1
|
Wang Y, Zhang G, Zhang Z, Zhang M, Chen J, Wang K, Liu L, Bao J, Chen M, Qi X, Gao M. Plasma DNMT1 Activity for Assessing Tumor Burden and Predicting Neoadjuvant Therapy Response in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501064. [PMID: 40317882 DOI: 10.1002/advs.202501064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/03/2025] [Indexed: 05/07/2025]
Abstract
DNA methylation is mediated by DNA methyltransferases (DNMTs), and the stability of their activity is essential for cellular fate. DNMT1 is considered one of the most promising targets for research. However, current detection techniques are limited in accurately quantifying its activity in peripheral blood. Here, a reaction system is developed known as DNMT1 Identification by Variable Activity (DIVA) for the highly sensitive detection of DNMT1 activity in the peripheral blood of breast cancer patients. DIVA can detect DNMT1 at levels as low as 10-7 U mL-1, with minimal time and cost. This method is applied to analyze 271 clinical samples, successfully evaluating tumor burden in patients staged I-IV. Finally, this method is utilized to assess the prognosis of 22 patients undergoing neoadjuvant therapy, demonstrating good consistency with ultrasound imaging results. It is believed that DIVA could serve as an effective auxiliary technique for both the early detection of breast cancer and evaluation of neoadjuvant therapy.
Collapse
Affiliation(s)
- Yingran Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Guozhi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Zhizhao Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Mengsi Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Jiao Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Ke Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Lu Liu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Jing Bao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Mingxuan Gao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| |
Collapse
|
2
|
El Gazzah E, Parker S, Pierobon M. Multi-omic profiling in breast cancer: utility for advancing diagnostics and clinical care. Expert Rev Mol Diagn 2025; 25:165-181. [PMID: 40193192 DOI: 10.1080/14737159.2025.2482639] [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: 07/29/2024] [Accepted: 03/18/2025] [Indexed: 04/09/2025]
Abstract
INTRODUCTION Breast cancer remains a major global health challenge. While advances in precision oncology have contributed to improvements in patient outcomes and provided a deeper understanding of the biological mechanisms that drive the disease, historically, research and patients' allocation to treatment have heavily relied on single-omic approaches, analyzing individual molecular dimensions such as genomics, transcriptomics, or proteomics. While these have provided deep insights into breast cancer biology, they often fail to offer a complete understanding of the disease's complex molecular landscape. AREAS COVERED In this review, the authors explore the recent advancements in multi-omic research in the realm of breast cancer and use clinical data to show how multi-omic integration can offer a more holistic understanding of the molecular alterations and their functional consequences underlying breast cancer. EXPERT OPINION The overall developments in multi-omic research and AI are expected to complement precision diagnostics through potentially refining prognostic models, and treatment selection. Overcoming challenges such as cost, data complexity, and lack of standardization is crucial for unlocking the full potential of multi-omics and AI in breast cancer patient care to enable the advancement of personalized treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Emna El Gazzah
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Scott Parker
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Mariaelena Pierobon
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| |
Collapse
|
3
|
Tamarindo GH, Novais AA, Frigieri BM, Alves DL, de Souza CA, Amadeu A, da Silveira JC, Souza FF, Bordin NA, Chuffa LGA, Zuccari DAPC. Distinct proteomic profiles of plasma-derived extracellular vesicles in healthy, benign, and triple-negative breast cancer: candidate biomarkers for liquid biopsy. Sci Rep 2025; 15:12122. [PMID: 40204835 PMCID: PMC11982211 DOI: 10.1038/s41598-025-95232-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Extracellular vesicles (EVs) derived from plasma, measuring up to 150 nm, act as molecular messengers transmitting critical information to recipient cells, making them valuable candidates for liquid biopsy applications in cancer diagnostics. Triple-negative breast cancer (TNBC) is particularly challenging due to its aggressive nature, metastasis potential, and limited treatment options. This study aimed to identify EV-associated proteins in blood samples that could serve as potential TNBC biomarkers. Using mass spectrometry-based proteomic analysis, we detected unique and differentially expressed proteins across healthy individuals, patients with benign breast conditions, and those with TNBC. While EVs size and concentration showed no differences, the proteomic profile varied significantly among these groups. Several immune-related proteins were found exclusively in healthy individuals but were diminished in both benign and malignant cases. We also assessed the impact of surgery on EVs protein content and identified Histone H2A as a TNBC-specific marker present before surgery. Its expression was further validated through immunohistochemistry and Western blotting in TNBC biopsies and cell lines. Notably, surgical intervention enhanced immune response pathways in TNBC patients. In conclusion, liquid biopsy has the potential to serve as a non-invasive tool for TNBC diagnosis and monitoring, revealing a post-surgery molecular landscape that supports combining immunotherapy with mastectomy.
Collapse
Affiliation(s)
- G H Tamarindo
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto/(FAMERP), São José do Rio Preto, SP, 15090-000, Brazil
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, 13083-100, Brazil
| | - A A Novais
- Institute of Health Science (ICS), Universidade Federal de Mato Grosso (UFMT), Sinop, MT, 78550-728, Brazil
| | - B M Frigieri
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto/(FAMERP), São José do Rio Preto, SP, 15090-000, Brazil
- Institute of Biosciences, Letters and Exact Sciences (IBILCE), UNESP, São José do Rio Preto, SP, Brazil
| | - D L Alves
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto/(FAMERP), São José do Rio Preto, SP, 15090-000, Brazil
- Institute of Biosciences, Letters and Exact Sciences (IBILCE), UNESP, São José do Rio Preto, SP, Brazil
| | - C A de Souza
- Laboratory of Molecular Morphophysiology and Development (LMMD/ZMV), University of São Paulo, Pirassununga, SP, Brazil
| | - A Amadeu
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto/(FAMERP), São José do Rio Preto, SP, 15090-000, Brazil
| | - J C da Silveira
- Laboratory of Molecular Morphophysiology and Development (LMMD/ZMV), University of São Paulo, Pirassununga, SP, Brazil
| | - F F Souza
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, FMVZ, São Paulo State University (UNESP), Botucatu, SP, 18618-681, Brazil
| | - N A Bordin
- Hospital de Base, FAMERP, Faculdade de Medicina de São José do Rio Preto/(FAMERP), São José do Rio Preto, SP, 15090-000, Brazil
| | - L G A Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - São Paulo State University, Botucatu, São Paulo, 18618-689, Brazil
| | - D A P C Zuccari
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto/(FAMERP), São José do Rio Preto, SP, 15090-000, Brazil.
| |
Collapse
|
4
|
e Silva LDS, Jordão PHF, Balieiro BC, Baracioli LDS, de Nóbrega DF, Novais AA, Chuffa LGDA, Zuccari DAPDC. Immunohistochemical Analysis of Inter-Alpha-Trypsin Inhibitor Heavy Chain 2 and Enolase 1 in Canine Mammary Tumors: Associations with Tumor Aggressiveness and Prognostic Significance. Vet Sci 2025; 12:110. [PMID: 40005870 PMCID: PMC11860171 DOI: 10.3390/vetsci12020110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Mammary neoplasms in dogs are a common clinical concern, especially in middle-aged and older intact females. These tumors share similarities with human breast cancer in terms of histology, disease progression, and risk factors, making dogs a relevant model for breast cancer research. The search for biomarkers in canine mammary tumors is essential to understand tumor progression and identify potential therapeutic targets. This study investigated the expression of two potential biomarkers-Inter-Alpha-Trypsin Inhibitor Heavy Chain 2 (ITIH2) and Enolase 1 (ENO1)-in the mammary glands of healthy and tumor-bearing dogs using immunohistochemistry. Both proteins were identified in previous proteomic analyses of extracellular vesicles derived from the plasma of healthy and tumor-bearing dogs. A total of fifty-one canine mammary tissue samples were analyzed and categorized into three groups: (i) the control group, composed of five samples of normal mammary tissue without neoplasia; (ii) benign tumors, composed of nineteen samples of benign mixed tumors; and (iii) malignant tumors, which included six carcinomas in grade 1 mixed tumors, five carcinomas in grade 2 mixed tumors, thirteen solid carcinomas of grade 3, one papillary carcinoma, and two tubular carcinomas. Regarding the intensity of staining, quantified by histoscore, there were no significant differences in the comparison between the groups; for ITIH2, the p-value was 0.33, and for ENO1, the p-value was 0.57. Regarding the predictive potential of their respective ROC curves, the proteins demonstrated low predictive power in canine mammary tumors. These findings indicate that neither ITIH2 nor ENO1 demonstrated strong prognostic value in this setting, as demonstrated by their moderate AUC values, wide confidence intervals, and lack of statistical significance. However, this study found distinct tissue localization patterns for ITIH2 and subcellular localization for ENO1. As an additional way to examine possible associations of these proteins with epithelial-mesenchymal transition, the ZEB1 antibody was tested by both single and double immunohistochemistry, demonstrating a tendency to be more intensely expressed in the malignant group and tending to be associated with ENO1 in canine mammary tumors.
Collapse
Affiliation(s)
- Luadna dos Santos e Silva
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, Brazil; (L.d.S.e.S.); (B.C.B.); (L.d.S.B.)
| | - Pedro Henrique Fogaça Jordão
- Biochemistry and Molecular Biology Research Center (NPBIM), Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, Brazil;
| | - Beatriz Castilho Balieiro
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, Brazil; (L.d.S.e.S.); (B.C.B.); (L.d.S.B.)
| | - Laura de Souza Baracioli
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, Brazil; (L.d.S.e.S.); (B.C.B.); (L.d.S.B.)
| | | | - Adriana Alonso Novais
- Institute of Health Science (ICS), Universidade Federal de Mato Grosso (UFMT), Sinop 78550-728, Brazil;
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP, São Paulo State University, Botucatu 18618-689, Brazil;
| | - Debora Aparecida Pires de Campos Zuccari
- Molecular Investigation of Cancer Laboratory (MICL), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, Brazil; (L.d.S.e.S.); (B.C.B.); (L.d.S.B.)
| |
Collapse
|
5
|
Patel D, Dickson AL, Zickuhr GM, Um IH, Read OJ, Czekster CM, Mullen P, Harrison DJ, Bré J. Defining the mode of action of cisplatin combined with NUC-1031, a phosphoramidate modification of gemcitabine. Transl Oncol 2024; 50:102114. [PMID: 39299019 PMCID: PMC11426158 DOI: 10.1016/j.tranon.2024.102114] [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: 05/31/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
The combination of gemcitabine with platinum agents is a widely used chemotherapy regimen for a number of tumour types. Gemcitabine plus cisplatin remains the current therapeutic choice for biliary tract cancer. Gemcitabine is associated with multiple cellular drug resistance mechanisms and other limitations and has thereforelined in use. NUC-1031 (Acelarin) is a phosphorylated form of gemcitabine, protected by the addition of a phosphoramidate moiety, developed to circumvent the key limitations and generate high levels of the cytotoxic metabolite, dFdCTP. The rationale for combination of gemcitabine and cisplatin is determined by in vitro cytotoxicity. This, however, does not offer an explanation of how these drugs lead to cell death. In this study we investigate the mechanism of action for NUC-1031 combined with cisplatin as a rationale for treatment. NUC-1031 is metabolised to dFdCTP, detectable up to 72 h post-treatment and incorporated into DNA, to stall the cell cycle and cause DNA damage in biliary tract and ovarian cancer cell lines. In combination with cisplatin, DNA damage was increased and occurred earlier compared to monotherapy. The damage associated with NUC-1031 may be potentiated by a second mechanism, via binding the RRM1 subunit of ribonucleotide reductase and perturbing the nucleotide pools; however, this may be mitigated by increased RRM1 expression. The implication of this was investigated in case studies from a Phase I clinical trial to observe whether baseline RRM1 expression in tumour tissue at time of diagnosis correlates with patient survival.
Collapse
Affiliation(s)
- Dillum Patel
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK.
| | - Alison L Dickson
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - Greice M Zickuhr
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - In Hwa Um
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - Oliver J Read
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - Clarissa M Czekster
- School of Biology, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - Peter Mullen
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - David J Harrison
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - Jennifer Bré
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| |
Collapse
|
6
|
Pantano F, Simonetti S, Iuliani M, Guillen MJ, Cuevas C, Aviles P, Cavaliere S, Napolitano A, Cortellini A, Mazzocca A, Nibid L, Sabarese G, Perrone G, Gambarotti M, Righi A, Palmerini E, Stacchiotti S, Barisella M, Gronchi A, Valeri S, Sbaraglia M, Dei Tos AP, Tonini G, Vincenzi B. S-p-bromobenzyl-glutathione cyclopentyl diester (BBGC) as novel therapeutic strategy to enhance trabectedin anti-tumor effect in soft tissue sarcoma preclinical models. Oncogene 2024; 43:2986-2994. [PMID: 39198616 PMCID: PMC11436363 DOI: 10.1038/s41388-024-03143-9] [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: 11/03/2023] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Trabectedin, approved for the treatment of soft tissue sarcoma (STS), interferes with cell division and genetic transcription processes. Due to its strong anti-tumor activity in only certain histotypes, several studies on trabectedin combinations are currently ongoing to improve its efficacy. In this study, we aimed to investigate novel potential therapeutic strategies to enhance the anti-tumor effect of trabectedin using integrated in silico, in vitro, and in vivo approaches. For in silico analysis, we screened two public datasets, GSEA M5190 and TCGA SARC. Fibrosarcoma, leiomyosarcoma, dedifferentiated, and myxoid liposarcoma cell lines were used for in vitro studies. For in vivo experiments, fibrosarcoma orthotopic murine model was developed. In silico analysis identified Glo1 as the only druggable target upregulated after trabectedin treatment and correlated with poor prognosis. The specific Glo1 inhibitor, S-p-bromobenzylglutathione cyclopentyl diester (BBGC), increased trabectedin cytotoxicity in STS cells, and restored drug sensitivity in myxoid liposarcoma cells resistant to trabectedin. Moreover, the combined treatment with BBGC and trabectedin had a synergistic antitumor effect in vivo without any additional toxicity to mice. Based on these results, we believe that BBGC warrants further investigation to evaluate its potential clinical use in combination with trabectedin.
Collapse
Affiliation(s)
- F Pantano
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - S Simonetti
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - M Iuliani
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy.
| | - M J Guillen
- Research Department, PharmaMar S.A, Madrid, Spain
| | - C Cuevas
- Research Department, PharmaMar S.A, Madrid, Spain
| | - P Aviles
- Research Department, PharmaMar S.A, Madrid, Spain
| | - S Cavaliere
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | | | - A Cortellini
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - A Mazzocca
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - L Nibid
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - G Sabarese
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - G Perrone
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - M Gambarotti
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - A Righi
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - E Palmerini
- Osteoncology, Soft Tissue and Bone Sarcomas, Innovative Therapy Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - S Stacchiotti
- Adult mesenchymal tumours and rare cancers unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Barisella
- Tissue Tumor Pathology Unit, Department of Advanced Diagnostics, Fondazione IRCSS Istituto Nazionale dei Tumori Milan, Milano, Italy
| | - A Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Valeri
- Sarcoma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - M Sbaraglia
- Department of Integrated Diagnostics, Azienda Ospedale-Università Padova; Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - A P Dei Tos
- Department of Integrated Diagnostics, Azienda Ospedale-Università Padova; Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - G Tonini
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - B Vincenzi
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| |
Collapse
|
7
|
Takatsuka D, Tachinami H, Suzuki N, Yamazaki M, Yonesi A, Takaichi M, Imaue S, Yamada SI, Tanuma JI, Noguchi M, Tomihara K. PAK4 inhibition augments anti-tumour effect by immunomodulation in oral squamous cell carcinoma. Sci Rep 2024; 14:14092. [PMID: 38890401 PMCID: PMC11189426 DOI: 10.1038/s41598-024-64126-0] [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: 11/02/2023] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumours, warranting novel treatments. Here, we examined the therapeutic efficacy of inhibiting p21 activated kinase 4 (PAK4) in OSCC and determined its immunomodulatory effect by focusing on the enhancement of anti-tumour effects. We examined PAK4 expression in OSCC cells and human clinical samples and analysed the proliferation and apoptosis of OSCC cells following PAK4 inhibition in vitro. We also investigated the effects of in vivo administration of a PAK4 inhibitor on immune cell distribution and T-cell immune responses in OSCC tumour-bearing mice. PAK4 was detected in all OSCC cells and OSCC tissue samples. PAK4 inhibitor reduced the proliferation of OSCC cells and induced apoptosis. PAK4 inhibitor significantly attenuated tumour growth in mouse and was associated with increased proportions of IFN-γ-producing CD8+ T-cells. Furthermore, PAK4 inhibitor increased the number of dendritic cells (DCs) and up-regulated the surface expression of various lymphocyte co-stimulatory molecules, including MHC-class I molecules, CD80, CD83, CD86, and CD40. These DCs augmented CD8+ T-cell activation upon co-culture. Our results suggest that PAK4 inhibition in OSCC can have direct anti-tumour and immunomodulatory effects, which might benefit the treatment of this malignancy.
Collapse
Affiliation(s)
- Danki Takatsuka
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Hidetake Tachinami
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Nihei Suzuki
- Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Manabu Yamazaki
- Divisions of Oral Pathology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514, Japan
| | - Amirmoezz Yonesi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Mayu Takaichi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Shuichi Imaue
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Shin-Ichi Yamada
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Jun-Ichi Tanuma
- Divisions of Oral Pathology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514, Japan
| | - Makoto Noguchi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Kei Tomihara
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan.
- Divisions of Oral and Maxillofacial Surgery, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514, Japan.
| |
Collapse
|
8
|
Saito N, Matsuo T, Tsuda H, Yokota H, Okada H. Novel approach to HER2 quantification using phosphor-integrated dots in human breast invasive cancer microarray. PLoS One 2024; 19:e0303614. [PMID: 38748758 PMCID: PMC11095758 DOI: 10.1371/journal.pone.0303614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/28/2024] [Indexed: 05/19/2024] Open
Abstract
HER2 expression in breast cancer is evaluated to select patients for anti-HER2 therapy. With the advent of newly approved HER2-targeted drugs for low HER2 expression breast cancer, more solid evidence on the whole spectrum of HER2 expression is needed. In this study, we quantitatively assessed HER2 expression from the whole core by combining high-intensity phosphor-integrated dot (PID) immunostaining and whole slide imaging (WSI) analysis. Two types of staining were performed using a 170-core tissue microarray of invasive breast cancer. First, HER2 was stained by immunohistochemistry (IHC), and IHC scores were determined by two practicing pathologists according to the ASCO/CAP HER2 guideline. Second, HER2 was stained with PID, and tentative PID scores were determined by quantitative analysis. The results show that PID can numerically classify HER2 expression status into scores 3+, 2+, 1+, and 0. The HER2 value quantified by PID strongly correlated with the 3, 3'-diaminobenzidine (DAB) IHC score determined by pathologists (R2 = 0.93). PID IHC score 1+ cases included both DAB IHC score 1+ and 0 cases, and low HER2 expression cases appeared to be often evaluated as DAB IHC score 0. Therefore, digital image analysis by PID and WSI can help stratify HER2 IHC. It may also help classify low HER2 expression.
Collapse
Affiliation(s)
- Naoya Saito
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| | - Tsukasa Matsuo
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Hiroyuki Yokota
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| | - Hisatake Okada
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| |
Collapse
|
9
|
Mustakim KR, Eo MY, Seo MH, Yang HC, Kim MK, Myoung H, Kim SM. Ultrastructural and immunohistochemical evaluation of hyperplastic soft tissues surrounding dental implants in fibular jaws. Sci Rep 2024; 14:10717. [PMID: 38730018 PMCID: PMC11087521 DOI: 10.1038/s41598-024-60474-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
In reconstructive surgery, complications post-fibula free flap (FFF) reconstruction, notably peri-implant hyperplasia, are significant yet understudied. This study analyzed peri-implant hyperplastic tissue surrounding FFF, alongside peri-implantitis and foreign body granulation (FBG) tissues from patients treated at the Department of Oral and Maxillofacial Surgery, Seoul National University Dental Hospital. Using light microscopy, pseudoepitheliomatous hyperplasia, anucleate and pyknotic prickle cells, and excessive collagen deposition were observed in FFF hyperplastic tissue. Ultrastructural analyses revealed abnormal structures, including hemidesmosome dilation, bacterial invasion, and endoplasmic reticulum (ER) swelling. In immunohistochemical analysis, unfolded protein-response markers ATF6, PERK, XBP1, inflammatory marker NFκB, necroptosis marker MLKL, apoptosis marker GADD153, autophagy marker LC3, epithelial-mesenchymal transition, and angiogenesis markers were expressed variably in hyperplastic tissue surrounding FFF implants, peri-implantitis, and FBG tissues. NFκB expression was higher in peri-implantitis and FBG tissues compared to hyperplastic tissue surrounding FFF implants. PERK expression exceeded XBP1 significantly in FFF hyperplastic tissue, while expression levels of PERK, XBP1, and ATF6 were not significantly different in peri-implantitis and FBG tissues. These findings provide valuable insights into the interconnected roles of ER stress, necroptosis, apoptosis, and angiogenesis in the pathogenesis of oral pathologies, offering a foundation for innovative strategies in dental implant rehabilitation management and prevention.
Collapse
Affiliation(s)
- Kezia Rachellea Mustakim
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Mi Young Eo
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Mi Hyun Seo
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Hyeong-Cheol Yang
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Min-Keun Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung, Korea
| | - Hoon Myoung
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Soung Min Kim
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Oral and Maxillofacial Microvascular Reconstruction LAB, Brong Ahafo Regional Hospital, P.O.Box 27, Sunyani, Ghana.
| |
Collapse
|
10
|
Ram S, Mojtahedzadeh S, Aguilar JK, Coskran T, Powell EL, O'Neil SP. Quantitative performance assessment of Ultivue multiplex panels in formalin-fixed, paraffin-embedded human and murine tumor specimens. Sci Rep 2024; 14:8496. [PMID: 38605049 PMCID: PMC11009312 DOI: 10.1038/s41598-024-58372-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
We present a rigorous validation strategy to evaluate the performance of Ultivue multiplex immunofluorescence panels. We have quantified the accuracy and precision of four different multiplex panels (three human and one mouse) in tumor specimens with varying levels of T cell density. Our results show that Ultivue panels are typically accurate wherein the relative difference in cell proportion between a multiplex image and a 1-plex image is less than 20% for a given biomarker. Ultivue panels exhibited relatively high intra-run precision (CV ≤ 25%) and relatively low inter-run precision (CV >> 25%) which can be remedied by using local intensity thresholding to gate biomarker positivity. We also evaluated the reproducibility of cell-cell distance estimates measured from multiplex images which show high intra- and inter-run precision. We introduce a new metric, multiplex labeling efficiency, which can be used to benchmark the overall fidelity of the multiplex data across multiple batch runs. Taken together our results provide a comprehensive characterization of Ultivue panels and offer practical guidelines for analyzing multiplex images.
Collapse
Affiliation(s)
- Sripad Ram
- Drug Safety Research and Development, Pfizer Inc., Groton, CT, USA.
| | | | | | - Timothy Coskran
- Drug Safety Research and Development, Pfizer Inc., Groton, CT, USA
| | - Eric L Powell
- Oncology Research and Development, Pfizer Inc., San Diego, CA, USA
| | - Shawn P O'Neil
- Drug Safety Research and Development, Pfizer Inc., Groton, CT, USA
| |
Collapse
|
11
|
Guo J, Tang B, Fu J, Zhu X, Xie W, Wang N, Ding Z, Song Z, Yang Y, Xu G, Xiao X. High-plex spatial transcriptomic profiling reveals distinct immune components and the HLA class I/DNMT3A/CD8 modulatory axis in mismatch repair-deficient endometrial cancer. Cell Oncol (Dordr) 2024; 47:573-585. [PMID: 37847338 PMCID: PMC11090934 DOI: 10.1007/s13402-023-00885-8] [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] [Accepted: 09/23/2023] [Indexed: 10/18/2023] Open
Abstract
PURPOSE Tumors bearing mismatch repair deficiency (MMRd) are characterized by a high load of neoantigens and are believed to trigger immunogenic reactions upon immune checkpoint blockade treatment such as anti-PD-1/PD-L1 therapy. However, the mechanisms are still ill-defined, as multiple cancers with MMRd exhibit variable responses to immune checkpoint inhibitors (ICIs). In endometrial cancer (EC), a distinct tumor microenvironment (TME) exists that may correspond to treatment-related efficacies. We aimed to characterize EC patients with aberrant MMR pathways to identify molecular subtypes predisposed to respond to ICI therapies. METHODS We applied digital spatial profiling, a high-plex spatial transcriptomic approach covering over 1,800 genes, to obtain a highly resolved TME landscape in 45 MMRd-EC patients. We cross-validated multiple biomarkers identified using immunohistochemistry and multiplexed immunofluorescence using in-study and independent cohorts totaling 123 MMRd-EC patients and validated our findings using external TCGA data from microsatellite instability endometrial cancer (MSI-EC) patients. RESULTS High-plex spatial profiling identified a 14-gene signature in the MMRd tumor-enriched regions stratifying tumors into "hot", "intermediate" and "cold" groups according to their distinct immune profiles, a finding highly consistent with the corresponding CD8 + T-cell infiltration status. Our validation studies further corroborated an existing coregulatory network involving HLA class I and DNMT3A potentially bridged through dynamic crosstalk incorporating CCL5. CONCLUSION Our study confirmed the heterogeneous TME status within MMRd-ECs and showed that these ECs can be stratified based on potential biomarkers such as HLA class I, DNMT3A and CD8 in pathological settings for improved ICI therapeutic efficacy in this subset of patients.
Collapse
Affiliation(s)
- Jingjing Guo
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- School of Medical and Life Sciences, Chengdu University of TCM, Chengdu, China
| | - Baijie Tang
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Fu
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuan Zhu
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- School of Medical and Life Sciences, Chengdu University of TCM, Chengdu, China
| | - Wenlong Xie
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- School of Medical and Life Sciences, Chengdu University of TCM, Chengdu, China
| | - Nan Wang
- Mills Institute for Personalized Cancer Care, Jinan, China
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Jinan, China
| | - Zhentao Song
- Mills Institute for Personalized Cancer Care, Jinan, China
| | - Yue Yang
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Gang Xu
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xue Xiao
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
12
|
Mon AM, Intuyod K, Klungsaeng S, Jusakul A, Pongking T, Lert-Itthiporn W, Luvira V, Pairojkul C, Plengsuriyakarn T, Na-Bangchang K, Pinlaor S, Pinlaor P. Overexpression of microRNA-205-5p promotes cholangiocarcinoma growth by reducing expression of homeodomain-interacting protein kinase 3. Sci Rep 2023; 13:22444. [PMID: 38105269 PMCID: PMC10725890 DOI: 10.1038/s41598-023-49694-x] [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: 02/20/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
The microRNA miR-205-5p has diverse effects in different malignancies, including cholangiocarcinoma (CCA), but its effects on CCA progression is unclear. Here we investigated the role and function of miR-205-5p in CCA. Three CCA cell lines and human serum samples were found to have much higher expression levels of miR-205-5p than seen in typical cholangiocyte cell lines and healthy controls. Inhibition of miR-205-5p suppressed CCA cell motility, invasion and proliferation of KKU-213B whereby overexpression of miR-205-5p promoted cell proliferation and motility of KKU-100 cells. Bioinformatics tools (miRDB, TargetScan, miRWalk, and GEPIA) all predicted various miR-205-5p targets. Experiments using miR-205-5p inhibitor and mimic indicated that homeodomain-interacting protein kinase 3 (HIPK3) was a potential direct target of miR-205-5p. Overexpression of HIPK3 using HIPK3 plasmid cloning DNA suppressed migration and proliferation of KKU-100 cells. Notably, HIPK3 expression was lower in human CCA tissues than in normal adjacent tissues. High HIPK3 expression was significantly associated with longer survival time of CCA patients. Multivariate regression analysis indicated tissue HIPK3 levels as an independent prognostic factor for CCA patients. These findings indicate that overexpression of miR-205-5p promotes CCA cells proliferation and migration partly via HIPK3-dependent way. Therefore, targeting miR-205-5p may be a potential treatment approach for CCA.
Collapse
Affiliation(s)
- Aye Myat Mon
- Medical Technology Program, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kitti Intuyod
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sirinapha Klungsaeng
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Apinya Jusakul
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Thatsanapong Pongking
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Worachart Lert-Itthiporn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Vor Luvira
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Tullayakorn Plengsuriyakarn
- Graduate Program in Bioclinical Sciences, Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Pathum Thani, 12120, Thailand
| | - Kesara Na-Bangchang
- Graduate Program in Bioclinical Sciences, Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Pathum Thani, 12120, Thailand
| | - Somchai Pinlaor
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Porntip Pinlaor
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
13
|
Hu Y, Jones D, Zhao W, Tozbikian G, Wesolowski R, Parwani AV, Li Z. Incidence, Clinicopathologic Features, HER2 Fluorescence In Situ Hybridization Profile, and Oncotype DX Results of Human Epidermal Growth Factor Receptor 2-Low Breast Cancers: Experience From a Single Academic Center. Mod Pathol 2023; 36:100164. [PMID: 36967073 DOI: 10.1016/j.modpat.2023.100164] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/08/2023] [Accepted: 03/20/2023] [Indexed: 04/20/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2)-low breast cancer, defined by an immunohistochemical (IHC) score of 1+ or 2+ with negative in situ hybridization, is emerging as a predictive marker for the use of the antibody-drug conjugate. To understand how this category differs from HER2-zero cases, we investigated clinicopathological characteristics and HER2 fluorescence in situ hybridization results in a large cohort of 1309 continuous HER2-negative invasive breast carcinomas from 2018 to 2021 evaluated by the Food and Drug Administration-approved HER2 IHC test. Additionally, we compared Oncotype DX recurrence scores and HER2 mRNA expression between HER-low and HER2-zero cases in a separate cohort of 438 estrogen receptor-positive (ER+) early-stage breast carcinoma cases from 2014 to 2016. Based on the cohort from 2018 to 2021, the incidence of HER2-low breast cancers was approximately 54%. HER2-low cases had less frequent grade 3 morphology, less frequent triple-negative results, ER and progesterone receptor negativity, and a higher mean HER2 copy number and HER2/CEP17 ratio than HER2-zero cases (P < .0001). Among ER+ cases, HER2-low cases showed significantly less frequent Nottingham grade 3 tumors. In the cohort from 2014 to 2016, HER2-low cases showed significantly higher ER+ percentages, fewer progesterone receptor-negative cases, lower Oncotype DX recurrence scores, and higher HER2 mRNA expression scores than HER2-zero cases. In summary, this is the first study, to our knowledge, using a large cohort of continuous cases evaluated by the Food and Drug Administration-approved HER2 IHC companion diagnostic test for HER2-low expression and HER2 fluorescence in situ hybridization profile in a real-world setting. Although HER2-low cases showed a higher HER2 copy number, ratio, and mRNA level than HER2-zero cases statistically, such small differences are unlikely to be biologically or clinically meaningful. However, our study suggests that HER2-low/ER+ early-stage breast carcinoma may represent a less aggressive group of breast carcinoma, given its association with a lower Nottingham grade and Oncotype DX recurrence score.
Collapse
Affiliation(s)
- Yan Hu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Dan Jones
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Weiqiang Zhao
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Gary Tozbikian
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Robert Wesolowski
- Department of Internal Medicine, Medical Oncology Division, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
14
|
Márquez BT, Leung TCS, Hui J, Charron F, McKinney RA, Watt AJ. A mitochondrial-targeted antioxidant (MitoQ) improves motor coordination and reduces Purkinje cell death in a mouse model of ARSACS. Neurobiol Dis 2023; 183:106157. [PMID: 37209925 DOI: 10.1016/j.nbd.2023.106157] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023] Open
Abstract
Mitochondrial deficits have been observed in animal models of Autosomal-recessive spastic ataxia of the Charlevoix-Saguenay (ARSACS) and in patient-derived fibroblasts. We investigated whether mitochondrial function could be restored in Sacs-/- mice, a mouse model of ARSACS, using the mitochondrial-targeted antioxidant ubiquinone MitoQ. After 10 weeks of chronic MitoQ administration in drinking water, we partially reversed motor coordination deficits in Sacs-/- mice but did not affect litter-matched wild-type control mice. MitoQ administration led to a restoration of superoxide dismutase 2 (SOD2) in cerebellar Purkinje cell somata without altering Purkinje cell firing deficits. Purkinje cells in anterior vermis of Sacs-/- mice normally undergo cell death in ARSACS; however, Purkinje cells numbers were elevated after chronic MitoQ treatment. Furthermore, Purkinje cell innervation of target neurons in the cerebellar nuclei of Sacs-/- mice were also partially restored with MitoQ treatment. Our data suggest that MitoQ is a potential therapeutic treatment for ARSACS and that it improves motor coordination via increasing cerebellar Purkinje cell mitochondria function and reducing Purkinje cell death.
Collapse
Affiliation(s)
| | | | - Jeanette Hui
- Department of Biology, McGill University, Montreal, QC, Canada
| | - François Charron
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - R Anne McKinney
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
| | - Alanna J Watt
- Department of Biology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
15
|
Elomaa H, Ahtiainen M, Väyrynen SA, Ogino S, Nowak JA, Lau MC, Helminen O, Wirta EV, Seppälä TT, Böhm J, Mecklin JP, Kuopio T, Väyrynen JP. Spatially resolved multimarker evaluation of CD274 (PD-L1)/PDCD1 (PD-1) immune checkpoint expression and macrophage polarisation in colorectal cancer. Br J Cancer 2023; 128:2104-2115. [DOI: 10.1038/s41416-023-02238-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Abstract
Background
The CD274 (PD-L1)/PDCD1 (PD-1) immune checkpoint interaction may promote cancer progression, but the expression patterns and prognostic significance of PD-L1 and PD-1 in the colorectal cancer microenvironment are inadequately characterised.
Methods
We used a custom 9-plex immunohistochemistry assay to quantify the expression patterns of PD-L1 and PD-1 in macrophages, T cells, and tumour cells in 910 colorectal cancer patients. We evaluated cancer-specific mortality according to immune cell subset densities using multivariable Cox regression models.
Results
Compared to PD-L1– macrophages, PD-L1+ macrophages were more likely M1-polarised than M2-polarised and located closer to tumour cells. PD-L1+ macrophage density in the invasive margin associated with longer cancer-specific survival [Ptrend = 0.0004, HR for the highest vs. lowest quartile, 0.52; 95% CI: 0.34–0.78]. T cell densities associated with longer cancer-specific survival regardless of PD-1 expression (Ptrend < 0.005 for both PD-1+ and PD-1– subsets). Higher densities of PD-1+ T cell/PD-L1+ macrophage clusters associated with longer cancer-specific survival (Ptrend < 0.005).
Conclusions
PD-L1+ macrophages show distinct polarisation profiles (more M1-like), spatial features (greater co-localisation with tumour cells and PD-1+ T cells), and associations with favourable clinical outcome. Our comprehensive multimarker assessment could enhance the understanding of immune checkpoints in the tumour microenvironment and promote the development of improved immunotherapies.
Collapse
|
16
|
Nicolò E, Boscolo Bielo L, Curigliano G, Tarantino P. The HER2-low revolution in breast oncology: steps forward and emerging challenges. Ther Adv Med Oncol 2023; 15:17588359231152842. [PMID: 36844387 PMCID: PMC9943960 DOI: 10.1177/17588359231152842] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/05/2023] [Indexed: 02/11/2023] Open
Abstract
Approximately half of breast cancers (BCs), historically categorized as human epidermal growth factor receptor 2 (HER2)-negative, have low expression of HER2 defined as an immunohistochemical (IHC) score of 1+ or 2+ with negative in situ hybridization. Retrospective evidence suggest that HER2-low BC does not represent a distinct subtype from a biological and prognostic perspective. Nonetheless, it currently constitutes an essential biomarker to guide treatment selection and its introduction has led to reconsidering the binary classification of HER2 status according to which only patients with HER2-positive BC were thought to derive benefit from anti-HER2 therapies. Trastuzumab deruxtecan has recently been approved by the U.S. Food and Drug Administration for the treatment of patients with HER2-low metastatic BC based on the results of the DESTINY-Breast04 phase III trial, and other antibody-drug conjugates (ADCs) targeting HER2 are showing promising results. Treatment paradigms for both triple-negative and hormone receptor-positive BCs exhibiting HER2-low expression are thus rapidly evolving. Given its therapeutic implications, it is essential to accurately recognize the level of HER2 expression, and the development of more sensitive and reliable methods for HER2 testing and scoring is warranted, especially since the minimum threshold of HER2 expression required for T-DXd efficacy is currently under investigation. Given the signs of activity of T-DXd even in patients with HER2-0 (IHC 0) disease, an evolution in the way we define HER2-low is anticipated. Considering the expansion of the therapeutic armamentarium for BC patients, with several ADCs approaching the clinic, research efforts are needed to clarify whether the expression level of targets can enrich for responders to a given ADC as well as to understand mechanisms of resistance with the goal of optimizing the sequencing of ADCs.
Collapse
Affiliation(s)
- Eleonora Nicolò
- Division of New Drugs and Early Drug
Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology,
University of Milan, Milan, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug
Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology,
University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug
Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology,
University of Milan, Milan, Italy
| | - Paolo Tarantino
- Division of New Drugs and Early Drug
Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology,
University of Milan, Milan, Italy
- Breast Oncology Center, Dana-Farber Cancer
Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, Boston, USA
| |
Collapse
|
17
|
Niwa K, Kubota H, Enomoto T, Ichino Y, Ohmiya Y. Quantitative Analysis of Bioluminescence Optical Signal. BIOSENSORS 2023; 13:223. [PMID: 36831989 PMCID: PMC9953788 DOI: 10.3390/bios13020223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
Bioluminescence is light emission based on the luciferin-luciferase enzymatic reaction in living organisms. Optical signals from bioluminescence (BL) reactions are available for bioanalysis and bioreporters for gene expression, in vitro, in vivo, and ex vivo bioimaging, immunoassay, and other applications. Although there are numerous bioanalysis methods based on BL signal measurements, the BL signal is measured as a relative value, and not as an absolute value. Recently, some approaches have been established to completely quantify the BL signal, resulting in, for instance, the redetermination of the quantum yield of the BL reaction and counting the photon number of the BL signal at the single-cell level. Reliable and reproducible understanding of biological events in the bioanalysis and bioreporter fields can be achieved by means of standardized absolute optical signal measurements, which is described in an International Organization for Standardization (ISO) document.
Collapse
Affiliation(s)
- Kazuki Niwa
- National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8563, Japan
| | | | | | - Yoshiro Ichino
- National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8563, Japan
| | - Yoshihiro Ohmiya
- National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8563, Japan
- Osaka Institute of Technology (OIT), Osaka 535-8585, Japan
| |
Collapse
|
18
|
Witt BL, Zhou W, Ambaye AB, Bellizzi A, Booth CN, Sundling K, Nguyen L, Russell DK, Schinstine M, Staats PN, Thomsen J, Troxell M, Souers RJ, Dvorak J, Lin X, Kurtycz DFI. Using American Type Culture Collection Cell Lines to Evaluate Interlaboratory Variables for Estrogen Receptor and Human Epidermal Growth Factor Receptor 2 Immunostaining. Arch Pathol Lab Med 2023; 147:143-148. [PMID: 35639575 DOI: 10.5858/arpa.2021-0152-cp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 02/05/2023]
Abstract
CONTEXT.— Most laboratories currently use patient tissues for validating immunohistochemical stains. OBJECTIVE.— To explore advantages of using cell lines with known antigenicity as a validation method. DESIGN.— Five American Type Culture Collection (ATCC) cell lines with known negative, low positive, and moderate to strong estrogen receptor (ER) expression as well as negative, equivocal, and positive human epidermal growth factor receptor 2 (HER2) expression were cultured and made into cell blocks. One block from each cell line was fixed in formalin and another in ethanol before cell block preparation. Two sets of paired unstained slides from each block were sent to 10 different laboratories for HER2 and ER staining to be stained on runs from different days according to each laboratory's defined protocol. RESULTS.— The 10 study participants evaluated 40 slides in a blinded fashion. For ER expression, all 80 interpretations for the ER strong and moderate positive cell lines had the target ER-positive result, and 74 of 80 ER-negative cell lines (92.5%) had agreement with the intended negative result. The ER low positive cell line showed varied but positive expression among all observers. The HER2 (3+)-positive cell lines yielded a target interpretation of 3+ in 65 of 80 interpretations (81.2%). For the HER2-negative cell line 69 of 78 interpretations (88.5%) were consistent with the target response (0 or 1+). No significant variation was observed between the ethanol- and non-ethanol-exposed cell lines, or between runs by the same laboratory. Variation from target results clustered within laboratories. CONCLUSIONS.— This study indicates that variability between laboratories can be identified by using cell lines for quantitative or semiquantitative immunohistochemistry when using cultured cell lines of known antigenicity. These cell lines could potentially play a role in aiding anatomic pathology laboratories in validating immunohistochemistry tests for formalin- and ethanol-fixed tissues.
Collapse
Affiliation(s)
- Benjamin L Witt
- From the Department of Pathology, University of Utah, Salt Lake City (Witt).,From the Institute for Clinical and Experimental Pathology, ARUP, Salt Lake City, Utah (Witt, Zhou)
| | - Wenhua Zhou
- From the Institute for Clinical and Experimental Pathology, ARUP, Salt Lake City, Utah (Witt, Zhou)
| | - Abiy B Ambaye
- From the Department of Pathology, University of Vermont Medical Center, Burlington (Ambaye, Schinstine)
| | - Andrew Bellizzi
- From the Department of Pathology, University of Iowa, Iowa City (Bellizzi)
| | - Christine N Booth
- From the Department of Pathology, Cleveland Clinic, Cleveland, Ohio (Booth)
| | - Kaitlin Sundling
- From the Department of Pathology, University of Wisconsin, Madison (Sundling, Kurtycz)
| | - Lananh Nguyen
- From the Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire (Nguyen)
| | - Donna K Russell
- From the Department of Pathology, University of Rochester, Rochester, New York (Russell)
| | - Malcolm Schinstine
- From the Department of Pathology, University of Vermont Medical Center, Burlington (Ambaye, Schinstine)
| | - Paul N Staats
- From the Department of Pathology, University of Maryland, Baltimore (Staats)
| | - Jean Thomsen
- From the Department of Pathology, Methodist Jennie Edmundson Hospital, Council Bluffs, Iowa (Thomsen)
| | - Megan Troxell
- From the Department of Pathology, Stanford University, Palo Alto, California (Troxell)
| | - Rhona J Souers
- From the Department of Biostatistics (Souers), College of American Pathologists, Northfield, Illinois
| | - James Dvorak
- From the Department of Proficiency Testing (Dvorak), College of American Pathologists, Northfield, Illinois
| | - Xiaoqi Lin
- From the Department of Pathology, Northwestern Medicine, Chicago, Illinois (Lin)
| | - Daniel F I Kurtycz
- From the Department of Pathology, University of Wisconsin, Madison (Sundling, Kurtycz)
| |
Collapse
|
19
|
Photofuel cell-based self-powered biosensor for HER2 detection by integration of plasmonic-metal/conjugated molecule hybrids and electrochemical sandwich structure. Biosens Bioelectron 2023; 220:114850. [DOI: 10.1016/j.bios.2022.114850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/18/2022] [Accepted: 10/21/2022] [Indexed: 11/23/2022]
|
20
|
Dabbs DJ, Huang RS, Ross JS. Novel markers in breast pathology. Histopathology 2023; 82:119-139. [PMID: 36468266 DOI: 10.1111/his.14770] [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: 06/27/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022]
Abstract
Breast pathology is an ever-expanding database of information which includes markers, or biomarkers, that detect or help treat the disease as prognostic or predictive information. This review focuses on these aspects of biomarkers which are grounded in immunohistochemistry, liquid biopsies and next-generation sequencing.
Collapse
Affiliation(s)
- David J Dabbs
- PreludeDx, Laguna Hills, CA, USA.,Department of Pathology, University of Pittsburgh, Board Member, CASI (Consortium for Analytical Standardization in Immunohistochemistry), Pittsburgh, PA, USA
| | - Richard S Huang
- Clinical Development, Foundation Medicine, Cambridge, MA, USA
| | | |
Collapse
|
21
|
Wu Y, Zhong R, Ma F. HER2-low breast cancer: Novel detections and treatment advances. Crit Rev Oncol Hematol 2023; 181:103883. [PMID: 36427769 DOI: 10.1016/j.critrevonc.2022.103883] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/20/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Breast cancer (BC), which has the highest cancer incidence in women, seriously threatens women's health. Since human epidermal growth factor receptor-2 (HER2) characterization, breast cancer treatment has entered an era of individualized targeted therapy. With the emergence of anti-HER2 targeting agents, monoclonal antibodies (mAbs) and tyrosine kinase inhibitors have considerably improved the prognosis of HER2-positive BC. However, HER2-low BC, accounting for 45-55% of BC patients, is less likely to benefit from conventional HER2-targeting mAbs. The growing success of the new generation of drugs, especially promising HER2-directed antibody-drug conjugates, has changed the treatment landscape for patients with HER2-low BC, leading to a research boom. HER-2-low BC is a heterogeneous entity, and there many areas remain to be explored. In this article, we review the literature on HER2-low BC, mainly focusing on its detection assays, clinicopathological profiles and treatment landscape, and hopefully provide insight into future perspectives.
Collapse
Affiliation(s)
- Yun Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruiqi Zhong
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
22
|
Cordova C, Muñoz R, Olivares R, Minonzio JG, Lozano C, Gonzalez P, Marchant I, González-Arriagada W, Olivero P. HER2 classification in breast cancer cells: A new explainable machine learning application for immunohistochemistry. Oncol Lett 2022; 25:44. [PMID: 36644146 PMCID: PMC9811637 DOI: 10.3892/ol.2022.13630] [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: 09/15/2022] [Accepted: 10/31/2022] [Indexed: 12/15/2022] Open
Abstract
The immunohistochemical (IHC) evaluation of epidermal growth factor 2 (HER2) for the diagnosis of breast cancer is still qualitative with a high degree of inter-observer variability, and thus requires the incorporation of complementary techniques such as fluorescent in situ hybridization (FISH) to resolve the diagnosis. Implementing automatic algorithms to classify IHC biomarkers is crucial for typifying the tumor and deciding on therapy for each patient with better performance. The present study aims to demonstrate that, using an explainable Machine Learning (ML) model for the classification of HER2 photomicrographs, it is possible to determine criteria to improve the value of IHC analysis. We trained a logistic regression-based supervised ML model with 393 IHC microscopy images from 131 patients, to discriminate between upregulated and normal expression of the HER2 protein. Pathologists' diagnoses (IHC only) vs. the final diagnosis complemented with FISH (IHC + FISH) were used as training outputs. Basic performance metrics and receiver operating characteristic curve analysis were used together with an explainability algorithm based on Shapley Additive exPlanations (SHAP) values to understand training differences. The model could discriminate amplified IHC from normal expression with better performance when the training output was the IHC + FISH final diagnosis (IHC vs. IHC + FISH: area under the curve, 0.94 vs. 0.81). This may be explained by the increased analytical impact of the membrane distribution criteria over the global intensity of the signal, according to SHAP value interpretation. The classification model improved its performance when the training input was the final diagnosis, downplaying the weighting of the intensity of the IHC signal, suggesting that to improve pathological diagnosis before FISH consultation, it is necessary to emphasize subcellular patterns of staining.
Collapse
Affiliation(s)
- Claudio Cordova
- Cell Function and Structure Laboratory (EFC Lab.), Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile,PhD Program in Health Sciences and Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile
| | - Roberto Muñoz
- PhD Program in Health Sciences and Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile,School of Informatics Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile
| | - Rodrigo Olivares
- School of Informatics Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile,Center for Research and Development in Health Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile
| | - Jean-Gabriel Minonzio
- PhD Program in Health Sciences and Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile,School of Informatics Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile,Center for Research and Development in Health Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile,Millennium Institute for Intelligent Healthcare: iHEALTH, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile
| | - Carlo Lozano
- Pathological Anatomy Service, Carlos Van Buren Hospital, Valparaíso 2340105, Chile
| | - Paulina Gonzalez
- Pathological Anatomy Service, Carlos Van Buren Hospital, Valparaíso 2340105, Chile,School of Medical Technology, Andrés Bello National University (UNAB), Viña del Mar, 2520000, Chile
| | - Ivanny Marchant
- Medical Modeling Laboratory, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2362735, Chile
| | - Wilfredo González-Arriagada
- Faculty of Dentistry, Universidad de los Andes, Santiago 7620086, Chile,Biomedical Research and Innovation Center (CIIB), Universidad de los Andes, Santiago 7620086, Chile
| | - Pablo Olivero
- Cell Function and Structure Laboratory (EFC Lab.), Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile,PhD Program in Health Sciences and Engineering, Faculty of Engineering, Universidad de Valparaíso, Valparaíso 2362735, Chile,Correspondence to: Dr Pablo Olivero, Cell Function and Structure Laboratory (EFC Lab.), Faculty of Engineering, Universidad de Valparaíso, 2664 Hontaneda, Valparaíso 2341386, Chile, E-mail:
| |
Collapse
|
23
|
Marui S, Nishikawa Y, Shiokawa M, Yokode M, Matsumoto S, Muramoto Y, Ota S, Nakamura T, Yoshida H, Okada H, Kuwada T, Matsumori T, Kuriyama K, Fukuda A, Saur D, Aoi T, Uza N, Kodama Y, Chiba T, Seno H. Context-Dependent Roles of Hes1 in the Adult Pancreas and Pancreatic Tumor Formation. Gastroenterology 2022; 163:1613-1629.e12. [PMID: 36075324 DOI: 10.1053/j.gastro.2022.08.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The Notch signaling pathway is an important pathway in the adult pancreas and in pancreatic ductal adenocarcinoma (PDAC), with hairy and enhancer of split-1 (HES1) as the core molecule in this pathway. However, the roles of HES1 in the adult pancreas and PDAC formation remain controversial. METHODS We used genetically engineered dual-recombinase mouse models for inducing Hes1 deletion under various conditions. RESULTS The loss of Hes1 expression in the adult pancreas did not induce phenotypic alterations. However, regeneration was impaired after caerulein-induced acute pancreatitis. In a pancreatic intraepithelial neoplasia (PanIN) mouse model, PanINs rarely formed when Hes1 deletion preceded PanIN formation, whereas more PanINs were formed when Hes1 deletion succeeded PanIN formation. In a PDAC mouse model, PDAC formation was also enhanced by Hes1 deletion after PanIN/PDAC development; therefore, Hes1 promotes PanIN initiation but inhibits PanIN/PDAC progression. RNA sequencing and chromatin immunoprecipitation-quantitative polymerase chain reaction revealed that Hes1 deletion enhanced epithelial-to-mesenchymal transition via Muc5ac up-regulation in PDAC progression. The results indicated that HES1 is not required for maintaining the adult pancreas under normal conditions, but is important for regeneration during recovery from pancreatitis; moreover, Hes1 plays different roles, depending on the tumor condition. CONCLUSIONS Our findings highlight the context-dependent roles of HES1 in the adult pancreas and pancreatic cancer.
Collapse
Affiliation(s)
- Saiko Marui
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshihiro Nishikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Hyogo, Japan; Department of Gastroenterology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Masataka Yokode
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shimpei Matsumoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuya Muramoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sakiko Ota
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeharu Nakamura
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Yoshida
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirokazu Okada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Kuwada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoaki Matsumori
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Katsutoshi Kuriyama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Dieter Saur
- Department of Internal Medicine II, Klinikum rechts der Isar Technische Universität München, München, Bayern, Germany; Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Baden-Württemberg, Germany
| | - Takashi Aoi
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Hyogo, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuzo Kodama
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kansai Electric Power Hospital, Osaka, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
24
|
Wang H, Ma Y. β-Elemene alleviates cisplatin resistance in oral squamous cell carcinoma cell via inhibiting JAK2/STAT3 pathway in vitro and in vivo. Cancer Cell Int 2022; 22:244. [PMID: 35909161 PMCID: PMC9341059 DOI: 10.1186/s12935-022-02650-7] [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: 03/21/2022] [Accepted: 06/25/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Objective
To investigate the effect of β-Elemene (β-Ele) on the cisplatin sensitivity of OSCC cells and its mechanism in vitro and in vivo.
Methods
The human OSCC cell lines Tca-8113 and the cisplatin-resistant cell line Tca-8113-CDDP were cultured with β-Ele or/and cisplatin. The cytotoxicity of cisplatin or β-Ele, cell viability, cell cycles and apoptosis were detected. And the expression of JAK2/STAT3 related protein were detected. The xenograft tumor model of OSCC was established in nude mice and treated with cisplatin and/or β-Ele. The volume and weight of the transplanted tumor was measured, and the expression of p-JAK2 and p-STAT3 and cell apoptosis in the xenograft tumor tissues were detected.
Results
The combination of β-Ele and cisplatin significantly suppressed the cell proliferation, induced cell cycle arrest, promoted the apoptosis of Tca-8113-CDDP cells, and suppressed the activation of JAK2/STAT3 signaling pathway. The rescue experiments suggested that β-Ele enhanced cisplatin sensitivity via down-regulating JAK2/STAT3 signaling pathway. In vivo, β-Ele and cisplatin synergistically suppressed the tumor growth and induced apoptosis, and down-regulated the expression of p-JAK2 and p-STAT3.
Conclusions
β-Ele inhibits the cell viability and enhances the cisplatin sensitivity of OSCC by blocking the activation of JAK/STAT3 signaling pathway in vitro and in vivo, and the combination of β-Ele and cisplatin maybe a novel treatment for OSCC.
Collapse
|
25
|
Perossi IFS, Saito MM, Varallo GR, de Godoy BLV, Colombo J, Zuccari DAPC. Protein Expression of PI3K/AKT/mTOR Pathway Targets Validated by Gene Expression and its Correlation with Prognosis in Canine Mammary Cancer. J Mammary Gland Biol Neoplasia 2022; 27:241-252. [PMID: 36323932 DOI: 10.1007/s10911-022-09527-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 12/31/2022] Open
Abstract
Mammary cancer is the main type of neoplasia in female dogs and is considered an adequate model for the biological and therapeutic study of cancer in women. The PIK3CA/AKT/mTOR pathway plays a central role in cellular homeostasis and is often dysregulated in cancer. The increased expression of PI3K protein in the literature is associated with a poor prognosis, and alterations in the PIK3CA gene can lead to changes in downstream pathways. Thus, the objective of this study was to validate the protein expression to confirm the gene expression of proteins belonging to the main pathway PI3K and PTEN, and their downstream pathways through ZEB1, ZEB2, HIF1A, VHL, CASP3 and PARP1 relating to prognosis in canine mammary cancer. For protein studies, the samples came from 58 female dogs with mammary neoplasia, immunohistochemistry was performed and its analysis by the histoscore method. For the genetic evaluation, the samples came from 13 patients, the DNA was extracted and the analysis for quantitative expression. Through immunohistochemistry, PI3K positivity was significantly associated with affected regional lymph node, distant metastasis, patients with HER2+, Triple Negative and Luminal B phenotypes, and the lowest survival rates. Through gene expression, we observed higher gene expression of ZEB2 and PARP1 both among patients who were alive and who died, which was not true for the expressions of PIK3CA and HIF1A. In conclusion, the data observed in this work are promising in the study of new molecular prognostic markers such as PI3K, ZEB2 and PARP1 for canine mammary cancer.
Collapse
Affiliation(s)
- Isabela F S Perossi
- Instituto de Biociências, Letras e Ciências Exatas (IBILCE) UNESP, São José do Rio Preto, Brazil.
| | - Mylena M Saito
- Centro Universitário de Rio Preto (UNIRP), São José do Rio Preto, Brazil
| | | | | | - Jucimara Colombo
- Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Debora A P C Zuccari
- Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| |
Collapse
|
26
|
Miles J, Ward SG, Larijani B. The Fusion of Quantitative Molecular Proteomics and Immune-Oncology: A Step Towards Precision Medicine in Cancer Therapeutics. FEBS Lett 2022; 596:2721-2735. [PMID: 36002439 DOI: 10.1002/1873-3468.14480] [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: 04/06/2022] [Revised: 07/25/2022] [Accepted: 08/13/2022] [Indexed: 11/08/2022]
Abstract
Innate and adaptive immune systems are built-in homeostatic functions of many multicellular organisms and protect the host against foreign pathogens and infections. Dysregulation of the molecular mechanisms of the immune system can result in autoimmune diseases. The immune system can also be harnessed and manipulated to provide targeted cancer therapies, some of them relying on the blockade of immune-checkpoint receptors. Two prominent immune checkpoints, PD-1/PD-L1 and CTLA-4/CD80, comprise receptor-ligand pairs that prevent the host immune cells from attacking host tissues. However, cancer cells upregulate the respective PD-L1 and CD80 ligands for PD-1 and CTLA-4 and thereby evade the host-immune response. Therapeutic drugs that block PD-1/PD-L1 and CTLA-4/CD80 interactions re-enable the immune system to attack cancer cells, but their prognostic biomarker remains challenging. In this review, we discuss how the use of quantitative molecular imaging can be exploited to predict the response to anti-PD-1/PD-L1 therapies and to identify cancer patients who would benefit from them.
Collapse
Affiliation(s)
- James Miles
- Centre for Therapeutic Innovation, Cell Biophysics Laboratory, Department of Pharmacy and Pharmacology, Department of Physics, University of Bath, Claverton Down, Bath, United Kingdom.,Cell Biophysics Laboratory, Research Centre for Experimental Marine Biology and Biotechnology (PiE) & Instituto Biofisika, (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain.,Early Phase Trials and Sarcoma, Institut Bergonié, Cours de l'Argonne, Bordeaux, France.,FASTBASE Solutions S.L. Astondo Bidea, Scientific and Technology Park of Bizkaia, Derio, Spain
| | - Stephen G Ward
- Department of Pharmacy & Pharmacology, Centre for Therapeutic Innovation Leukocyte Biology Laboratory, University of Bath, Claverton Down, Bath, United Kingdom
| | - Banafshé Larijani
- Centre for Therapeutic Innovation, Cell Biophysics Laboratory, Department of Pharmacy and Pharmacology, Department of Physics, University of Bath, Claverton Down, Bath, United Kingdom.,Cell Biophysics Laboratory, Research Centre for Experimental Marine Biology and Biotechnology (PiE) & Instituto Biofisika, (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain
| |
Collapse
|
27
|
Zhang H, Karakas C, Tyburski H, Turner BM, Peng Y, Wang X, Katerji H, Schiffhauer L, Hicks DG. HER2-low breast cancers: Current insights and future directions. Semin Diagn Pathol 2022; 39:305-312. [PMID: 35872032 DOI: 10.1053/j.semdp.2022.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/11/2022]
Abstract
In light of the significant clinical benefits of novel HER2-targeting antibody-drug conjugates in advanced HER2-low expressing breast cancers in recent phases I and III clinical trials, particularly trastuzumab-deruxtecan (T-Dxd), the new "HER2-low" category in breast cancers (breast cancer with a HER2 IHC score of 1+, or 2+ without gene amplification) has gained increasing attention. In the past year, "HER2-low" breast cancers have been under active investigation by both oncologists and pathologists. In this current review, we update the recent cutting-edge research on HER2-low breast cancers, with a focus on the biology of HER2-low breast cancers, the issues on the identification of HER2-low breast cancers by immunohistochemistry in current practice of pathology, and the future directions in this emerging category in breast cancers.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States.
| | - Cansu Karakas
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| | - Haley Tyburski
- Class of 2024, University of Rochester, Rochester, NY, United States
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| | - Yan Peng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xi Wang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| | - Hani Katerji
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| | - Linda Schiffhauer
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| | - David G Hicks
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
28
|
Bang K, Cheon J, Park YS, Kim HD, Ryu MH, Park Y, Moon M, Lee H, Kang YK. Association between HER2 heterogeneity and clinical outcomes of HER2-positive gastric cancer patients treated with trastuzumab. Gastric Cancer 2022; 25:794-803. [PMID: 35524883 DOI: 10.1007/s10120-022-01298-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/04/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND The GASTHER1 study showed that re-evaluation of HER2 status rescued 8% of HER2-positive gastric cancer (GC) patients with initially HER2-negative GC. Since rescued HER2 positivity represents HER2 heterogeneity, we aimed to investigate this in a larger cohort with longer follow-up duration. METHODS Data of 153 HER2-positive advanced GC patients who received first-line trastuzumab-based chemotherapy were analyzed. Repeat endoscopic biopsy was performed in patients with initially HER2-negative GC. Survival outcomes were analyzed according to the immunohistochemistry (IHC) score (IHC 2+ /in situ hybridization [ISH] + vs IHC 3+), HER2 status (initially vs rescued HER2 positive), and H-score. RESULTS IHC 2+ /ISH + patients showed worse progression-free survival (PFS) and overall survival (OS) than those with IHC 3+ (p < 0.05). Rescued HER2-positive patients showed worse PFS and OS than initially HER2-positive patients (p < 0.05). Although survival outcomes were comparable according to HER2 status in IHC 2+ /ISH + patients, initially HER2-positive patients showed more favorable PFS and OS than rescued HER2-positive patients (p < 0.05) among those with IHC 3+ . Among the subgroups determined by HER2 status and IHC score, the initially IHC 3+ subgroup had the highest H-score. The low H-score group (H-score ≤ 210) had significantly worse survival outcomes than the high H-score group (H-score > 210) (p < 0.05). An H-score of ≤ 210 was independently associated with shorter OS (HR = 1.54, 95% CI 1.02-2.31, p = 0.04). CONCLUSIONS Rescued HER2-positive patients showed worse clinical outcomes than initially HER2-positive patients, especially those with IHC 3+ . This finding highlights the impact of HER2 heterogeneity, which can be quantified indirectly as an H-score.
Collapse
Affiliation(s)
- Kyunghye Bang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jaekyung Cheon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Yangsoon Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Meesun Moon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Hyungeun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
29
|
Cosuppression of NF-κB and AICDA Overcomes Acquired EGFR-TKI Resistance in Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14122940. [PMID: 35740609 PMCID: PMC9221089 DOI: 10.3390/cancers14122940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/04/2022] [Accepted: 06/12/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Since the first discovery of EGFR-tyrosine kinase inhibitors (TKIs), they have become the gold standard treatment for EGFR-mutated non-small cell lung cancer. However, the inevitable acquisition of secondary TKI resistance after treatment with TKIs remains an unresolved issue. Here, we evaluated the expression of NF-κB, AICDA, Akt, IL-6, Jak2, and Stat3 by EGFR-TKI-resistant lung adenocarcinoma (LAC), and found that NF-κB and AICDA are major players in the acquired resistance of lung cancer to TKIs. Therefore, treatment with an EGFR-TKI plus cosuppression of NF-κB and AICDA may be a promising strategy to overcome EGFR-TKI resistance in LACs. Abstract Background: Acquired resistance after EGFR-tyrosine kinase inhibitor (TKI) treatment is the rule rather than the exception. Overcoming resistance to EGFR-TKIs is essential if we are to develop better therapeutic strategies for lung cancer patients. Here, we examine the effector signaling pathways underlying TKI resistance and propose targets to overcome the resistance of lung adenocarcinoma (LAC) to TKI. Methods: We compared the expression of NF-κB, AICDA, Akt, IL-6, Jak2, and Stat3 by EGFR-TKI-resistant and EGFR-TKI-sensitive LAC cell lines, and by LAC patients treated with EGFR-TKIs; we then evaluated links between expression and treatment responses. We also examined the therapeutic effects of NF-κB and AICDA inhibition in EGFR-TKI-resistant LACs. Results: NF-κB and AICDA were more expressed by EGFR-TKI-resistant LACs than by EGFR-TKI-sensitive LACs. EGFR-TKIs induced a dose-dependent increase in the expression of NF-κB, AICDA, and IL-6. Inhibition of NF-κB suppressed the expression of AICDA, Akt, and IL-6 in EGFR-TKI-resistant and EGFR-TKI-sensitive LACs, whereas knockdown of AICDA suppressed the expression of NF-κB and Akt in both cell types. Treating EGFR-TKI-resistant LACs with an EGFR-TKI, alongside cosuppression of NF-κB and AICDA, had a significant therapeutic effect. Conclusion: Treatment with an EGFR-TKI plus cosuppression of NF-κB and AICDA may be a promising strategy to overcome EGFR-TKI resistance in LACs.
Collapse
|
30
|
Kanbar M, de Michele F, Poels J, Van Loo S, Giudice MG, Gilet T, Wyns C. Microfluidic and Static Organotypic Culture Systems to Support Ex Vivo Spermatogenesis From Prepubertal Porcine Testicular Tissue: A Comparative Study. Front Physiol 2022; 13:884122. [PMID: 35721544 PMCID: PMC9201455 DOI: 10.3389/fphys.2022.884122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Background:In vitro maturation of immature testicular tissue (ITT) cryopreserved for fertility preservation is a promising fertility restoration strategy. Organotypic tissue culture proved successful in mice, leading to live births. In larger mammals, including humans, efficiently reproducing spermatogenesis ex vivo remains challenging. With advances in biomaterials technology, culture systems are becoming more complex to better mimic in vivo conditions. Along with improving culture media components, optimizing physical culture conditions (e.g., tissue perfusion, oxygen diffusion) also needs to be considered. Recent studies in mice showed that by using silicone-based hybrid culture systems, the efficiency of spermatogenesis can be improved. Such systems have not been reported for ITT of large mammals. Methods: Four different organotypic tissue culture systems were compared: static i.e., polytetrafluoroethylene membrane inserts (OT), agarose gel (AG) and agarose gel with polydimethylsiloxane chamber (AGPC), and dynamic i.e., microfluidic (MF). OT served as control. Porcine ITT fragments were cultured over a 30-day period using a single culture medium. Analyses were performed at days (d) 0, 5, 10, 20 and 30. Seminiferous tubule (ST) integrity, diameters, and tissue core integrity were evaluated on histology. Immunohistochemistry was used to identify germ cells (PGP9.5, VASA, SYCP3, CREM), somatic cells (SOX9, INSL3) and proliferating cells (Ki67), and to assess oxidative stress (MDA) and apoptosis (C-Caspase3). Testosterone was measured in supernatants using ELISA. Results: ITT fragments survived and grew in all systems. ST diameters, and Sertoli cell (SOX9) numbers increased, meiotic (SYCP3) and post-meiotic (CREM) germ cells were generated, and testosterone was secreted. When compared to control (OT), significantly larger STs (d10 through d30), better tissue core integrity (d5 through d20), higher numbers of undifferentiated spermatogonia (d30), meiotic and post-meiotic germ cells (SYCP3: d20 and 30, CREM: d20) were observed in the AGPC system. Apoptosis, lipid peroxidation (MDA), ST integrity, proliferating germ cell (Ki67/VASA) numbers, Leydig cell (INSL3) numbers and testosterone levels were not significantly different between systems. Conclusions: Using a modified culture system (AGPC), germ cell survival and the efficiency of porcine germ cell differentiation were moderately improved ex vivo. We assume that further optimization can be obtained with concomitant modifications in culture media components.
Collapse
Affiliation(s)
- Marc Kanbar
- Andrology Lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Francesca de Michele
- Andrology Lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jonathan Poels
- Andrology Lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Stéphanie Van Loo
- Microfluidics Lab, Department of Aerospace and Mechanical Engineering, University of Liege, Liege, Belgium
| | - Maria Grazia Giudice
- Andrology Lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Tristan Gilet
- Microfluidics Lab, Department of Aerospace and Mechanical Engineering, University of Liege, Liege, Belgium
| | - Christine Wyns
- Andrology Lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- *Correspondence: Christine Wyns,
| |
Collapse
|
31
|
Miglietta F, Griguolo G, Bottosso M, Giarratano T, Lo Mele M, Fassan M, Cacciatore M, Genovesi E, De Bartolo D, Vernaci G, Amato O, Porra F, Conte P, Guarneri V, Dieci MV. HER2-low-positive breast cancer: evolution from primary tumor to residual disease after neoadjuvant treatment. NPJ Breast Cancer 2022; 8:66. [PMID: 35595761 PMCID: PMC9122970 DOI: 10.1038/s41523-022-00434-w] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/14/2022] [Indexed: 11/09/2022] Open
Abstract
Approximately a half of breast tumors classified as HER2-negative exhibit HER2-low-positive expression. We recently described a high instability of HER2-low-positive expression from primary breast cancer (BC) to relapse. Previous studies reporting discordance in HER2 status between baseline biopsy and residual disease (RD) in patients undergoing neoadjuvant treatment did not include the HER2-low-positive category. The aim of this study is to track the evolution of HER2-low-positive expression from primary BC to RD after neoadjuvant treatment. Patients undergoing neoadjuvant treatment with available baseline tumor tissue and matched samples of RD (in case of no pCR) were included. HER2-negative cases were sub-classified as HER2-0 or HER2-low-positive (IHC 1+ or 2+ and ISH negative). Four-hundred forty-six patients were included. Primary BC phenotype was: HR-positive/HER2-negative 23.5%, triple-negative (TN) 35%, HER2-positive 41.5%. HER2-low-positive cases were 55.6% of the HER2-negative cohort and were significantly enriched in the HR-positive/HER2-negative vs. TN subgroup (68.6% vs. 46.8%, p = 0.001 χ2 test). In all, 35.3% of non-pCR patients (n = 291) had a HER2-low-positive expression on RD. The overall rate of HER2 expression discordance was 26.4%, mostly driven by HER2-negative cases converting either from (14.8%) or to (8.9%) HER2-low-positive phenotype. Among HR-positive/HER2-negative patients with HER2-low-positive expression on RD, 32.0% and 57.1% had an estimated high risk of relapse according to the residual proliferative cancer burden and CPS-EG score, respectively. In conclusion, HER2-low-positive expression showed high instability from primary BC to RD after neoadjuvant treatment. HER2-low-positive expression on RD may guide personalized adjuvant treatment for high-risk patients in the context of clinical trials with novel anti-HER2 antibody-drug conjugates.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Michele Bottosso
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Tommaso Giarratano
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Marcello Lo Mele
- Surgical Pathology Unit, University Hospital of Padua, 35121, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121, Padua, Italy
- Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - Matilde Cacciatore
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Elisa Genovesi
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Debora De Bartolo
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121, Padua, Italy
| | - Grazia Vernaci
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Ottavia Amato
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Francesca Porra
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - PierFranco Conte
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy.
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy.
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35128, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128, Padova, Italy
| |
Collapse
|
32
|
Kang EY, Millstein J, Popovic G, Meagher NS, Bolithon A, Talhouk A, Chiu DS, Anglesio MS, Leung B, Tang K, Lambie N, Pavanello M, Da-Anoy A, Lambrechts D, Loverix L, Olbrecht S, Bisinotto C, Garcia-Donas J, Ruiz-Llorente S, Yagüe-Fernandez M, Edwards RP, Elishaev E, Olawaiye A, Taylor S, Ataseven B, du Bois A, Harter P, Lester J, Høgdall CK, Armasu SM, Huang Y, Vierkant RA, Wang C, Winham SJ, Heublein S, Kommoss FKF, Cramer DW, Sasamoto N, van-Wagensveld L, Lycke M, Mateoiu C, Joseph J, Pike MC, Odunsi K, Tseng CC, Pearce CL, Bilic S, Conrads TP, Hartmann A, Hein A, Jones ME, Leung Y, Beckmann MW, Ruebner M, Schoemaker MJ, Terry KL, El-Bahrawy MA, Coulson P, Etter JL, LaVigne-Mager K, Andress J, Grube M, Fischer A, Neudeck N, Robertson G, Farrell R, Barlow E, Quinn C, Hettiaratchi A, Casablanca Y, Erber R, Stewart CJR, Tan A, Yu Y, Boros J, Brand AH, Harnett PR, Kennedy CJ, Nevins N, Morgan T, Fasching PA, Vergote I, Swerdlow AJ, Candido Dos Reis FJ, Maxwell GL, Neuhausen SL, Barquin-Garcia A, Modugno F, Moysich KB, Crowe PJ, Hirasawa A, Heitz F, Karlan BY, Goode EL, Sinn P, Horlings HM, Høgdall E, Sundfeldt K, Kommoss S, Staebler A, et alKang EY, Millstein J, Popovic G, Meagher NS, Bolithon A, Talhouk A, Chiu DS, Anglesio MS, Leung B, Tang K, Lambie N, Pavanello M, Da-Anoy A, Lambrechts D, Loverix L, Olbrecht S, Bisinotto C, Garcia-Donas J, Ruiz-Llorente S, Yagüe-Fernandez M, Edwards RP, Elishaev E, Olawaiye A, Taylor S, Ataseven B, du Bois A, Harter P, Lester J, Høgdall CK, Armasu SM, Huang Y, Vierkant RA, Wang C, Winham SJ, Heublein S, Kommoss FKF, Cramer DW, Sasamoto N, van-Wagensveld L, Lycke M, Mateoiu C, Joseph J, Pike MC, Odunsi K, Tseng CC, Pearce CL, Bilic S, Conrads TP, Hartmann A, Hein A, Jones ME, Leung Y, Beckmann MW, Ruebner M, Schoemaker MJ, Terry KL, El-Bahrawy MA, Coulson P, Etter JL, LaVigne-Mager K, Andress J, Grube M, Fischer A, Neudeck N, Robertson G, Farrell R, Barlow E, Quinn C, Hettiaratchi A, Casablanca Y, Erber R, Stewart CJR, Tan A, Yu Y, Boros J, Brand AH, Harnett PR, Kennedy CJ, Nevins N, Morgan T, Fasching PA, Vergote I, Swerdlow AJ, Candido Dos Reis FJ, Maxwell GL, Neuhausen SL, Barquin-Garcia A, Modugno F, Moysich KB, Crowe PJ, Hirasawa A, Heitz F, Karlan BY, Goode EL, Sinn P, Horlings HM, Høgdall E, Sundfeldt K, Kommoss S, Staebler A, Wu AH, Cohen PA, DeFazio A, Lee CH, Steed H, Le ND, Gayther SA, Lawrenson K, Pharoah PDP, Konecny G, Cook LS, Ramus SJ, Kelemen LE, Köbel M. MCM3 is a novel proliferation marker associated with longer survival for patients with tubo-ovarian high-grade serous carcinoma. Virchows Arch 2022; 480:855-871. [PMID: 34782936 PMCID: PMC9035053 DOI: 10.1007/s00428-021-03232-0] [Show More Authors] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 12/14/2022]
Abstract
Tubo-ovarian high-grade serous carcinomas (HGSC) are highly proliferative neoplasms that generally respond well to platinum/taxane chemotherapy. We recently identified minichromosome maintenance complex component 3 (MCM3), which is involved in the initiation of DNA replication and proliferation, as a favorable prognostic marker in HGSC. Our objective was to further validate whether MCM3 mRNA expression and possibly MCM3 protein levels are associated with survival in patients with HGSC. MCM3 mRNA expression was measured using NanoString expression profiling on formalin-fixed and paraffin-embedded tissue (N = 2355 HGSC) and MCM3 protein expression was assessed by immunohistochemistry (N = 522 HGSC) and compared with Ki-67. Kaplan-Meier curves and the Cox proportional hazards model were used to estimate associations with survival. Among chemotherapy-naïve HGSC, higher MCM3 mRNA expression (one standard deviation increase in the score) was associated with longer overall survival (HR = 0.87, 95% CI 0.81-0.92, p < 0.0001, N = 1840) in multivariable analysis. MCM3 mRNA expression was highest in the HGSC C5.PRO molecular subtype, although no interaction was observed between MCM3, survival and molecular subtypes. MCM3 and Ki-67 protein levels were significantly lower after exposure to neoadjuvant chemotherapy compared to chemotherapy-naïve tumors: 37.0% versus 46.4% and 22.9% versus 34.2%, respectively. Among chemotherapy-naïve HGSC, high MCM3 protein levels were also associated with significantly longer disease-specific survival (HR = 0.52, 95% CI 0.36-0.74, p = 0.0003, N = 392) compared to cases with low MCM3 protein levels in multivariable analysis. MCM3 immunohistochemistry is a promising surrogate marker of proliferation in HGSC.
Collapse
Affiliation(s)
- Eun Young Kang
- Department of Pathology and Laboratory Medicine, Foothills Medical Center, University of Calgary, Calgary, AB, Canada
| | - Joshua Millstein
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Gordana Popovic
- Mark Wainwright Analytical Centre, Stats Central, University of New South Wales Sydney, Sydney, Australia
| | - Nicola S Meagher
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, NSW, Australia
| | - Adelyn Bolithon
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, NSW, Australia
| | - Aline Talhouk
- British Columbia's Ovarian Cancer Research (OVCARE) Program, BC Cancer, Vancouver General Hospital, and University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Derek S Chiu
- British Columbia's Ovarian Cancer Research (OVCARE) Program, BC Cancer, Vancouver General Hospital, and University of British Columbia, Vancouver, BC, Canada
| | - Michael S Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
- Histopathology/ISH Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Betty Leung
- Prince of Wales Clinical School, University of NSW Sydney, Sydney, NSW, Australia
| | - Katrina Tang
- Department of Anatomical Pathology, Prince of Wales Hospital, Sydney, Australia
| | - Neil Lambie
- NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
| | - Marina Pavanello
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Annalyn Da-Anoy
- Department of Pathology and Laboratory Medicine, Foothills Medical Center, University of Calgary, Calgary, AB, Canada
| | - Diether Lambrechts
- VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, University of Leuven, Leuven, Belgium
| | - Liselore Loverix
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Siel Olbrecht
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Christiani Bisinotto
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jesus Garcia-Donas
- HM Sanchinarro Centro Integral Oncológico Clara Campal, University Hospital, Madrid, Spain
| | - Sergio Ruiz-Llorente
- HM Sanchinarro Centro Integral Oncológico Clara Campal, University Hospital, Madrid, Spain
| | - Monica Yagüe-Fernandez
- HM Sanchinarro Centro Integral Oncológico Clara Campal, University Hospital, Madrid, Spain
| | - Robert P Edwards
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Esther Elishaev
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexander Olawaiye
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Beyhan Ataseven
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte (KEM), Essen, Germany
- Department of Gynecology and Obstetrics, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte (KEM), Essen, Germany
- Department of Gynecology and Gynecological Oncology, Dr. Horst-Schmidt Klinik Wiesbaden, Wiesbaden, Germany
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte (KEM), Essen, Germany
- Department of Gynecology and Gynecological Oncology, Dr. Horst-Schmidt Klinik Wiesbaden, Wiesbaden, Germany
| | - Jenny Lester
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Claus K Høgdall
- Department of Gynaecology, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Sebastian M Armasu
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Yajue Huang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Robert A Vierkant
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Chen Wang
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Stacey J Winham
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Sabine Heublein
- Department of Obstetrics and Gynecology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix K F Kommoss
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel W Cramer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Naoko Sasamoto
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lilian van-Wagensveld
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maria Lycke
- Department of Obstetrics and Gynecology, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - Constantina Mateoiu
- Department of Pathology and Cytology, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - Janine Joseph
- Division of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Malcolm C Pike
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Kunle Odunsi
- Department of Oncology, University of Chicago, Chicago, IL, USA
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Chiu-Chen Tseng
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Celeste L Pearce
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sanela Bilic
- Department of Gynaecological Oncology, St John of God Subiaco Hospital, Subiaco, Australia
| | - Thomas P Conrads
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - Arndt Hartmann
- Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen-Nürnberg, Germany
| | - Alexander Hein
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Michael E Jones
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Yee Leung
- Division of Obstetrics and Gynaecology, Faculty of Health and Medical Sciences, University of Western Australia, Crawley, WA, Australia
- Department of Gynaecological Oncology, King Edward Memorial Hospital, Subiaco, Australia
- Australia New Zealand Gynaecological Oncology Group, Camperdown, Australia
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Minouk J Schoemaker
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Kathryn L Terry
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mona A El-Bahrawy
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Penny Coulson
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - John L Etter
- Division of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Juergen Andress
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Marcel Grube
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Anna Fischer
- Institute of Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Nina Neudeck
- Institute of Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Greg Robertson
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia
- St George Private Hospital, Kogarah, Australia
| | | | - Ellen Barlow
- Gynaecological Cancer Centre, Royal Hospital for Women, Sydney, Australia
| | - Carmel Quinn
- Mark Wainwright Analytical Centre, Stats Central, University of New South Wales Sydney, Sydney, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, NSW, Australia
- Prince of Wales Clinical School, University of NSW Sydney, Sydney, NSW, Australia
- Translational Cancer Research Network, University of New South Wales Sydney, Sydney, Australia
- UNSW Biorepository, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Sydney, Australia
| | - Anusha Hettiaratchi
- UNSW Biorepository, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Sydney, Australia
| | - Yovanni Casablanca
- Uniformed Services University of the Health Sciences, USAF, Bethesda, MD, USA
| | - Ramona Erber
- Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen-Nürnberg, Germany
| | - Colin J R Stewart
- School for Women's and Infants' Health, University of Western Australia, Perth, Australia
| | - Adeline Tan
- Division of Obstetrics and Gynaecology, Faculty of Health and Medical Sciences, University of Western Australia, Crawley, WA, Australia
- Western Women's Pathology, Western Diagnostic Pathology, Wembley, Australia
| | - Yu Yu
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Jessica Boros
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Alison H Brand
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Paul R Harnett
- The Crown Princess Mary Cancer Centre Westmead, Sydney-West Cancer Network, Westmead Hospital, Sydney, NSW, Australia
- The University of Sydney, Sydney, Australia
| | - Catherine J Kennedy
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Nikilyn Nevins
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
- The University of Sydney, Sydney, Australia
| | - Terry Morgan
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Ignace Vergote
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Francisco J Candido Dos Reis
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - G Larry Maxwell
- Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | | | - Francesmary Modugno
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Kirsten B Moysich
- Division of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Philip J Crowe
- Prince of Wales Clinical School, University of NSW Sydney, Sydney, NSW, Australia
| | - Akira Hirasawa
- Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Florian Heitz
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte (KEM), Essen, Germany
- Department of Gynecology and Gynecological Oncology, Dr. Horst-Schmidt Klinik Wiesbaden, Wiesbaden, Germany
- Department for Gynecology with the Center for Oncologic Surgery, Charité Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Beth Y Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Ellen L Goode
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hugo M Horlings
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Estrid Høgdall
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pathology, Molecular Unit, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Karin Sundfeldt
- Department of Obstetrics and Gynecology, Sahlgrenska Center for Cancer Research, Gothenburg University, Gothenburg, Sweden
| | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Annette Staebler
- Institute of Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Anna H Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Paul A Cohen
- Division of Obstetrics and Gynaecology, Faculty of Health and Medical Sciences, University of Western Australia, Crawley, WA, Australia
- Bendat Family Comprehensive Cancer Centre, St John of God Subiaco Hospital, Subiaco, WA, Australia
| | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
- The Crown Princess Mary Cancer Centre Westmead, Sydney-West Cancer Network, Westmead Hospital, Sydney, NSW, Australia
- The University of Sydney, Sydney, Australia
| | - Cheng-Han Lee
- Department of Pathology and Laboratory Medicine, University of Alberta, Edmonton, AB, Canada
| | - Helen Steed
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Royal Alexandra Hospital, Edmonton, AB, Canada
| | - Nhu D Le
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Simon A Gayther
- Center for Bioinformatics and Functional Genomics and the Cedars Sinai Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kate Lawrenson
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Women's Cancer Program at the Samuel Oschin Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul D P Pharoah
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Gottfried Konecny
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Linda S Cook
- School of Public Health, University of Colorado, Aurora, CO, USA
- Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, AB, Canada
| | - Susan J Ramus
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, NSW, Australia
| | - Linda E Kelemen
- Bureau of Population Health Data Analytics & Informatics, South Carolina Department of Health and Environmental Control, Columbia, SC, USA
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, Foothills Medical Center, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
33
|
Zhang H, Katerji H, Turner BM, Hicks DG. HER2-Low Breast Cancers. Am J Clin Pathol 2022; 157:328-336. [PMID: 34519765 DOI: 10.1093/ajcp/aqab117] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/31/2021] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Recent clinical trials have demonstrated significant clinical benefits from novel therapeutic compounds in breast cancer patient with human epidermal growth factor receptor 2 (HER2) immunohistochemical (IHC) score of 1+ or 2+ and negative in situ hybridization (ISH) result. A new concept of "HER2-low" breast cancer has been proposed and applied in the recent and ongoing clinical trials. In this article, we review the literature on the topic of HER2-low breast cancer. METHODS A literature search in PubMed was performed using key words related to HER2-low breast cancer. Major relevant studies that were presented in international breast cancer conferences were also included. RESULTS HER2-low breast cancer is currently defined as breast cancer with HER2 IHC score of 1+ or 2+ and negative ISH result. It likely represents a group of tumors with significant biological heterogeneity. Reports of clinical activity using the next generation of HER2-targeting antibody-drug conjugates in HER2-low breast cancers suggest that some strategies of targeting HER2 might be effective in this patient population while raising considerable concerns over limitations in our current testing methodologies and our ability to accurately identify such patients. CONCLUSIONS The promising efficacy of novel HER2-targeted therapy in advanced HER2-low breast cancers has raised the possibility for changing the clinical interpretation of HER2 status in breast cancer to include a HER2-low category; however, the definition of HER2-low breast cancer, the corresponding reliable and accurate quantitative HER2 testing methodology, and the biology of HER2-low breast cancer remain poorly defined.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Hani Katerji
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - David G Hicks
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
34
|
QuantISH: RNA in situ hybridization image analysis framework for quantifying cell type-specific target RNA expression and variability. J Transl Med 2022; 102:753-761. [PMID: 35169222 PMCID: PMC9249626 DOI: 10.1038/s41374-022-00743-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
RNA in situ hybridization (RNA-ISH) is a powerful spatial transcriptomics technology to characterize target RNA abundance and localization in individual cells. This allows analysis of tumor heterogeneity and expression localization, which are not readily obtainable through transcriptomic data analysis. RNA-ISH experiments produce large amounts of data and there is a need for automated analysis methods. Here we present QuantISH, a comprehensive open-source RNA-ISH image analysis pipeline that quantifies marker expressions in individual carcinoma, immune, and stromal cells on chromogenic or fluorescent in situ hybridization images. QuantISH is designed to be modular and can be adapted to various image and sample types and staining protocols. We show that in chromogenic RNA in situ hybridization images of high-grade serous carcinoma (HGSC) QuantISH cancer cell classification has high precision, and signal expression quantification is in line with visual assessment. We further demonstrate the power of QuantISH by showing that CCNE1 average expression and DDIT3 expression variability, as captured by the variability factor developed herein, act as candidate biomarkers in HGSC. Altogether, our results demonstrate that QuantISH can quantify RNA expression levels and their variability in carcinoma cells, and thus paves the way to utilize RNA-ISH technology.
Collapse
|
35
|
Sato R, Ohmori K, Umetsu M, Takao M, Tano M, Grant G, Porter B, Bet A, Terasaki T, Uchida Y. An Atlas of the Quantitative Protein Expression of Anti-Epileptic-Drug Transporters, Metabolizing Enzymes and Tight Junctions at the Blood-Brain Barrier in Epileptic Patients. Pharmaceutics 2021; 13:pharmaceutics13122122. [PMID: 34959403 PMCID: PMC8708024 DOI: 10.3390/pharmaceutics13122122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/02/2021] [Accepted: 12/08/2021] [Indexed: 01/06/2023] Open
Abstract
The purpose of the present study was to quantitatively elucidate the levels of protein expression of anti-epileptic-drug (AED) transporters, metabolizing enzymes and tight junction molecules at the blood–brain barrier (BBB) in the focal site of epilepsy patients using accurate SWATH (sequential window acquisition of all theoretical fragment ion spectra) proteomics. Brain capillaries were isolated from focal sites in six epilepsy patients and five normal brains; tryptic digests were produced and subjected to SWATH analysis. MDR1 and BCRP were significantly downregulated in the epilepsy group compared to the normal group. Out of 16 AED-metabolizing enzymes detected, the protein expression levels of GSTP1, GSTO1, CYP2E1, ALDH1A1, ALDH6A1, ALDH7A1, ALDH9A1 and ADH5 were significantly 2.13-, 6.23-, 2.16-, 2.80-, 1.73-, 1.67-, 2.47- and 2.23-fold greater in the brain capillaries of epileptic patients than those of normal brains, respectively. The protein expression levels of Claudin-5, ZO-1, Catenin alpha-1, beta-1 and delta-1 were significantly lower, 1.97-, 2.51-, 2.44-, 1.90- and 1.63-fold, in the brain capillaries of epileptic patients compared to those of normal brains, respectively. Consistent with these observations, leakage of blood proteins was also observed. These results provide for a better understanding of the therapeutic effect of AEDs and molecular mechanisms of AED resistance in epileptic patients.
Collapse
Affiliation(s)
- Risa Sato
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Kotaro Ohmori
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Mina Umetsu
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Masaki Takao
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki 372-0006, Japan; (M.T.); (M.T.)
- Department of Clinical Laboratory, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira 187-8551, Japan
| | - Mitsutoshi Tano
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki 372-0006, Japan; (M.T.); (M.T.)
| | - Gerald Grant
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (G.G.); (A.B.)
| | - Brenda Porter
- Department of Neurology, Stanford University, Stanford, CA 94305, USA;
| | - Anthony Bet
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (G.G.); (A.B.)
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
- Correspondence: ; Tel.: +81-22-795-6832
| |
Collapse
|
36
|
Imron A, Hernowo B, Hilmanto D, Wiriadisastra K, Hermanto Y. The Effects of Glucagon and Insulin Combination toward on Neurodegeneration Following Traumatic Brain Injury in Rat Model. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Traumatic brain injury (TBI) is a major cause of death and disability in the productive age. Glutamate excitotoxicity and hyperglycemia those occur following TBI are among the factors those influence secondary brain injury.
AIM: This study aimed to determine the effect of glucagon and insulin combination on neuronal necrosis following TBI.
METHODS: A total of 28 male wistar rats were randomized into four experimental groups: placebo, insulin, glucagon, and combination of glucagon and insulin. Each animal underwent controlled cortical impact model of TBI. The blood glucose and glutamate levels were measured before and 4 h following TBI. The brain tissues were collected to evaluate neuronal necrosis.
RESULTS: Glucagon or glucagon and insulin combination were able to prevent the increased of blood glutamate levels following TBI (p < 0.05). Glucagon administration was associated high blood glucose level (198.10 ± 32.58 mg/dL); a combination with insulin was able to minimize the increased of blood glucose level (166.53 ± 18.48 mg/dL). Combination of glucagon and insulin had a lower number of neuronal necrosis compare to the other groups (p < 0.005).
CONCLUSION: The combination of glucagon and insulin potentially exhibit neuroprotection effect on rats following TBI as being demonstrated by lower number of neuronal necrosis. This finding further indicates the role of glucose homeostasis in neuroprotection.
Collapse
|
37
|
Hana S, Peterson M, McLaughlin H, Marshall E, Fabian AJ, McKissick O, Koszka K, Marsh G, Craft M, Xu S, Sorets A, Torregrosa T, Sun C, Henderson CE, Lo SC. Highly efficient neuronal gene knockout in vivo by CRISPR-Cas9 via neonatal intracerebroventricular injection of AAV in mice. Gene Ther 2021; 28:646-658. [PMID: 33558692 PMCID: PMC8599009 DOI: 10.1038/s41434-021-00224-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/29/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
CRISPR-Cas systems have emerged as a powerful tool to generate genetic models for studying normal and diseased central nervous system (CNS). Targeted gene disruption at specific loci has been demonstrated successfully in non-dividing neurons. Despite its simplicity, high specificity and low cost, the efficiency of CRISPR-mediated knockout in vivo can be substantially impacted by many parameters. Here, we used CRISPR-Cas9 to disrupt the neuronal-specific gene, NeuN, and optimized key parameters to achieve effective gene knockout broadly in the CNS in postnatal mice. Three cell lines and two primary neuron cultures were used to validate the disruption of NeuN by single-guide RNAs (sgRNA) harboring distinct spacers and scaffold sequences. This triage identified an optimal sgRNA design with the highest NeuN disruption in in vitro and in vivo systems. To enhance CRISPR efficiency, AAV-PHP.B, a vector with superior neuronal transduction, was used to deliver this sgRNA in Cas9 mice via neonatal intracerebroventricular (ICV) injection. This approach resulted in 99.4% biallelic indels rate in the transduced cells, leading to greater than 70% reduction of total NeuN proteins in the cortex, hippocampus and spinal cord. This work contributes to the optimization of CRISPR-mediated knockout and will be beneficial for fundamental and preclinical research.
Collapse
|
38
|
Miglietta F, Griguolo G, Bottosso M, Giarratano T, Lo Mele M, Fassan M, Cacciatore M, Genovesi E, De Bartolo D, Vernaci G, Amato O, Conte P, Guarneri V, Dieci MV. Evolution of HER2-low expression from primary to recurrent breast cancer. NPJ Breast Cancer 2021; 7:137. [PMID: 34642348 PMCID: PMC8511010 DOI: 10.1038/s41523-021-00343-4] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/28/2021] [Indexed: 12/20/2022] Open
Abstract
About a half of HER2-negative breast cancer (BC) show HER2-low expression that can be targeted by new antibody-drug conjugates. The main aim of this study is to describe the evolution of HER2 expression from primary BC to relapse by including HER2-low category in both primary and recurrent BC samples. Patients with matched primary and relapse BC samples were included. HER2 was evaluated according to ASCO/CAP recommendations in place at the time of diagnosis. A cutoff of >10% cells staining for HER2-positivity was applied. HER2-negative cases were sub-classified as HER2-low (IHC = 1 + /2+ and ISH not amplified), or HER2-0 (IHC-0). 547 patients were included. The proportion of HER2-low cases was 34.2% on the primary tumor and 37.3% on the relapse samples. Among HER2-negative cases, HER2-low status was more frequent in HR-positive vs triple-negative tumors (47.3% vs 35.4% on primary tumor samples, 53.8% vs 36.2% on relapse samples). The overall rate of HER2 discordance was 38.0%, mostly represented by HER2-0 switching to HER2-low (15%) and HER2-low switching to HER2-0 (14%). Among patients with a primary HER2-negative tumor, the rate of HER2 discordance was higher in HR-positive/HER2-negative vs triple-negative cases (45.5% vs 36.7% p = 0.170). This difference was mostly driven by cases switching from HER2-0 to HER2-low. HER2-low expression is highly unstable during disease evolution. Relapse biopsy in case of a primary HER2-0 tumor may open new therapeutic opportunities in a relevant proportion of patients.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Michele Bottosso
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | | | - Marcello Lo Mele
- Surgical Pathology Unit, University Hospital of Padua, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Matilde Cacciatore
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Elisa Genovesi
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Debora De Bartolo
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Grazia Vernaci
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Ottavia Amato
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - PierFranco Conte
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy.
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| |
Collapse
|
39
|
Floranović MP, Petrović AR, Veličković LJ. Expression of the CXCR4 and CXCR7 in renal cancers; can "the orphan receptor" predict the mortality? Ann Diagn Pathol 2021; 55:151829. [PMID: 34563828 DOI: 10.1016/j.anndiagpath.2021.151829] [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] [Received: 06/23/2021] [Revised: 08/26/2021] [Accepted: 09/05/2021] [Indexed: 02/05/2023]
Abstract
CXCR4 and CXCR7 are chemokine receptors that bind with chemokine CXCL12 and influence various physiological and pathological processes. In renal cell carcinoma, their expression has been mostly associated with tumour aggressiveness. However, there are some contradictory results regarding the localization of immunohistochemical staining and predictive potential of these markers. The expression of CXCR4 and CXCR7 was immunohistochemicaly analyzed in 98 tumour samples, including 85 clear cell type (ccRCC) and 13 papillary type (pRCC). Depending on the staining localization (cytoplasmatic or membranous), intensity and percentage of stained cells, histoscores were calculated, and their association with clinicopathological parameters was analyzed. PRCC was associated with both CXCR7 and CXCR4 cytoplasmatic expression. We have also found that higher CXCR7 expression can be expected in tumours of greater size. In our study, mortality could be predicted by membranous CXCR7 histoscore, tumour size and pRCC type. With each centimetre in tumour size, survival decreases 1.2 times. CXCR7M histoscore higher by 50 units was associated with 1.5 greater risk of mortality. Neither membranous nor cytoplasmatic CXCR4 histoscore was found to be mortality predictor. Our data showed that CXCR7 could be considered as a valid prognostic marker regarding survival of RCC patients.
Collapse
Affiliation(s)
- Milena Potić Floranović
- Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Zoran Đinđić Boulevard 81, 18000 Niš, Serbia.
| | - Ana Ristić Petrović
- Pathology and Pathological Anatomy Center - Clinical Center of Niš, Zoran Đinđić Boulevard 48, 18000 Niš, Serbia
| | - Ljubinka Janković Veličković
- Pathology and Pathological Anatomy Center - Clinical Center of Niš, Zoran Đinđić Boulevard 48, 18000 Niš, Serbia
| |
Collapse
|
40
|
Carbonara K, Andonovski M, Coorssen JR. Proteomes Are of Proteoforms: Embracing the Complexity. Proteomes 2021; 9:38. [PMID: 34564541 PMCID: PMC8482110 DOI: 10.3390/proteomes9030038] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/24/2021] [Accepted: 08/29/2021] [Indexed: 12/17/2022] Open
Abstract
Proteomes are complex-much more so than genomes or transcriptomes. Thus, simplifying their analysis does not simplify the issue. Proteomes are of proteoforms, not canonical proteins. While having a catalogue of amino acid sequences provides invaluable information, this is the Proteome-lite. To dissect biological mechanisms and identify critical biomarkers/drug targets, we must assess the myriad of proteoforms that arise at any point before, after, and between translation and transcription (e.g., isoforms, splice variants, and post-translational modifications [PTM]), as well as newly defined species. There are numerous analytical methods currently used to address proteome depth and here we critically evaluate these in terms of the current 'state-of-the-field'. We thus discuss both pros and cons of available approaches and where improvements or refinements are needed to quantitatively characterize proteomes. To enable a next-generation approach, we suggest that advances lie in transdisciplinarity via integration of current proteomic methods to yield a unified discipline that capitalizes on the strongest qualities of each. Such a necessary (if not revolutionary) shift cannot be accomplished by a continued primary focus on proteo-genomics/-transcriptomics. We must embrace the complexity. Yes, these are the hard questions, and this will not be easy…but where is the fun in easy?
Collapse
Affiliation(s)
| | | | - Jens R. Coorssen
- Faculties of Applied Health Sciences and Mathematics & Science, Departments of Health Sciences and Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (K.C.); (M.A.)
| |
Collapse
|
41
|
Mojtahedzadeh S, Opsahl A, Aguilar JK, Li D, Streiner N, Wang J, Trajkovic D, Boucher G, Coskran T, O'Neil SP, Ram S. Characterizing Intra-Tumor and Inter-Tumor Variability of Immune Cell Infiltrates in Murine Syngeneic Tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2133-2146. [PMID: 34428423 DOI: 10.1016/j.ajpath.2021.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022]
Abstract
Murine tumors are indispensable model systems in preclinical immuno-oncology research. While immunologic heterogeneity is well-known to be an important factor that can influence treatment outcome, there is a severe paucity of data concerning the nature of this heterogeneity in murine tumor models. Using serial sectioning methodology combined with IHC analysis and whole-slide image analysis, the depth-dependent variation in immune-cell abundance in tumor specimens was investigated at single-cell resolution. Specifically, intra- and intertumor variability in cell density of nine immune-cell biomarkers was quantified in multiple murine tumor models. The analysis showed that intertumor variability was typically the dominant source of variation in measurements of immune-cell densities. Statistical power analysis revealed the effect of group size and variance in immune-cell density on the predictive power of detecting a statistically meaningful fold-change in immune-cell density. Intertumor variability in the ratio of immune-cell densities showed distinct patterns in select tumor models and revealed the existence of strong correlations between select biomarker pairs. Furthermore, the relative proportion of immune cells at different depths across tumor samples was preserved in some but not all tumor models, thereby revealing the existence of compositional heterogeneity. Taken together, these results reveal novel insights into the nature of immunologic heterogeneity, which is not accessible through typical omics approaches.
Collapse
MESH Headings
- Animals
- Biological Variation, Individual
- Cell Count
- Chemotaxis, Leukocyte/physiology
- Female
- Gene Expression Regulation, Neoplastic
- Immunophenotyping
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Transplantation, Isogeneic
- Tumor Cells, Cultured
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Sepideh Mojtahedzadeh
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California
| | - Alan Opsahl
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California
| | - Joan-Kristel Aguilar
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California
| | - Dingzhou Li
- Drug Safety Statistics, Drug Safety Research and Development, Pfizer, Inc., San Diego, California
| | - Nicole Streiner
- Oncology Research and Development, Pfizer, Inc., San Diego, California
| | - Jinwei Wang
- Oncology Research and Development, Pfizer, Inc., San Diego, California
| | - Dusko Trajkovic
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California
| | - Germaine Boucher
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California
| | - Timothy Coskran
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California
| | - Shawn P O'Neil
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California
| | - Sripad Ram
- Departments of Global Pathology and Investigative Toxicology, Pfizer, Inc., San Diego, California.
| |
Collapse
|
42
|
Richter M, Piwocka O, Musielak M, Piotrowski I, Suchorska WM, Trzeciak T. From Donor to the Lab: A Fascinating Journey of Primary Cell Lines. Front Cell Dev Biol 2021; 9:711381. [PMID: 34395440 PMCID: PMC8356673 DOI: 10.3389/fcell.2021.711381] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/21/2021] [Indexed: 12/02/2022] Open
Abstract
Primary cancer cell lines are ex vivo cell cultures originating from resected tissues during biopsies and surgeries. Primary cell cultures are objects of intense research due to their high impact on molecular biology and oncology advancement. Initially, the patient-derived specimen must be subjected to dissociation and isolation. Techniques for tumour dissociation are usually reliant on the organisation of connecting tissue. The most common methods include enzymatic digestion (with collagenase, dispase, and DNase), chemical treatment (with ethylene diamine tetraacetic acid and ethylene glycol tetraacetic acid), or mechanical disaggregation to obtain a uniform cell population. Cells isolated from the tissue specimen are cultured as a monolayer or three-dimensional culture, in the form of multicellular spheroids, scaffold-based cultures (i.e., organoids), or matrix-embedded cultures. Every primary cell line must be characterised to identify its origin, purity, and significant features. The process of characterisation should include different assays utilising specific (extra- and intracellular) markers. The most frequently used approaches comprise immunohistochemistry, immunocytochemistry, western blot, flow cytometry, real-time polymerase chain reaction, karyotyping, confocal microscopy, and next-generation sequencing. The growing body of evidence indicates the validity of the usage of primary cancer cell lines in the formulation of novel anti-cancer treatments and their contribution to drug development.
Collapse
Affiliation(s)
- Magdalena Richter
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, Poznań, Poland
| | - Oliwia Piwocka
- Radiobiology Lab, Department of Medical Physics, Greater Poland Cancer Center, Poznań, Poland
| | - Marika Musielak
- Department of Electroradiology, Poznan University of Medical Sciences, Poznań, Poland
| | - Igor Piotrowski
- Department of Electroradiology, Poznan University of Medical Sciences, Poznań, Poland
| | - Wiktoria M. Suchorska
- Radiobiology Lab, Department of Medical Physics, Greater Poland Cancer Center, Poznań, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznań, Poland
| | - Tomasz Trzeciak
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
43
|
Xu D, Li T, Wang R, Mu R. Expression and Pathogenic Analysis of Integrin Family Genes in Systemic Sclerosis. Front Med (Lausanne) 2021; 8:674523. [PMID: 34355002 PMCID: PMC8329247 DOI: 10.3389/fmed.2021.674523] [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: 03/01/2021] [Accepted: 06/17/2021] [Indexed: 12/05/2022] Open
Abstract
Objectives: Emerging evidence shows that integrin members are involved in inflammation and fibrosis in systemic sclerosis (SSc). This study aimed at evaluating the expression of integrin family genes in the skin tissue from SSc patients and exploring the potential pathogenic mechanism. Methods: We utilized the public datasets of SSc skin tissue from the Gene Expression Omnibus (GEO) database to analyze the expression and clinical significance of integrin family genes in SSc. The expression of integrin members in skin tissue was also assessed by immunohistochemistry. In addition, functional enrichment and pathway analysis were conducted. Results: Compared with healthy controls, the mRNA and protein levels of ITGA5, ITGB2, and ITGB5 were upregulated in the skin of SSc patients. Further analysis indicated that the mRNA expression levels of ITGA5, ITGB2, and ITGB5 were positively correlated with modified Rodnan skin thickness score (mRSS). Functional enrichment and pathway analysis showed that integrin members may play multiple roles in the pathogenesis of SSc. Among them, ITGA5, ITGB2, and ITGB5 might synergistically promote SSc through affecting extracellular matrix (ECM) turnover, ECM–receptor interaction, focal adhesion, and leukocyte trans-endothelial migration, while ITGA5 and ITGB5 also might affect angiogenesis and endothelial cell function. In addition, ITGA5, ITGB2, and ITGA5 were associated with different pathways, respectively. ITGA5 was uniquely enriched for actin organization, while ITGB5 was for TGF-β signaling and ITGB2 for immune cell activation. Conclusion: Our results implied that the abnormal expression of integrin family genes including ITGA5, ITGB2, and ITGB5 may participate in multiple pathological processes in SSc. Further investigations are required for confirming this speculation.
Collapse
Affiliation(s)
- Dan Xu
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, China
| | - Ting Li
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, China
| | - Ruikang Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Rong Mu
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
44
|
Immunohistochemical Analysis of Expression, Phosphorylation, and Nuclear Translocation of NF-κB Proteins in Human Tissues. Methods Mol Biol 2021. [PMID: 34236631 DOI: 10.1007/978-1-0716-1669-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Immunohistochemistry (IHC) is a technique aimed at detecting specific antigens on tissue sections by the use of targeting reagents labeled with reporter molecules. This technique allows a snapshot of the structure of tissue and determines the cellular and subcellular localization of a target antigen. This chapter describes how to identify and localize NF-κB proteins in human tissue using immunohistochemical staining on formalin-fixed paraffin-embedded and frozen tissue.
Collapse
|
45
|
Expression and prognostic value of CXCL12/CXCR4/CXCR7 axis in clear cell renal cell carcinoma. Clin Exp Nephrol 2021; 25:1057-1069. [PMID: 34109508 DOI: 10.1007/s10157-021-02081-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND CXCL12 or stromal-derived factor-1 is a chemokine that binds to two receptors CXCR4 and CXCR7 and takes part in both physiological and pathological cell functions. The disruption of the CXCL12/CXCR4/CXCR7 chemokine axis is seen in various types of cancers. METHODS We have immunohistochemically analyzed the expression of CXCL12 and its receptors in clear cell renal cell carcinoma patients. The study included 85 tissue samples. Since samples exhibited heterogeneity of expression intensity and staining localization (cytoplasmatic and membranous), histoscores were calculated, and their associations with clinicopathological parameters were analyzed. RESULTS Both cytoplasmatic CXCR7 and CXCL12 histoscores were associated with greater tumour size, while CXCL12 staining was associated with a higher grade as well. Mortality was associated with tumour size and both membranous and cytoplasmatic CXCL12 histoscores. With each centimetre in tumour size, survival decreases 1.3 times, while CXCL12C histoscore higher than 73 was associated with 2.3 greater risk of mortality. CXCR4 histoscore could only be predicted by female gender and neither cytoplasmatic nor membranous CXCR4 expression was found to be a mortality predictor. CONCLUSION Our data suggest that regarding overall survival, CXCL12 could be considered a valuable prognostic marker.
Collapse
|
46
|
Oungsakul P, Choi E, Shah AK, Mohamed A, O’Leary C, Duffy D, Hill MM, Bielefeldt-Ohmann H. Candidate Glycoprotein Biomarkers for Canine Visceral Hemangiosarcoma and Validation Using Semi-Quantitative Lectin/Immunohistochemical Assays. Vet Sci 2021; 8:vetsci8030038. [PMID: 33673507 PMCID: PMC7997418 DOI: 10.3390/vetsci8030038] [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: 01/29/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 11/16/2022] Open
Abstract
Visceral hemangiosarcoma (HSA) is one of the more frequent cancers in dogs and has a high metastatic rate and poor prognosis, as clinical signs only become apparent in advanced stages of tumor development. In order to improve early and differential diagnostic capabilities and hence, prognosis for dogs with HSA, two types of biomarker are needed: a point-of-care diagnostic biomarker and a prognostic biomarker—preferentially based on samples obtained with minimally invasive methods. In this study, we applied a lectin magnetic bead array-coupled tandem mass spectrometry (LeMBA-MS/MS) workflow through discovery and validation phases to discover serum glycoprotein biomarker candidates for canine HSA. By this approach, we found that Datura stramonium (DSA), wheat germ agglutinin (WGA), Sambucus nigra (SNA), and Pisum sativum (PSA) lectins captured the highest number of validated candidate glycoproteins. Secondly, we independently validated serum LeMBA-MS/MS results by demonstrating the in situ relationship of lectin-binding with tumor cells. Using lectin-histochemistry and immunohistochemistry (IHC) for key proteins on tissues with HSA and semi-quantitation of the signals, we demonstrate that a combination of DSA histochemistry and IHC for complement C7 greatly increases the prospect of a more specific diagnosis of canine HSA.
Collapse
Affiliation(s)
- Patharee Oungsakul
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia; (P.O.); (C.O.)
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; (E.C.); (A.K.S.); (D.D.); (M.M.H.)
| | - Eunju Choi
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; (E.C.); (A.K.S.); (D.D.); (M.M.H.)
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Alok K. Shah
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; (E.C.); (A.K.S.); (D.D.); (M.M.H.)
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
| | - Ahmed Mohamed
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
| | - Caroline O’Leary
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia; (P.O.); (C.O.)
| | - David Duffy
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; (E.C.); (A.K.S.); (D.D.); (M.M.H.)
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
| | - Michelle M. Hill
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; (E.C.); (A.K.S.); (D.D.); (M.M.H.)
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Helle Bielefeldt-Ohmann
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia; (P.O.); (C.O.)
- School of Chemistry & Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
- Correspondence:
| |
Collapse
|
47
|
Li Z, Goebel S, Reimann A, Ungerer M. Histo-ELISA technique for quantification and localization of tissue components. Sci Rep 2020; 10:19849. [PMID: 33199754 PMCID: PMC7669848 DOI: 10.1038/s41598-020-76950-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
A novel Histo-ELISA technique is intended to facilitate quantification of target tissue proteins in a tissue section and involves the selection of target regions in the tissue section, application of streptavidin-conjugated HRP (horseradish peroxidase), coupled with peroxidase substrate—TMB (3,3′,5,5′-tetramethylbenzidine), and staining dye evaluation with ELISA reader. The target protein content (weight per volume unit) was translated from optical densities by a reference standard curve, obtained via parallel staining of the targeted protein-coated slides. To validate the technique, we carried out quantifications of IgG extravasation in ischemic and nonischemic brain sections in a mouse stroke model. With those obtained data and the reference of immunohistochemistry scores assessed on the adjacent sections, accuracy, sensitivity, and precision for the technique were evaluated. For all evaluated parameters, Histo-ELISA performance was either comparable to or better than the standard immunohistochemistry. A comparison with the data from the repeated measurements yielded a rather low coefficient of variation. The results confirmed that the technique is a fairly reliable quantitative test with rather high sensitivity, accuracy, precision, and reproducibility for detecting target protein content in tissue sections and that its tissue distribution and related subsequent morphological changes can be observed at the same time.
Collapse
Affiliation(s)
- Zhongmin Li
- Advancecor GmbH, 82152, Martinsried, Germany.
| | | | | | | |
Collapse
|
48
|
The design and characterization of a gravitational microfluidic platform for drug sensitivity assay in colorectal perfused tumoroid cultures. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 30:102294. [DOI: 10.1016/j.nano.2020.102294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/24/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022]
|
49
|
Ko CJ, Harigopal M, Gehlhausen JR, Bosenberg M, McNiff JM, Damsky W. Discordant anti-SARS-CoV-2 spike protein and RNA staining in cutaneous perniotic lesions suggests endothelial deposition of cleaved spike protein. J Cutan Pathol 2020; 48:47-52. [PMID: 32895985 DOI: 10.1111/cup.13866] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/10/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Prior studies have shown the presence of immunohistochemical staining for the SARS-CoV-2 spike protein (SP) in endothelial cells and eccrine epithelium of acral perniosis classified as "COVID toes." Yet, other studies have been unable to detect SARS-CoV-2 RNA in skin biopsies of "COVID toes" by reverse-transcriptase polymerase chain reaction testing. OBJECTIVE In order to address these apparently conflicting findings, we compared detection of SARS-CoV-2 SP, through RNA in situ hybridization (ISH) vs immunohistochemistry (IHC), in skin biopsies of acral perniotic lesions presenting during the COVID-19 pandemic. RESULTS Three of six cases showed positive immunohistochemical labeling of endothelial cells, with one of three cases with sufficient depth also having labeling of eccrine glands, using an anti-SP SARS-CoV-2 antibody. These three cases positive with IHC were negative for SP by RNA ISH. CONCLUSION While the gold standard for detection of SARS-CoV-2 in tissue sections has yet to be determined, the detection of SARS-CoV-2 SP alone without spike RNA suggests that cleaved SP may be present in cutaneous endothelial cells and eccrine epithelium, providing a potential pathogenetic mechanism of COVID-19 endotheliitis.
Collapse
Affiliation(s)
- Christine J Ko
- Department of Dermatology, Yale Medical School, New Haven, Connecticut, USA.,Department of Pathology, Yale Medical School, New Haven, Connecticut, USA
| | - Malini Harigopal
- Department of Pathology, Yale Medical School, New Haven, Connecticut, USA
| | - Jeff R Gehlhausen
- Department of Dermatology, Yale Medical School, New Haven, Connecticut, USA
| | - Marcus Bosenberg
- Department of Dermatology, Yale Medical School, New Haven, Connecticut, USA.,Department of Pathology, Yale Medical School, New Haven, Connecticut, USA
| | - Jennifer M McNiff
- Department of Dermatology, Yale Medical School, New Haven, Connecticut, USA.,Department of Pathology, Yale Medical School, New Haven, Connecticut, USA
| | - William Damsky
- Department of Dermatology, Yale Medical School, New Haven, Connecticut, USA.,Department of Pathology, Yale Medical School, New Haven, Connecticut, USA
| |
Collapse
|
50
|
Kuznietsova H, Dziubenko N, Herheliuk T, Prylutskyy Y, Tauscher E, Ritter U, Scharff P. Water-Soluble Pristine C 60 Fullerene Inhibits Liver Alterations Associated with Hepatocellular Carcinoma in Rat. Pharmaceutics 2020; 12:pharmaceutics12090794. [PMID: 32842595 PMCID: PMC7559840 DOI: 10.3390/pharmaceutics12090794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022] Open
Abstract
Excessive production of reactive oxygen species is the main cause of hepatocellular carcinoma (HCC) initiation and progression. Water-soluble pristine C60 fullerene is a powerful and non-toxic antioxidant, therefore, its effect under rat HCC model and its possible mechanisms were aimed to be discovered. Studies on HepG2 cells (human HCC) demonstrated C60 fullerene ability to inhibit cell growth (IC50 = 108.2 μmol), to induce apoptosis, to downregulate glucose-6-phosphate dehydrogenase, to upregulate vimentin and p53 expression and to alter HepG2 redox state. If applied to animals experienced HCC in dose of 0.25 mg/kg per day starting at liver cirrhosis stage, C60 fullerene improved post-treatment survival similar to reference 5-fluorouracil (31 and 30 compared to 17 weeks) and inhibited metastasis unlike the latter. Furthermore, C60 fullerene substantially attenuated liver injury and fibrosis, decreased liver enzymes, and normalized bilirubin and redox markers (elevated by 1.7–7.7 times under HCC). Thus, C60 fullerene ability to inhibit HepG2 cell growth and HCC development and metastasis and to improve animal survival was concluded. C60 fullerene cytostatic action might be realized through apoptosis induction and glucose-6-phosphate dehydrogenase downregulation in addition to its antioxidant activity.
Collapse
Affiliation(s)
- Halyna Kuznietsova
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
- Correspondence: (H.K.); (U.R.); Tel.: +38-095-277-4370 (H.K.); +49-3677-69-3603 (U.R.)
| | - Natalia Dziubenko
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
| | - Tetiana Herheliuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
| | - Yuriy Prylutskyy
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine; (N.D.); (T.H.); (Y.P.)
| | - Eric Tauscher
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer str. 25, 98693 Ilmenau, Germany; (E.T.); (P.S.)
| | - Uwe Ritter
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer str. 25, 98693 Ilmenau, Germany; (E.T.); (P.S.)
- Correspondence: (H.K.); (U.R.); Tel.: +38-095-277-4370 (H.K.); +49-3677-69-3603 (U.R.)
| | - Peter Scharff
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer str. 25, 98693 Ilmenau, Germany; (E.T.); (P.S.)
| |
Collapse
|