens in the Presence of Frequent Intratumoral Heterogeneity. Int J Gynecol Pathol 2021; 40:263-271. [PMID: 32897955 DOI: 10.1097/pgp.0000000000000690] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A recent clinical trial showed prolonged progression-free survival in human epidermal growth factor receptor 2 (HER2)-positive advanced stage and recurrent endometrial serous carcinomas when trastuzumab was added to traditional chemotherapy. Approximately one third of these tumors are HER2-positive and have been described to show unique characteristics of HER2 protein expression and gene amplification, including significant intratumoral heterogeneity, in recent studies. However, currently, there are no standard protocols for the selection of optimal specimen type or algorithm for HER2 testing in endometrial serous carcinomas. The current study aimed to evaluate the concordance of HER2 status between endometrial biopsy/curettage and subsequent hysterectomy specimens in endometrial serous carcinoma. A total of 57 patients with endometrial serous carcinoma with available HER2 status were identified during the study period, 14 of which (14/57, 25%) were HER2-positive by immunohistochemistry and/or fluorescent in situ hybridization (FISH). The final study cohort consisted of 40 paired endometrial biopsies/curettings and hysterectomies to include all 14 HER2-positive tumors and 26 selected HER2-negative tumors to represent an equal distribution of HER2 immunohistochemical scores. HER2 FISH was performed on all tumors with an immunohistochemical score of 2+. HER2 immunohistochemical scores, heterogeneity of HER2 expression, FISH results, and the overall HER2 status were compared between the 2 specimen types. HER2 status was successfully assigned in both specimen types in 37 cases, as three specimens showed inadequate FISH signals. Concordant HER2 status was observed in 84% of cases (31/37), with identical HER2 immunohistochemical scores in 65% (26/40) of tumors. Among the 6 tumors with a discordant HER2 status, 2 were HER2 negative in the biopsy and positive in the hysterectomy, and 4 were HER2-positive in the biopsy and negative in the hysterectomy. The false-negative rate would be 15.4% and 26.7% if only the biopsy or only the hysterectomy would be the basis for the result, respectively. Intratumoral heterogeneity of HER2 protein expression was present in 22 tumors (55%), including all cases with a discordant HER2 status. The concordance rate of HER2 status between paired endometrial biopsies/curettings and hysterectomies of endometrial serous carcinoma is lower than the reported rates of breast cancer, and comparable to those of gastric carcinomas. Frequent heterogeneity of HER2 protein expression combined with the possibility of a spatially more heterogenous sampling of endometrial cavity in biopsies and curettings, and the potential differences in specimen handling/fixation between the 2 specimen types may explain our findings. HER2 testing of multiple specimens may help identify a greater proportion of patients eligible for targeted trastuzumab therapy and should be taken into account in future efforts of developing endometrial cancer-specific HER2 testing algorithm.
Collapse
Affiliation(s)
- Douglas Rottmann
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | | | | | | | | | | |
Collapse
|