1
|
Bountas D, Bountas M, Exadactylou P, Tziafalia C, Dimitriadis C, Doumas A. Erdheim-Chester disease and nuclear medicine imaging. A case report and brief review. Skeletal Radiol 2025; 54:359-365. [PMID: 38842690 DOI: 10.1007/s00256-024-04718-z] [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: 10/31/2023] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Erdheim-Chester disease (ECD) is a rare clonal myeloid neoplasm typically affecting adults over 50 years old, with bone lesions in almost all patients. The prognosis is poor in most cases if left untreated. Clinical manifestations are not specific, which hinders early diagnosis. The disease has distinct radiological features. However, three-phase bone scintigraphy exhibits the most typical pattern of all imaging modalities, which is the prominent strikingly symmetrical radiotracer uptake in the distal ends of the femurs and proximal and distal ends of the tibiae, sparing the epiphyses. We report a case of a 54-year-old female patient, presenting with atypical persistent knee joint pain. After an MRI scan, she underwent a three-phase bone scan, revealing the characteristic pattern, thus indicating a possible ECD diagnosis, which was eventually confirmed in biopsy material. Novel aspects of the pathophysiology and treatment of the disease, as well as a differential diagnosis from the perspective of an MSK radiologist and nuclear medicine physician, are also discussed.
Collapse
Affiliation(s)
- D Bountas
- Ippokratis Nuclear Medicine Center, Thessaloniki, Greece.
- Medical Multidiagnostic Center, Euromedica Karditsa, Karditsa, Greece.
| | - M Bountas
- Aristotle University of Thessaloniki Medical School, Thessaloniki, Greece
| | - P Exadactylou
- Dpt of Nuclear Medicine, Aristotle University of Thessaloniki Medical School, AHEPA University Hospital, Thessaloniki, Greece
| | - C Tziafalia
- Medical Multidiagnostic Center, Euromedica Karditsa, Karditsa, Greece
| | - C Dimitriadis
- Internal Medicine Private Practice, Karditsa, Greece
| | - A Doumas
- Ippokratis Nuclear Medicine Center, Thessaloniki, Greece
- Dpt of Nuclear Medicine, Aristotle University of Thessaloniki Medical School, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
2
|
He X, Deng H, Liu W, Hu L, Tan X. Advances in Understanding Drug Resistance Mechanisms and Innovative Clinical Treatments for Melanoma. Curr Treat Options Oncol 2024; 25:1615-1633. [PMID: 39633237 DOI: 10.1007/s11864-024-01279-0] [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] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
OPINION STATEMENT Melanoma, a highly invasive skin cancer resulting from melanocyte malignant transformation, is the third most common skin malignancy. Despite accounting for only 4% to 5% of all skin malignancies, it is responsible for 80% of skin cancer-related deaths. Targeted therapies and immune checkpoint inhibitors have improved survival rates, yet drug resistance remains a major challenge. In this review, I explore the latest research progress on melanoma drug resistance mechanisms and clinical treatment methods. This aims to provide insights for more effective treatment strategies and improve patient prognosis and quality of life. I also discuss potential strategies to overcome drug resistance based on the latest scientific findings, with a particular focus on the complex and multi-factorial drug resistance mechanisms of melanomas, including genetic mutations, epigenetic changes, and tumor microenvironment factors. Understanding these mechanisms is crucial for developing new drugs and combination therapies targeting drug-resistant tumors. Analyzing complex drug resistance pathways paves the way for personalized medical approaches, which is expected to provide enlightenment on breaking through drug resistance barriers and enhancing the effectiveness of melanoma treatment.
Collapse
Affiliation(s)
- Xiaoya He
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Hao Deng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Wei Liu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, China
| | - Liling Hu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, China.
| | - Xiao Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, China.
| |
Collapse
|
3
|
Kennedy VE, Smith CC. FLT3 targeting in the modern era: from clonal selection to combination therapies. Int J Hematol 2024; 120:528-540. [PMID: 38112995 PMCID: PMC11513752 DOI: 10.1007/s12185-023-03681-0] [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: 06/21/2023] [Revised: 10/14/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023]
Abstract
Fms-like tyrosine kinase 3 (FLT3) is the most frequently mutated gene in acute myeloid leukemia (AML). Modern targeting of FLT3 with inhibitors has improved clinical outcomes and FLT3 inhibitors have been incorporated into the treatment of AML in all phases of the disease, including the upfront, relapsed/refractory and maintenance settings. This review will discuss the current understanding of FLT3 biology, the clinical use of FLT3 inhibitors, resistance mechanisms and emerging combination treatment strategies.
Collapse
Affiliation(s)
- Vanessa E Kennedy
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, 505 Parnassus Ave, Box 1270, San Francisco, CA, 94143, USA
| | - Catherine C Smith
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, 505 Parnassus Ave, Box 1270, San Francisco, CA, 94143, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Naghib SM, Ahmadi B, Mikaeeli Kangarshahi B, Mozafari MR. Chitosan-based smart stimuli-responsive nanoparticles for gene delivery and gene therapy: Recent progresses on cancer therapy. Int J Biol Macromol 2024; 278:134542. [PMID: 39137858 DOI: 10.1016/j.ijbiomac.2024.134542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
Recent cancer therapy research has found that chitosan (Ch)-based nanoparticles show great potential for targeted gene delivery. Chitosan, a biocompatible and biodegradable polymer, has exceptional properties, making it an ideal carrier for therapeutic genes. These nanoparticles can respond to specific stimuli like pH, temperature, and enzymes, enabling precise delivery and regulated release of genes. In cancer therapy, these nanoparticles have proven effective in delivering genes to tumor cells, slowing tumor growth. Adjusting the nanoparticle's surface, encapsulating protective agents, and using targeting ligands have also improved gene delivery efficiency. Smart nanoparticles based on chitosan have shown promise in improving outcomes by selectively releasing genes in response to tumor conditions, enhancing targeted delivery, and reducing off-target effects. Additionally, targeting ligands on the nanoparticles' surface increases uptake and effectiveness. Although further investigation is needed to optimize the structure and composition of these nanoparticles and assess their long-term safety, these advancements pave the way for innovative gene-focused cancer therapies.
Collapse
Affiliation(s)
- Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Bahar Ahmadi
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Babak Mikaeeli Kangarshahi
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
5
|
Kamal R, Awasthi A, Paul P, Mir MS, Singh SK, Dua K. Novel drug delivery systems in colorectal cancer: Advances and future prospects. Pathol Res Pract 2024; 262:155546. [PMID: 39191194 DOI: 10.1016/j.prp.2024.155546] [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: 02/20/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Colorectal cancer (CRC) is an abnormal proliferation of cells within the colon and rectum, leading to the formation of polyps and disruption of mucosal functions. The disease development is influenced by a combination of factors, including inflammation, exposure to environmental mutagens, genetic alterations, and impairment in signaling pathways. Traditional treatments such as surgery, radiation, and chemotherapy are often used but have limitations, including poor solubility and permeability, treatment resistance, side effects, and post-surgery issues. Novel Drug Delivery Systems (NDDS) have emerged as a superior alternative, offering enhanced drug solubility, precision in targeting cancer cells, and regulated drug release. Thereby addressing the shortcomings of conventional therapies and showing promise for more effective CRC management. The present review sheds light on the pathogenesis, signaling pathways, biomarkers, conventional treatments, need for NDDS, and application of NDDS against CRC. Additionally, clinical trials, ongoing clinical trials, marketed formulations, and patents on CRC are also covered in the present review.
Collapse
Affiliation(s)
- Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab 142001, India; School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab 147301, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab 142001, India; Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Priyanka Paul
- Department of Pharmaceutical Science, PCTE Group of Institute, Ludhiana, Punjab, India
| | - Mohammad Shabab Mir
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab 147301, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
6
|
Kugler V, Schwaighofer S, Feichtner A, Enzler F, Fleischmann J, Strich S, Schwarz S, Wilson R, Tschaikner P, Troppmair J, Sexl V, Meier P, Kaserer T, Stefan E. Impact of protein and small molecule interactions on kinase conformations. eLife 2024; 13:RP94755. [PMID: 39088265 PMCID: PMC11293870 DOI: 10.7554/elife.94755] [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] [Indexed: 08/02/2024] Open
Abstract
Protein kinases act as central molecular switches in the control of cellular functions. Alterations in the regulation and function of protein kinases may provoke diseases including cancer. In this study we investigate the conformational states of such disease-associated kinases using the high sensitivity of the kinase conformation (KinCon) reporter system. We first track BRAF kinase activity conformational changes upon melanoma drug binding. Second, we also use the KinCon reporter technology to examine the impact of regulatory protein interactions on LKB1 kinase tumor suppressor functions. Third, we explore the conformational dynamics of RIP kinases in response to TNF pathway activation and small molecule interactions. Finally, we show that CDK4/6 interactions with regulatory proteins alter conformations which remain unaffected in the presence of clinically applied inhibitors. Apart from its predictive value, the KinCon technology helps to identify cellular factors that impact drug efficacies. The understanding of the structural dynamics of full-length protein kinases when interacting with small molecule inhibitors or regulatory proteins is crucial for designing more effective therapeutic strategies.
Collapse
Affiliation(s)
- Valentina Kugler
- Institute for Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
| | - Selina Schwaighofer
- Institute for Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
| | - Andreas Feichtner
- Institute for Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
| | - Florian Enzler
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of InnsbruckInnsbruckAustria
| | - Jakob Fleischmann
- Institute for Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
| | - Sophie Strich
- Institute for Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
| | - Sarah Schwarz
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
| | - Rebecca Wilson
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer ResearchLondonUnited Kingdom
| | - Philipp Tschaikner
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
- KinCon biolabs GmbHInnsbruckAustria
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of InnsbruckInnsbruckAustria
| | | | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer ResearchLondonUnited Kingdom
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of InnsbruckInnsbruckAustria
| | - Eduard Stefan
- Institute for Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research Institute (TKFI)InnsbruckAustria
- KinCon biolabs GmbHInnsbruckAustria
| |
Collapse
|
7
|
Dragomir M, Călugăru OT, Popescu B, Jardan C, Jardan D, Popescu M, Aposteanu S, Bădeliță S, Nedelcu G, Șerban C, Popa C, Vassu-Dimov T, Coriu D. DNA Sequencing of CD138 Cell Population Reveals TP53 and RAS-MAPK Mutations in Multiple Myeloma at Diagnosis. Cancers (Basel) 2024; 16:358. [PMID: 38254847 PMCID: PMC10813921 DOI: 10.3390/cancers16020358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Multiple myeloma is a hematologic neoplasm caused by abnormal proliferation of plasma cells. Sequencing studies suggest that plasma cell disorders are caused by both cytogenetic abnormalities and oncogene mutations. Therefore, it is necessary to detect molecular abnormalities to improve the diagnosis and management of MM. The main purpose of this study is to determine whether NGS, in addition to cytogenetics, can influence risk stratification and management. Additionally, we aim to establish whether mutational analysis of the CD138 cell population is a suitable option for the characterization of MM compared to the bulk population. Following the separation of the plasma cells harvested from 35 patients newly diagnosed with MM, we performed a FISH analysis to detect the most common chromosomal abnormalities. Consecutively, we used NGS to evaluate NRAS, KRAS, BRAF, and TP53 mutations in plasma cell populations and in bone marrow samples. NGS data showed that sequencing CD138 cells provides a more sensitive approach. We identified several variants in BRAF, KRAS, and TP53 that were not previously associated with MM. Considering that the presence of somatic mutations could influence risk stratification and therapeutic approaches of patients with MM, sensitive detection of these mutations at diagnosis is essential for optimal management of MM.
Collapse
Affiliation(s)
- Mihaela Dragomir
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (M.D.); (T.V.-D.)
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
| | - Onda-Tabita Călugăru
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
| | - Bogdan Popescu
- Hematology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Cerasela Jardan
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
- Hematology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Dumitru Jardan
- Molecular Biology Laboratory, Medlife Bucharest, 010093 Bucharest, Romania;
| | - Monica Popescu
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
| | - Silvia Aposteanu
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
| | - Sorina Bădeliță
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
| | - Gabriela Nedelcu
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
| | - Cătălin Șerban
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
| | - Codruța Popa
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
- Hematology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Tatiana Vassu-Dimov
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (M.D.); (T.V.-D.)
| | - Daniel Coriu
- Fundeni Clinical Institute, 022328 Bucharest, Romania; (C.J.); (M.P.); (S.A.); (S.B.); (G.N.); (C.Ș.); (C.P.); (D.C.)
- Hematology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
8
|
Baygin RC, Yilmaz KC, Acar A. Characterization of dabrafenib-induced drug insensitivity via cellular barcoding and collateral sensitivity to second-line therapeutics. Sci Rep 2024; 14:286. [PMID: 38167959 PMCID: PMC10762103 DOI: 10.1038/s41598-023-50443-3] [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: 05/14/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Drug insensitivity is arguably one of the biggest challenges in cancer therapeutics. Although effective therapeutic solutions in cancer are limited due to the emergence of drug insensitivity, exploiting evolutionary understanding in this context can provide potential second-line therapeutics sensitizing the drug insensitive populations. Targeted therapeutic agent dabrafenib is used to treat CRC patients with BRAF V600E genotype and insensitivity to dabrafenib is often observed. Understanding underlying clonal architecture of dabrafenib-induced drug insensitivity and identification of potential second-line therapeutics that could sensitize dabrafenib insensitive populations remain to be elucidated. For this purpose, we utilized cellular barcoding technology to decipher dabrafenib-induced clonal evolution in BRAF V600E mutant HT-29 cells. This approach revealed the detection of both pre-existing and de novo barcodes with increased frequencies as a result of dabrafenib insensitivity. Furthermore, our longitudinal monitoring of drug insensitivity based on barcode detection from floating DNA within used medium enabled to identify temporal dynamics of pre-existing and de novo barcodes in relation to dabrafenib insensitivity in HT-29 cells. Moreover, whole-exome sequencing analysis exhibited possible somatic CNVs and SNVs contributing to dabrafenib insensitivity in HT-29 cells. Last, collateral drug sensitivity testing demonstrated oxaliplatin and capecitabine, alone or in combination, as successful second-like therapeutics in inducing collateral sensitivity in dabrafenib-insensitive HT-29 cells. Overall, our findings demonstrate clonal dynamics of dabrafenib-insensitivity in HT-29 cells. In addition, oxaliplatin and capecitabine, alone or in combination, were successful second-line therapeutics in inducing collateral sensitivity in dabrafenib-insensitive HT-29 cells.
Collapse
Affiliation(s)
- Rana Can Baygin
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, Çankaya, 06800, Ankara, Turkey
| | - Kubra Celikbas Yilmaz
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, Çankaya, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, Çankaya, 06800, Ankara, Turkey.
| |
Collapse
|
9
|
Blay JY, Cropet C, Mansard S, Loriot Y, De La Fouchardière C, Haroche J, Topart D, Tougeron D, You B, Italiano A, Le Brun-Ly V, Ferrero JM, Penel N, Fabbro M, Troussard X, Malka D, Ray-Coquard I, Leboulleux S, Fléchon A, Maubec E, Charles J, Dalle S, Taieb S, Garcia GCTE, Mandache AM, Colignon N, Gavrel M, Nowak F, Hoog Labouret N, Mahier Aït Oukhatar C, Gomez-Roca C. Long term activity of vemurafenib in cancers with BRAF mutations: the ACSE basket study for advanced cancers other than BRAF V600-mutated melanoma. ESMO Open 2023; 8:102038. [PMID: 37922690 PMCID: PMC10774964 DOI: 10.1016/j.esmoop.2023.102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND BRAF inhibitors are approved in BRAFV600-mutated metastatic melanoma, non-small-cell lung cancer (NSCLC), Erdheim-Chester disease (ECD), and thyroid cancer. We report here the efficacy, safety, and long-term results of single-agent vemurafenib given in the AcSé vemurafenib basket study to patients with various BRAF-mutated advanced tumours other than BRAFV600-mutated melanoma and NSCLC. PATIENTS AND METHODS Patients with advanced tumours other than BRAFV600E melanoma and progressing after standard treatment were eligible for inclusion in nine cohorts (including a miscellaneous cohort) and received oral vemurafenib 960 mg two times daily. The primary endpoint was the objective response rate (ORR) estimated with a Bayesian design. The secondary outcomes were disease control rate, duration of response, progression-free survival (PFS), overall survival (OS), and vemurafenib safety. RESULTS A total of 98 advanced patients with various solid or haematological cancers, 88 with BRAFV600 mutations and 10 with BRAFnonV600 mutations, were included. The median follow-up duration was 47.7 months. The Bayesian estimate of ORR was 89.7% in hairy cell leukaemias (HCLs), 33.3% in the glioblastomas cohort, 18.2% in cholangiocarcinomas, 80.0% in ECD, 50.0% in ovarian cancers, 50.0% in xanthoastrocytomas, 66.7% in gangliogliomas, and 60.0% in sarcomas. The median PFS of the whole series was 8.8 months. The 12-, 24-, and 36-month PFS rates were 42.2%, 23.8%, and 17.9%, respectively. Overall, 54 patients died with a median OS of 25.9 months, with a projected 4-year OS of 40%. Adverse events were similar to those previously reported with vemurafenib. CONCLUSION Responses and prolonged PFS were observed in many tumours with BRAF mutations, including HCL, ECD, ovarian carcinoma, gliomas, ganglioglioma, and sarcomas. Although not all cancer types responded, vemurafenib is an agnostic oncogene therapy of cancers.
Collapse
Affiliation(s)
- J Y Blay
- Department of Medicine, CentreLeon bErard, Lyon.
| | | | - S Mansard
- Dermatology Department, Hôpital Estaing, University Hospital of Clermont Ferrand, Clermont-Ferrand
| | - Y Loriot
- Department of Medicine, Gustave Roussy, Villejuif
| | | | - J Haroche
- Department of Internal Medicine, Institut E3M, French Reference Centre for Histiocytosis, Pitié-Salpȇtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris
| | - D Topart
- Onco-urology Department, Hôpital Saint ELOI, Montpellier
| | - D Tougeron
- Gastroenterology and Hepatology Department, Poitiers University Hospital and Faculty of Medicine of Poitiers, Poitiers
| | - B You
- Centre d'Investigation des Thérapeutiques en Oncologie et Hématologie de Lyon (CITOHL), Hospices Civils de Lyon (IC-HCL), EA 3738 CICLY, Lyon
| | - A Italiano
- Department of Medicine, Institut Bergonié, Bordeaux; Faculty of Medicine, University of Bordeaux, Bordeaux
| | - V Le Brun-Ly
- Department of Medicine, CHU Limoges, Medical Oncology, Limoges
| | - J M Ferrero
- Department of Medicine, Centre A. Lacassagne, Nice
| | - N Penel
- Department of Medical Oncology, Centre Oscar Lambret, Lille; Université de Lille, CHU Lille, ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales, Lille
| | - M Fabbro
- Department of Medicine, Institut de Cancerologie de Montpellier, Montpellier
| | | | - D Malka
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris
| | | | - S Leboulleux
- Department of Medicine, Gustave Roussy, Villejuif
| | | | - E Maubec
- Assistance Publique-Hôpitaux de Paris, Department of Dermatology, Hôpital Avicenne, Bobigny; University Sorbonne Paris Nord - Campus de Bobigny, Bobigny and UMR 1124, Campus Saint-Germain-des-Prés, Paris
| | - J Charles
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Grenoble; Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, La Tronche
| | - S Dalle
- Department of Dermatology, Hospices Civils de Lyon, CRCL, Université Claude Bernard Lyon1, Lyon
| | - S Taieb
- Department of Medical Oncology, Centre Oscar Lambret, Lille
| | | | | | - N Colignon
- Department of Radiology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris
| | - M Gavrel
- Department of Medicine, Gustave Roussy, Villejuif
| | - F Nowak
- Institut National Du Cancer, Boulogne-Billancourt
| | | | | | - C Gomez-Roca
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse (IUCT-Oncopole), Clinical Research Unit, Toulouse, France
| |
Collapse
|
10
|
Laschinsky C, Theurer S, Herold T, Rawitzer J, Weber F, Herrmann K, Brandenburg T, Führer-Sakel D, Fendler WP, Weber M. Molecular Markers Are Associated with Onset of Radioiodine Refractoriness in Patients with Papillary Thyroid Carcinoma. J Nucl Med 2023; 64:1865-1868. [PMID: 37884333 DOI: 10.2967/jnumed.123.266044] [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: 05/22/2023] [Revised: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The onset of radioiodine-refractory thyroid carcinoma (RR-TC) is a negative predictor of survival and has been linked to the presence of BRAFV600E mutations in papillary thyroid cancer. We aimed to identify further genetic alterations associated with RR-TC. Methods: We included 38 patients with papillary thyroid cancer who underwent radioiodine imaging and 18F-FDG PET/CT after total thyroidectomy. The molecular profile was assessed by next-generation sequencing. The time to the onset of RR-TC for different genetic alterations was compared using the log-rank test. Results: The median onset to RR-TC was 0.7 and 19.8 mo in patients with and without, respectively, telomerase reverse transcriptase promoter mutations (P = 0.02) and 1.7 and 19.8 mo in patients with and without, respectively, a tumor protein 53 mutation (P < 0.01). This association was not observed for BRAFV600E mutations (P = 0.49). Conclusion: Our data show a significant association between the onset of RR-TC and mutations in telomerase reverse transcriptase promoter and tumor protein 53, indicating the need for a more extensive diagnostic workup in these patients. Certain genetic changes put patients with thyroid cancer at risk of developing cancer spread that does not respond to radioiodine therapy.
Collapse
Affiliation(s)
- Christina Laschinsky
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany;
- German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Sarah Theurer
- German Cancer Consortium, Partner Site Essen, Essen, Germany
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Thomas Herold
- German Cancer Consortium, Partner Site Essen, Essen, Germany
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Josefine Rawitzer
- German Cancer Consortium, Partner Site Essen, Essen, Germany
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Frank Weber
- German Cancer Consortium, Partner Site Essen, Essen, Germany
- Department of Surgery, Section of Endocrine Surgery, Essen University Hospital, Essen, Germany; and
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Tim Brandenburg
- German Cancer Consortium, Partner Site Essen, Essen, Germany
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dagmar Führer-Sakel
- German Cancer Consortium, Partner Site Essen, Essen, Germany
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Manuel Weber
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium, Partner Site Essen, Essen, Germany
| |
Collapse
|
11
|
Katkat E, Demirci Y, Heger G, Karagulle D, Papatheodorou I, Brazma A, Ozhan G. Canonical Wnt and TGF-β/BMP signaling enhance melanocyte regeneration but suppress invasiveness, migration, and proliferation of melanoma cells. Front Cell Dev Biol 2023; 11:1297910. [PMID: 38020918 PMCID: PMC10679360 DOI: 10.3389/fcell.2023.1297910] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Melanoma is the deadliest form of skin cancer and develops from the melanocytes that are responsible for the pigmentation of the skin. The skin is also a highly regenerative organ, harboring a pool of undifferentiated melanocyte stem cells that proliferate and differentiate into mature melanocytes during regenerative processes in the adult. Melanoma and melanocyte regeneration share remarkable cellular features, including activation of cell proliferation and migration. Yet, melanoma considerably differs from the regenerating melanocytes with respect to abnormal proliferation, invasive growth, and metastasis. Thus, it is likely that at the cellular level, melanoma resembles early stages of melanocyte regeneration with increased proliferation but separates from the later melanocyte regeneration stages due to reduced proliferation and enhanced differentiation. Here, by exploiting the zebrafish melanocytes that can efficiently regenerate and be induced to undergo malignant melanoma, we unravel the transcriptome profiles of the regenerating melanocytes during early and late regeneration and the melanocytic nevi and malignant melanoma. Our global comparison of the gene expression profiles of melanocyte regeneration and nevi/melanoma uncovers the opposite regulation of a substantial number of genes related to Wnt signaling and transforming growth factor beta (TGF-β)/(bone morphogenetic protein) BMP signaling pathways between regeneration and cancer. Functional activation of canonical Wnt or TGF-β/BMP pathways during melanocyte regeneration promoted melanocyte regeneration but potently suppressed the invasiveness, migration, and proliferation of human melanoma cells in vitro and in vivo. Therefore, the opposite regulation of signaling mechanisms between melanocyte regeneration and melanoma can be exploited to stop tumor growth and develop new anti-cancer therapies.
Collapse
Affiliation(s)
- Esra Katkat
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Türkiye
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Izmir, Türkiye
| | - Yeliz Demirci
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Türkiye
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Izmir, Türkiye
| | | | - Doga Karagulle
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Türkiye
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Türkiye
| | - Irene Papatheodorou
- European Molecular Biology Laboratory—European Bioinformatics Institute (EMBL-EBI), Cambridge, United Kingdom
| | - Alvis Brazma
- European Molecular Biology Laboratory—European Bioinformatics Institute (EMBL-EBI), Cambridge, United Kingdom
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Türkiye
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Türkiye
| |
Collapse
|
12
|
Janke LJ, Rehg JE. The many faces of mouse histiocytic sarcoma in C57BL/6J mice. Vet Pathol 2023; 60:443-460. [PMID: 37132518 DOI: 10.1177/03009858231166658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Histiocytic sarcoma is a tumor of the hematopoietic system considered to be derived from macrophages. Although rare in humans, it occurs frequently in mice. Histiocytic sarcoma can be a difficult tumor to diagnose due to its diverse cellular morphologies, growth patterns, and organ distributions. The varying morphology of histiocytic sarcomas makes it easy to confuse them with other types of neoplasia, including hepatic hemangiosarcoma, uterine schwannoma, leiomyosarcoma, uterine stromal cell tumor, intramedullary osteosarcoma, and myeloid leukemia. As such, immunohistochemistry (IHC) is often needed to differentiate histiocytic sarcomas from other common tumors in mice that they can morphologically mimic. The goal of this article is to present a broader perspective of the diverse cellular morphologies, growth patterns, organ distributions, and IHC labeling of histiocytic sarcomas encountered by the authors. This article describes these features in a set of 62 mouse histiocytic sarcomas, including the IHC characterization of the tumors using a panel of markers for the macrophage antigens F4/80, IBA1, MAC2, CD163, CD68, and lysozyme, and describes differentiating features of histiocytic sarcomas from other morphologically similar tumors. The genetic changes underlying the pathogenesis of histiocytic sarcoma in humans are beginning to be elucidated, but this is difficult due to its rarity. The higher prevalence of this tumor in mice provides opportunities to investigate mechanisms of its development and to test potential treatments.
Collapse
|
13
|
Kollara A, Burt BD, Ringuette MJ, Brown TJ. The adaptor protein VEPH1 interacts with the kinase domain of ERBB2 and impacts EGF signaling in ovarian cancer cells. Cell Signal 2023; 106:110634. [PMID: 36828346 DOI: 10.1016/j.cellsig.2023.110634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023]
Abstract
Upregulation of ERBB2 and activating mutations in downstream KRAS/BRAF and PIK3CA are found in several ovarian cancer histotypes. ERBB2 enhances signaling by the ERBB family of EGF receptors, and contains docking positions for proteins that transduce signaling through multiple pathways. We identified the adaptor protein ventricular zone-expressed pleckstrin homology domain-containing protein 1 (VEPH1) as a potential interacting partner of ERBB2 in a screen of proteins co-immunoprecipitated with VEPH1. In this study, we confirm a VEPH1 - ERBB2 interaction by co-immunoprecipitation and biotin proximity labelling and show that VEPH1 interacts with the juxtamembrane-kinase domain of ERBB2. In SKOV3 ovarian cancer cells, which bear a PIK3CA mutation and ERBB2 overexpression, ectopic VEPH1 expression enhanced EGF activation of ERK1/2, and mTORC2 activation of AKT. In contrast, in ES2 ovarian cancer cells, which bear a BRAFV600E mutation with VEPH1 amplification but low ERBB2 expression, loss of VEPH1 expression enabled further activation of ERK1/2 by EGF and enhanced EGF activation of AKT. VEPH1 expression in SKOV3 cells enhanced EGF-induced cell migration consistent with increased Snail2 and decreased E-cadherin levels. In comparison, loss of VEPH1 expression in ES2 cells led to decreased cell motility independent of EGF treatment despite higher levels of N-cadherin and Snail2. Importantly, we found that loss of VEPH1 expression rendered ES2 cells less sensitive to BRAF and MEK inhibition. This study extends the range of adaptor function of VEPH1 to ERBB2, and indicates VEPH1 has differential effects on EGF signaling in ovarian cancer cells that may be influenced by driver gene mutations.
Collapse
Affiliation(s)
- Alexandra Kollara
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Brian D Burt
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Maurice J Ringuette
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Theodore J Brown
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
14
|
Microbiota, Oxidative Stress, and Skin Cancer: An Unexpected Triangle. Antioxidants (Basel) 2023; 12:antiox12030546. [PMID: 36978794 PMCID: PMC10045429 DOI: 10.3390/antiox12030546] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Mounting evidence indicates that the microbiota, the unique combination of micro-organisms residing in a specific environment, plays an essential role in the development of a wide range of human diseases, including skin cancer. Moreover, a persistent imbalance of microbial community, named dysbiosis, can also be associated with oxidative stress, a well-known emerging force involved in the pathogenesis of several human diseases, including cutaneous malignancies. Although their interplay has been somewhat suggested, the connection between microbiota, oxidative stress, and skin cancer is a largely unexplored field. In the present review, we discuss the current knowledge on these topics, suggesting potential therapeutic strategies.
Collapse
|
15
|
Wang Q, Ren H, Zheng L, Wang J, Zhong D. Recurrent central nervous system Rosai-Dorfman disease with KRAS mutation: a case report. Diagn Pathol 2023; 18:21. [PMID: 36782249 PMCID: PMC9926849 DOI: 10.1186/s13000-022-01276-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 12/19/2022] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Rosai-Dorfman disease (RDD) is a rare, non-Langerhans cell histiocytosis of unknown etiology. we report a very rare case of recurrent central nervous system RDD with KRAS gene mutation and review the literature to improve our understanding of this disease. CASE PRESENTATION A 19-year-old male patient was admitted to our hospital for headache. Cranial magnetic resonance imaging revealed a mass of abnormal signal shadows in the prepontine cistern. The mass was surgically removed and the patient was consequently diagnosed with intracranial Rosai-Dorfman disease. Seven months later, pathological examination confirmed that the RDD had recurred. Next-generation sequencing found KRAS mutation in exon 4 (C.351A > C. P. K117n). CONCLUSION RDD of the CNS has no distinct clinical manifestations and imaging characteristics, and the final diagnosis should be based on the results of the pathological examination. Although RDD is not currently classified as a neoplastic disorder, some evidence of clonality has changed our understanding of it. Follow up examinations over a long period are necessary to determine the efficacy of treatment.
Collapse
Affiliation(s)
- Qingyang Wang
- grid.506261.60000 0001 0706 7839Graduate School of Peking Union Medical College, Beijing, 100006 China ,grid.415954.80000 0004 1771 3349Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Hongxiang Ren
- grid.415954.80000 0004 1771 3349Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Liyuan Zheng
- grid.415954.80000 0004 1771 3349Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Juan Wang
- grid.506261.60000 0001 0706 7839Graduate School of Peking Union Medical College, Beijing, 100006 China ,grid.415954.80000 0004 1771 3349Department of Radiology, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Dingrong Zhong
- Graduate School of Peking Union Medical College, Beijing, 100006, China. .,Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
16
|
Dey A, Mitra A, Pathak S, Prasad S, Zhang AS, Zhang H, Sun XF, Banerjee A. Recent Advancements, Limitations, and Future Perspectives of the use of Personalized Medicine in Treatment of Colon Cancer. Technol Cancer Res Treat 2023; 22:15330338231178403. [PMID: 37248615 PMCID: PMC10240881 DOI: 10.1177/15330338231178403] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/18/2023] [Accepted: 03/13/2023] [Indexed: 08/29/2024] Open
Abstract
Due to the heterogeneity of colon cancer, surgery, chemotherapy, and radiation are ineffective in all cases. The genomic profile and biomarkers associated with the process are considered in personalized medicine, along with the patient's personal history. It is based on the response of the targeted therapies to specific genetic variations. The patient's genetic transcriptomic and epigenetic features are evaluated, and the best therapeutic approach and diagnostic testing are identified through personalized medicine. This review aims to summarize all the necessary, updated information on colon cancer related to personalized medicine. Personalized medicine is gaining prominence as generalized treatments are finding it challenging to contain colon cancer cases which currently rank fourth among global cancer incidence while being the fifth largest in total death cases worldwide. In personalized therapy, patients are grouped into specific categories, and the best therapeutic approach is chosen based on evaluating their molecular features. Various personalized strategies are currently being explored in the treatment of colon cancer involving immunotherapy, phytochemicals, and other biomarker-specific targeted therapies. However, significant challenges must be overcome to integrate personalized medicine into healthcare systems completely. We look at the various signaling pathways and genetic and epigenetic alterations associated with colon cancer to understand and identify biomarkers useful in targeted therapy. The current personalized therapies available in colon cancer treatment and the strategies being explored to improve the existing methods are discussed. This review highlights the advantages and limitations of personalized medicine in colon cancer therapy. The current scenario of personalized medicine in developed countries and the challenges faced in middle- and low-income countries are also summarized. Finally, we discuss the future perspectives of personalized medicine in colon cancer and how it could be integrated into the healthcare systems.
Collapse
Affiliation(s)
- Amit Dey
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Abhijit Mitra
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Suhanya Prasad
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Białystok, Poland
| | | | - Hong Zhang
- School of Medicine, Department of Medical Sciences, Orebro University, Örebro, Sweden
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| |
Collapse
|
17
|
BRAF gene as a potential target to attenuate drug resistance and treat cancer. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
18
|
Mirallas O, López-Valbuena D, García-Illescas D, Fabregat-Franco C, Verdaguer H, Tabernero J, Macarulla T. Advances in the systemic treatment of therapeutic approaches in biliary tract cancer. ESMO Open 2022; 7:100503. [PMID: 35696747 PMCID: PMC9198382 DOI: 10.1016/j.esmoop.2022.100503] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Biliary tract cancers (BTCs) are a rare and heterogenous group with an increasing incidence and high mortality rate. The estimated new cases and deaths of BTC worldwide are increasing, but the incidence and mortality rates in South East Asia are the highest worldwide, representing a real public health problem in these regions. BTC has a poor prognosis with a median overall survival <12 months. Thus, an urgent unmet clinical need for BTC patients exists and must be addressed. RESULTS The backbone treatment of these malignancies is chemotherapy in first- and second-line setting, but in the last decade a rich molecular landscape has been discovered, expanding conceivable treatment options. Some druggable molecular aberrations can be treated with new targeted therapies and have already demonstrated efficacy in patients with BTC, improving clinical outcomes, such as the FGFR2 or IDH1 inhibitors. Many other molecular alterations are being discovered and the treatment of BTC will change in the near future from our current clinical practice. CONCLUSIONS In this review we discuss the epidemiology, molecular characteristics, present treatment approaches, review the recent therapeutic advances, and explore future directions for patients with BTC. Due to the rich molecular landscape of BTC, molecular profiling should be carried out early. Ongoing research will bring new targeted treatments and immunotherapy in the near future.
Collapse
Affiliation(s)
- O Mirallas
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - D López-Valbuena
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - D García-Illescas
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - C Fabregat-Franco
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - H Verdaguer
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - J Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - T Macarulla
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
19
|
Bukhari SNA. Synthesis and evaluation of new chalcones and oximes as anticancer agents. RSC Adv 2022; 12:10307-10320. [PMID: 35424971 PMCID: PMC8973297 DOI: 10.1039/d2ra01198k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/28/2022] [Indexed: 12/28/2022] Open
Abstract
Complex illnesses, such as cancer, are often caused by many disorders, gene mutations, or pathways. Biological pathways play a significant part in the development of these diseases. Multi-target directed ligands (MTDLs) have been used by medicinal chemists recently in an effort to find single molecules that can affect many targets concurrently. In this work, several chalcones containing the ligustrazine moiety were synthesized and tested for their in vitro anticancer activity and several cancer markers, including EGFR, BRAFV600E, c-Met, and tubulin polymerization, in order to uncover multitarget bioactive compounds. In assays using multiple cancer cell lines, the majority of the compounds examined showed strong anticancer activity against them. To synthesize oximes, all of the chalcones were used as precursors. The IC50 values of two compounds (11g and 11e) were found to be 0.87, 0.28, 2.43, 1.04 μM and 11d, 1.47, 0.79, 3.8, 1.63 μM respectively, against A-375, MCF-7, HT-29 and H-460 cell lines. These IC50 values revealed an excellent antiproliferative activity compared to those of the positive control foretinib, (IC50 = 1.9, 1.15, 3.97, and 2.86 μM). Careful examination of their structure and configuration revealed that both compounds had an oxime functional group with z configuration, in place of carbonyl functional group, along with a 2-phenyl thiophenyl moiety with or without a bromo group at position-5. The possible binding pattern was implied by docking simulation, inferring the possibility of introducing interactions with the nearby tubulin chain. Since the novel structural trial has been conducted with a detailed structure activity relationship discussion, this work might stimulate new ideas in further modification of multitarget anti-cancer agents and therapeutic approaches. Discovery of multitarget anticancer agents by modifications of natural compound.![]()
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka Aljouf 72388 Saudi Arabia +96 6565738896
| |
Collapse
|
20
|
El Sissy FN, Wassef M, Faucon B, Salvan D, Nadaud S, Coulet F, Adle-Biassette H, Soubrier F, Bisdorff A, Eyries M. Somatic Mutational Landscape of Extracranial Arteriovenous Malformations and Phenotypic Correlations. J Eur Acad Dermatol Venereol 2022; 36:905-912. [PMID: 35238086 DOI: 10.1111/jdv.18046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/15/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Somatic genetic variants may be the cause of extracranial arteriovenous malformations, but few studies have explored these genetic anomalies, and no genotype-phenotype correlations have been identified. OBJECTIVES To characterize the somatic genetic landscape of extracranial arteriovenous malformations and correlate these findings with the phenotypic characteristics of these lesions. METHODS This study included twenty-three patients with extracranial arteriovenous malformations that were confirmed clinically and treated by surgical resection, and for whom frozen tissue samples were available. Targeted next-generation sequencing analysis of tissues was performed using a gene panel that included vascular disease-related genes and tumor-related genes. RESULTS We identified a pathogenic variant in 18 out of 23 samples (78.3%). Pathogenic variants were mainly located in MAP2K1 (n=7) and KRAS (n=6), and more rarely in BRAF (n=2) and RASA1 (n=3). KRAS variants were significantly (p<0.005) associated with severe extended facial arteriovenous malformations, for which relapse after surgical resection is frequently observed, while MAP2K1 variants were significantly (p<0.005) associated with less severe, limited arteriovenous malformations located on the lips. CONCLUSIONS Our study highlights a high prevalence of pathogenic somatic variants, predominantly in MAP2K1 and KRAS, in extracranial arteriovenous malformations. In addition, our study identifies for the first time a correlation between the genotype, clinical severity and angiographic characteristics of extracranial arteriovenous malformations. The RAS/MAPK variants identified in this study are known to be associated with malignant tumors for which targeted therapies have already been developed. Thus, identification of these somatic variants could lead to new therapeutic options to improve the management of patients with extracranial arteriovenous malformations.
Collapse
Affiliation(s)
- F N El Sissy
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Department of Pathology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Faculty of Medicine, Paris, France
| | - M Wassef
- Department of Pathology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Faculty of Medicine, Paris, France
| | - B Faucon
- Department of Otorhinolaryngology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - D Salvan
- Department of Otorhinolaryngology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - S Nadaud
- Sorbonne Université, INSERM, UMR_S1166, Unité de recherche sur les maladies cardiovasculaires, ICAN, le métabolisme et la nutrition, Paris, France
| | - F Coulet
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - H Adle-Biassette
- Department of Pathology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Faculty of Medicine, Paris, France
| | - F Soubrier
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, INSERM, UMR_S1166, Unité de recherche sur les maladies cardiovasculaires, ICAN, le métabolisme et la nutrition, Paris, France
| | - A Bisdorff
- Department of Neuroradiology, Lariboisère Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - M Eyries
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, INSERM, UMR_S1166, Unité de recherche sur les maladies cardiovasculaires, ICAN, le métabolisme et la nutrition, Paris, France
| |
Collapse
|
21
|
Molecular profile of FLT3-mutated relapsed/refractory AML patients in the phase 3 ADMIRAL study of gilteritinib. Blood Adv 2022; 6:2144-2155. [PMID: 35130342 PMCID: PMC9006281 DOI: 10.1182/bloodadvances.2021006489] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/14/2022] [Indexed: 11/22/2022] Open
Abstract
Patients with FLT3-mutated R/R AML benefitted from gilteritinib regardless of comutations, FLT3-ITD allelic ratio, or FLT3-ITD length. Resistance to gilteritinib was associated with acquired Ras/MAPK pathway gene mutations and FLT3 F691L mutations.
The phase 3 Study of ASP2215 Versus Salvage Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With FMS-like Tyrosine Kinase (FLT3) Mutation (ADMIRAL) trial demonstrated the superiority of the FLT3 inhibitor, gilteritinib, to salvage chemotherapy (SC) in patients with FLT3-mutated relapsed or refractory (R/R) AML. Baseline comutations, FLT3-internal tandem duplication (ITD) allelic ratio and length, and treatment-emergent mutations were analyzed in patients in the ADMIRAL trial. Baseline comutations were grouped according to gene subgroups (DNA methylation/hydroxymethylation, transcription, chromatin–spliceosome, receptor tyrosine kinase-Ras signaling, TP53-aneuploidy, NPM1, DNMT3A, DNMT3A/NPM1, WT-1, and IDH1/IDH2). Across all but 1 gene subgroup (TP53-aneuploidy), higher pretransplant response rates and a trend toward longer overall survival were observed with gilteritinib vs SC. Patients with DNMT3A/NPM1 comutations who received gilteritinib had the most favorable outcomes of any molecular subgroup analyzed. Survival outcomes with gilteritinib were not adversely affected by FLT3-ITD allelic ratio, FLT3-ITD length, or multiple FLT3-ITD mutations. Among patients who relapsed on gilteritinib, Ras/mitogen-activated protein kinase (MAPK) pathway and FLT3 F691L gene mutations were the most common mutational events associated with treatment resistance. However, the occurrence of Ras/MAPK pathway gene mutations at baseline did not preclude a clinical benefit from gilteritinib. Acquisition of multiple Ras/MAPK pathway gene mutations at relapse suggests a high level of pathway reactivation is needed to overcome the gilteritinib treatment effect. These findings provide insight into the R/R AML molecular profile and the impact of FLT3 inhibitors on mutational evolution associated with treatment resistance and benefit of gilteritinib across a wide spectrum of molecular and genetic subgroups in FLT3-mutated R/R AML. This trial was registered at www.clinicaltrials.gov as #NCT02421939.
Collapse
|
22
|
Potential of Withaferin-A, Withanone and Caffeic Acid Phenethyl ester as ATP-competitive inhibitors of BRAF: A bioinformatics study. Curr Res Struct Biol 2022; 3:301-311. [PMID: 35028596 PMCID: PMC8714769 DOI: 10.1016/j.crstbi.2021.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/22/2022] Open
Abstract
Serine/threonine-protein kinase B-raf (BRAF) plays a significant role in regulating cell division and proliferation through MAPK/ERK pathway. The constitutive expression of wild-type BRAF (BRAFWT) and its mutant forms, especially V600E (BRAFV600E), has been linked to multiple cancers. Various synthetic drugs have been approved and are in clinical trials, but most of them are reported to become ineffective within a short duration. Therefore, combinational therapy involving multiple drugs are often recruited for cancer treatment. However, they lead to toxicity and adverse side effects. In this computational study, we have investigated three natural compounds, namely Withaferin-A (Wi-A), Withanone (Wi-N) and Caffeic Acid Phenethyl ester (CAPE) for anti-BRAFWT and anti-BRAFV600E activity. We found that these compounds could bind stably at ATP-binding site in both BRAFWT and BRAFV600E proteins. In-depth analysis revealed that these compounds maintained the active conformation of wild-type BRAF protein by inducing αC-helix-In, DFG-In, extended activation segment and well-aligned R-spine residues similar to already known drugs Vemurafenib (VEM), BGB283 and Ponatinib. In terms of binding energy, among the natural compounds, CAPE showed better affinity towards both wild-type and V600E mutant proteins than the other two compounds. These data suggested that CAPE, Wi-A and Wi-N have potential to block constitutive autophosphorylation of BRAF and hence warrant in vitro and in vivo experimental validation. Out of all the human cancers approximately 8% involve BRAF mutations. The 40–50% of the commercialized drugs in the market are from the natural sources or inspired by it. Three natural compounds Withaferin-A , Withanone and Caffeic acid phenethyl ester (CAPE) have been studied against BRAF. CAPE binds with higher binding affinity with BRAF wild type protein and BRAF V600E mutant protein than other natural compounds.
Collapse
|
23
|
Coperchini F, Melillo RM, Rotondi M. Editorial: Further advances in understanding the endocrine cancer microenvironment. Front Endocrinol (Lausanne) 2022; 13:1009963. [PMID: 36093100 PMCID: PMC9449897 DOI: 10.3389/fendo.2022.1009963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Francesca Coperchini
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia, Italy
| | - Rosa Marina Melillo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, Naples, Italy
| | - Mario Rotondi
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- *Correspondence: Mario Rotondi,
| |
Collapse
|
24
|
gcMECM: graph clustering of mutual exclusivity of cancer mutations. BMC Bioinformatics 2021; 22:592. [PMID: 34906079 PMCID: PMC8670134 DOI: 10.1186/s12859-021-04505-w] [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: 02/01/2021] [Accepted: 11/30/2021] [Indexed: 11/29/2022] Open
Abstract
Background Next-generation sequencing platforms allow us to sequence millions of small fragments of DNA simultaneously, revolutionizing cancer research. Sequence analysis has revealed that cancer driver genes operate across multiple intricate pathways and networks with mutations often occurring in a mutually exclusive pattern. Currently, low-frequency mutations are understudied as cancer-relevant genes, especially in the context of networks. Results Here we describe a tool, gcMECM, that enables us to visualize the functionality of mutually exclusive genes in the subnetworks derived from mutation associations, gene–gene interactions, and graph clustering. These subnetworks have revealed crucial biological components in the canonical pathway, especially those mutated at low frequency. Examining the subnetwork, and not just the impact of a single gene, significantly increases the statistical power of clinical analysis and enables us to build models to better predict how and why cancer develops. Conclusions gcMECM uses a computationally efficient and scalable algorithm to identify subnetworks in a canonical pathway with mutually exclusive mutation patterns and distinct biological functions.
Collapse
|
25
|
Chao WR, Lee YJ, Lee MY, Sheu GT, Han CP. High frequency of BRAF mutations in primary mucinous ovarian carcinoma of Taiwanese patients. Taiwan J Obstet Gynecol 2021; 60:1072-1077. [PMID: 34794740 DOI: 10.1016/j.tjog.2021.09.019] [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] [Accepted: 05/26/2021] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE Considering the clinical evidence of BRAF inhibitors that can treat melanoma patients successfully, we aimed to investigate the status of BRAF mutations of primary mucinous ovarian carcinomas (MOC) in Taiwanese women, and apply the emerging paradigm classification of BRAF mutation groups. MATERIALS AND METHODS 20 archived primary MOC samples were analyzed. The BRAF mutations of activation segment (exon 15), CR3 (conserved regions 3), kinase domain of the BRAF gene were analyzed using the highly sensitive BRAF mutant enriched kit (FemtoPath®) with Sanger sequencing method. Additionally, we extended our prior reported data of HER2 aberrations and KRAS mutation into this study in order to compare with the status of BRAF mutation. RESULTS Of them (n = 20), 16 (80%) harbored BRAF missense mutations. Their mutation profile and case number (n) were categorized as (1) class I: V600E (n=1), V600M (n = 1); (2) class II: A598V (n = 1), T599I (n = 10); (3) class III: none (n = 0); and (4) unclassified variants: S602F (n = 2), T599I/S602F (n = 1). The BRAF S602F is novel. The prevalence of BRAF mutation is significantly higher than either HER2 mutation (80% vs. 35%; p = 0.022) or HER2 amplification (80% vs. 35%; p = 0.022). However, the mutation rates of BRAF and KRAS were not significantly different (80% vs. 60%; p = 0.289). CONCLUSION Activating BRAF mutation, HER2 amplification, HER2 mutation and KRAS mutation were not mutually exclusive. However, they may even have a synergistic effect in tumorigenesis. BRAF mutation is not uncommon in primary MOC of Taiwanese. The BRAF mutant (T599I) stands the majority. We suggested that there was a lower potential response to the existing V600 BRAF inhibitors, but may be responsive to dual BRAF plus MEK inhibitors or single MEK inhibitor. Further studies are warranted to investigate the clinical benefits of newly targeted therapy in recurrent or advanced stage primary MOC patients carrying different classes of BRAF mutation.
Collapse
Affiliation(s)
- Wan-Ru Chao
- Department of Pathology, Chung-Shan Medical University, Taichung, Taiwan; Department of Pathology, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Ju Lee
- Department of Pathology, Chung-Shan Medical University, Taichung, Taiwan; Department of Pathology, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Yung Lee
- Department of Statistics and Informatics Science, Providence University, Taichung, Taiwan
| | - Gwo-Tarng Sheu
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Chih-Ping Han
- Department of Pathology, Chung-Shan Medical University, Taichung, Taiwan; Department of Pathology, Chung-Shan Medical University Hospital, Taichung, Taiwan; Department of Obstetrics and Gynecology, Chung-Shan Medical University and Chung-Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
26
|
Babačić H, Eriksson H, Pernemalm M. Plasma proteome alterations by MAPK inhibitors in BRAF V600-mutated metastatic cutaneous melanoma. Neoplasia 2021; 23:783-791. [PMID: 34246984 PMCID: PMC8274243 DOI: 10.1016/j.neo.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/22/2022]
Abstract
Approximately half of metastatic cutaneous melanomas (CM) harbor a mutation in the BRAF protooncogene, upregulating the mitogen-activated protein kinase (MAPK)-pathway. The development of inhibitors targeting the MAPK pathway (MAPKi), i.e., BRAF- and MEK-inhibitors (BRAFi and MEKi), have substantially improved the survival in BRAFV600E/K-mutated stage IV metastatic CM. However, most patients develop resistance to treatment and no predictive biomarkers exist in practice. This study aimed at discovering plasma proteome changes during treatment MAPKi in patients with metastatic (stage IV) CM. Matched plasma samples before (pre) and during treatment (trm) from 23 patients with stage IV CM, treated with BRAF-inhibitors (BRAFi) alone or BRAF- and MEK- inhibitors combined (BRAFi and MEKi), were collected and analyzed with targeted proteomics by proximity extension assays. Additionally, plasma from 9 patients treated with BRAFi and MEKi was analyzed with in-depth high-resolution isoelectric focusing liquid-chromatography mass-spectrometry proteomics. Alterations of plasma proteins involved in granzyme and interferon gamma pathways were detected in patients treated with BRAFi, and cell adhesion-, neutrophil degranulation-, and proteolysis pathways in patients treated with BRAFi and MEKi. Several proteins were associated with progression-free survival after MAPKi treatment. We show that the majority of the altered plasma proteins were traceable to BRAFV600E-mutant metastatic CM tissue at mRNA level in 154 patients from the TCGA, further strengthening their involvement in tumoral response to treatment. This wide screen of plasma proteins unravels proteins that may serve as predictive and/or prognostic biomarkers of MAPKi treatment, opening a window of opportunity for plasma biomarker discovery in MAPKi-treatment of BRAFV600-mutant metastatic CM.
Collapse
Affiliation(s)
- Haris Babačić
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Hanna Eriksson
- Theme Cancer / Department of Oncology, Karolinska University Hospital, Stockholm, Sweden.
| | - Maria Pernemalm
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
27
|
Cen S, Liu K, Zheng Y, Shan J, Jing C, Gao J, Pan H, Bai Z, Liu Z. BRAF Mutation as a Potential Therapeutic Target for Checkpoint Inhibitors: A Comprehensive Analysis of Immune Microenvironment in BRAF Mutated Colon Cancer. Front Cell Dev Biol 2021; 9:705060. [PMID: 34381786 PMCID: PMC8350390 DOI: 10.3389/fcell.2021.705060] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
BRAF mutated colon cancer presents with poor survival, and the treatment strategies are controversial. The tumor microenvironment, which plays a key role in tumorigenesis as well as responses to treatments, of this subtype is largely unknown. In the present study, we analyzed the differences of immune microenvironments between BRAF mutated and BRAF wild-type colon cancer utilizing datasets from The Cancer Genome Atlas and Gene Expression Omnibus and confirmed the findings by tissue specimens of patients. We found that BRAF mutated colon cancer had more stromal cells, more immune cell infiltration, and lower tumor purity. Many immunotherapeutic targets, including PD-1, PD-L1, CTLA-4, LAG-3, and TIM-3, were highly expressed in BRAF mutated patients. BRAF mutation was also correlated with higher proportions of neutrophils and macrophages M1, and lower proportions of plasma cells, dendritic cells resting, and T cells CD4 naïve. In conclusion, our study demonstrates a different pattern of the immune microenvironment in BRAF mutated colon cancer and provides insights into the future use of checkpoint inhibitors in this subgroup of patients.
Collapse
Affiliation(s)
- Shuyi Cen
- Department of Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Colorectal Cancerm, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yu Zheng
- Department of Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianzhen Shan
- Department of Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Jing
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Colorectal Cancerm, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jiale Gao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Colorectal Cancerm, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Hongming Pan
- Department of Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Colorectal Cancerm, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhen Liu
- Department of Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Lassen UN, Makaroff LE, Stenzinger A, Italiano A, Vassal G, Garcia-Foncillas J, Avouac B. Precision oncology: a clinical and patient perspective. Future Oncol 2021; 17:3995-4009. [PMID: 34278817 DOI: 10.2217/fon-2021-0688] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Molecular characterization of tumors has shifted cancer treatment strategies away from nonspecific cytotoxic treatment of histology-specific tumors toward targeting of actionable mutations that can be found across multiple cancer types. The development of high-throughput technologies such as next-generation sequencing, combined with decision support applications and availability of patient databases, has provided tools that optimize disease management. Precision oncology has proven success in improving outcomes and quality of life, as well as identifying and overcoming mechanisms of drug resistance and relapse. Addressing challenges that impede its use will improve matching of therapies to patients. Here we review the current status of precision oncology medicine, emphasizing its impact on patients - what they understand about precision oncology medicine and their hopes for the future.
Collapse
Affiliation(s)
| | - Lydia E Makaroff
- Fight Bladder Cancer, Oxfordshire, OX39 4DJ, UK.,World Bladder Cancer Patient Coalition, Brussels, Belgium
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, 69120, Germany
| | | | - Gilles Vassal
- Gustave Roussy Comprehensive Cancer Center, & Unversity Paris-Saclay, Villejuif, 94805, France
| | - Jesus Garcia-Foncillas
- University Cancer Institute & The Department of Oncology, University Hospital Fundacion Jimenez Diaz, Autonomous University, Madrid, 28033, Spain
| | | |
Collapse
|
29
|
Chanda M, Cohen MS. Advances in the discovery and development of melanoma drug therapies. Expert Opin Drug Discov 2021; 16:1319-1347. [PMID: 34157926 DOI: 10.1080/17460441.2021.1942834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Therapeutic strategies for melanoma have evolved significantly over the last decade shifting from cytotoxic chemotherapies like dacarbazine to targeted therapies and immunotherapies including immune checkpoint inhibitors. These new drug therapies have improved overall as well as progression-free survival, lowering the mortality of this cancer for melanoma patients with advanced disease. Newer strategies incorporate combination therapies that harness synergies between mechanisms of anticancer efficacy as well as help overcome resistance issues of monotherapies, which remain a challenge. AREAS COVERED This review looks at each class of drug therapy for melanoma and provides an overview of the preclinical mechanism of action, the clinical efficacy data, and their applications in combination therapy regimens. NCCN treatment guidelines, safety, toxicity, and immune-related adverse events are also described as well as a note on cost. EXPERT OPINION Numerous ongoing trials continue to evaluate the role of novel therapies and combinations for this challenging disease and understanding their mechanism of action, risks, benefits, and treatment guidelines can help care providers and patients have a more comprehensive and tailored discussion of treatment options and expectations.
Collapse
Affiliation(s)
- Monica Chanda
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Mark S Cohen
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA.,Department of Surgery, University of Michigan, Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Ying H, Lin A, Liang J, Zhang J, Luo P. Association Between FSIP2 Mutation and an Improved Efficacy of Immune Checkpoint Inhibitors in Patients With Skin Cutaneous Melanoma. Front Mol Biosci 2021; 8:629330. [PMID: 34113648 PMCID: PMC8186463 DOI: 10.3389/fmolb.2021.629330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have shown remarkable success in treating skin cutaneous melanoma (SKCM); however, the response to treatment varies greatly between patients. Considering that the efficacy of ICI treatment is influenced by many factors, we selected the Fibrosheath interacting protein 2 (FSIP2) gene and systematically analyzed its potential to predict the efficacy of ICI treatment. Methods Patient data were collected from an ICI treatment cohort (n = 120) and a The Cancer Genome Atlas (TCGA)-SKCM cohort (n = 467). The data were divided into an FSIP2-mutant (MT) group and FSIP2-wild-type (WT) group according to FSIP2 mutation status. In this study, we analyzed the patients' overall survival rate, tumor mutational burden (TMB), neoantigen load (NAL), copy number variation (CNV), cell infiltration data and immune-related genes. We used gene set enrichment analysis (GSEA) to delineate biological pathways and processes associated with the efficacy of immunotherapy. Results The efficacy of ICI treatment of SKCM patients with FSIP2 mutation was significantly better than that of patients without FSIP2 mutation. The patients in the FSIP2-MT group had higher tumor immunogenicity and lower regulatory T cell (Treg) infiltration. Results of GSEA showed that pathways related to tumor progression (MAPK and FGFR), immunomodulation, and IL-2 synthesis inhibition were significantly downregulated in the FSIP2-MT group. Conclusion Our research suggests that the FSIP2 gene has the potential to predict the efficacy of ICI treatment. The high tumor immunogenicity and low Treg levels observed may be closely related to the fact that patients with FSIP2-MT can benefit from ICI treatment.
Collapse
Affiliation(s)
- Haoxuan Ying
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Southern Medical University, Guangzhou, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junyi Liang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Discontinuation of BRAF/MEK-Directed Targeted Therapy after Complete Remission of Metastatic Melanoma-A Retrospective Multicenter ADOReg Study. Cancers (Basel) 2021; 13:cancers13102312. [PMID: 34065877 PMCID: PMC8151093 DOI: 10.3390/cancers13102312] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
The advent of BRAF/MEK inhibitors (BRAFi/MEKi) has significantly improved progression-free (PFS) and overall survival (OS) for patients with advanced BRAF-V600-mutant melanoma. Long-term survivors have been identified particularly among patients with a complete response (CR) to BRAF/MEK-directed targeted therapy (TT). However, it remains unclear which patients who achieved a CR maintain a durable response and whether treatment cessation might be a safe option in these patients. Therefore, this study investigated the impact of treatment cessation on the clinical course of patients with a CR upon BRAF/MEK-directed-TT. We retrospectively selected patients with BRAF-V600-mutant advanced non-resectable melanoma who had been treated with BRAFi ± MEKi therapy and achieved a CR upon treatment out of the multicentric skin cancer registry ADOReg. Data on baseline patient characteristics, duration of TT, treatment cessation, tumor progression (TP) and response to second-line treatments were collected and analyzed. Of 461 patients who received BRAF/MEK-directed TT 37 achieved a CR. TP after initial CR was observed in 22 patients (60%) mainly affecting patients who discontinued TT (n = 22/26), whereas all patients with ongoing TT (n = 11) maintained their CR. Accordingly, patients who discontinued TT had a higher risk of TP compared to patients with ongoing treatment (p < 0.001). However, our data also show that patients who received TT for more than 16 months and who discontinued TT for other reasons than TP or toxicity did not have a shorter PFS compared to patients with ongoing treatment. Response rates to second-line treatment being initiated in 21 patients, varied between 27% for immune-checkpoint inhibitors (ICI) and 60% for BRAFi/MEKi rechallenge. In summary, we identified a considerable number of patients who achieved a CR upon BRAF/MEK-directed TT in this contemporary real-world cohort of patients with BRAF-V600-mutant melanoma. Sustained PFS was not restricted to ongoing TT but was also found in patients who discontinued TT.
Collapse
|
32
|
Wang N, Wen J, Ren W, Wu Y, Deng C. Upregulation of TRIB2 by Wnt/β-catenin activation in BRAF V600E papillary thyroid carcinoma cells confers resistance to BRAF inhibitor vemurafenib. Cancer Chemother Pharmacol 2021; 88:155-164. [PMID: 33860836 DOI: 10.1007/s00280-021-04270-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/25/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE The BRAFV600E mutation is an oncogenic driver associated with aggressive tumor behaviors and increased mortality among patients with papillary thyroid cancer (PTC). Although the BRAF inhibitor vemurafenib gave promising results in BRAFV600E-mutant PTC, resistance development remains a major clinical challenge. This study aimed to explore the mechanisms underlying drug resistance in PTC. METHODS Two vemurafenib-resistant PTC cell lines (KTC1 and BCPAP) were established by continuous treatment with vemurafenib for 5 months. The knockdown and upregulation of Tribbles homolog 2 (TRIB2) in PTC cells were achieved by the transfection with short hairpin RNA against TRIB2 or recombinant lentiviral vector carrying TRIB2, respectively. The β-catenin inhibitor, ICG-001, was used for the inhibition of the Wnt/β-catenin signaling in PTC cells. RESULTS Vemurafenib-resistant PTC cells showed higher TRIB2 expression, upregulated ERK and AKT activation, enhanced invasive capacity, and increased epithelial-mesenchymal transition compared to the drug-sensitive groups. TRIB2 knockdown repressed the activation of ERK and AKT, inhibited invasion and EMT, and induced apoptosis of PTC cells. TRIB2 deficiency also enhanced the sensitivity of both PTC cells to vemurafenib. Vemurafenib-resistant PTC cells showed elevated expression of β-catenin in both cytoplasm and nucleus. The pre-incubation of cells with β-catenin inhibitor significantly inhibited TRIB2 expression, suppressed EMT, and repressed the activation of ERK and AKT in vemurafenib-resistant cells. CONCLUSION Our study showed that the upregulation of TRIB2 by the Wnt/β-catenin activation confers resistance to vemurafenib in PTC with BRAFV600 mutation. These findings support the potential use of TRIB2 as a therapeutic target for resistant PTC.
Collapse
Affiliation(s)
- Nianxue Wang
- Department of Immunology, Guizhou Medical University, Guiyang City, 550025, Guizhou Province, China
| | - Jing Wen
- Department of Ultrasonic Center, Affiliated Hospital of Guizhou Medical University, Guiyang City, 550004, Guizhou Province, China
| | - Wei Ren
- Department of Immunology, Guizhou Medical University, Guiyang City, 550025, Guizhou Province, China
| | - Yuting Wu
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Guiyang City, 550004, Guizhou Province, China
| | - Chaonan Deng
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Guiyang City, 550004, Guizhou Province, China.
| |
Collapse
|
33
|
Bourque MS, Salek M, Sabin ND, Canale M, Upadhyaya SA. Comment on: Response to the BRAF/MEK inhibitors dabrafenib/trametinib in an adolescent with a BRAF V600E mutated anaplastic ganglioglioma intolerant to vemurafenib. Pediatr Blood Cancer 2021; 68:e28814. [PMID: 33211390 DOI: 10.1002/pbc.28814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Melissa S Bourque
- Department of Pharmaceutical Services, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Marta Salek
- Department of Hematology/Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Noah D Sabin
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Meredith Canale
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Santhosh A Upadhyaya
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
34
|
Xing H, Song Y, Zhang Z, Koch PD. Clinical Characteristics of BRAF V600E Gene Mutation in Patients of Epilepsy-Associated Brain Tumor: a Meta-analysis. J Mol Neurosci 2021; 71:1815-1824. [PMID: 33791912 DOI: 10.1007/s12031-021-01837-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/24/2021] [Indexed: 11/27/2022]
Abstract
Epilepsy-associated brain tumors (EATs) are usually slow-growing, with seizures as the primary and most dominant symptom. BRAF (v-raf murine sarcoma viral oncogene homolog B1) gene mutations have been found in several subsets of EATs; the V600E mutation is currently believed to contribute to the intrinsic epileptogenicity and tumor growth. However, the relationship between BRAF V600E gene mutation and clinical characteristics in EAT patients is not clear. In this study, we aimed to systematically review the frequency of BRAF V600E gene mutation, as well as the relationship between BRAF V600E gene mutation and clinical characteristics, which may help with the diagnosis and treatment of EATs. Cochrane Library, PubMed, Embase, CNKI, WanFang Data, CQVIP, and SinoMed databases were searched up to October 2020 to identify peer-reviewed human studies on assessing the relationship between BRAF V600E gene mutations and clinical characteristics in EATs. The following data were calculated: the frequency of BRAF V600E mutation and clinical feature comparison between BRAF V600E mutations and wild type in EATs, such as gender, age of seizure onset, duration of epilepsy, location of tumors, and Engel outcome. A total of 12 articles were included in the analysis. Five hundred and nine patients with epilepsy-associated brain tumors were screened for the BRAF V600E gene mutation. Among them, 193 patients had the BRAF V600E mutation (34.06%, 95% CI = 0.25 to 0.43). The subgroup analyses of BRAF V600E mutation showed positive frequency of 44.76% (95% CI = 0.36 to 0.54) in ganglioglioma, 24.75% (95% CI = 0.14 to 0.37) in gysembryoplastic neuroepithelial tumor, 2.15% (95% CI = 0 to 0.19) in angiocentric glioma, and 50.16% (95% CI = 0.33 to 0.68) in pleomorphic xanthoastrocytoma. Compared with the overall frequency, the BRAF V600E positive frequency in ganglioglioma was significantly higher (P = 0.0283). We also found that BRAF V600E gene mutation was significantly associated with age at seizure onset (MD = -2.37; 95% CI = -4.33 to -0.41; P = 0.02). There was no statistical difference between BRAF V600E mutations and wild type in gender, duration of epilepsy, tumor site, and Engel outcome comparison. In conclusion, our updated and comprehensive meta-analysis based on a large number of clinical data demonstrated that BRAF V600E mutation is a specific biomarker and could be a pharmacological target for ganglioglioma patients and an exclusion diagnostic criterion for angiocentric glioma. This meta-analysis suggested the critical role of BRAF V600E mutation in the occurrence and development of EATs. Our findings help to elucidate the progression mechanisms in EATs and develop future therapeutic strategies for EATs.
Collapse
Affiliation(s)
- Hang Xing
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Yi Song
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Zhiqi Zhang
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Peter David Koch
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA, 02114, USA
| |
Collapse
|
35
|
Elkoshi Z. The Binary Classification of Protein Kinases. J Inflamm Res 2021; 14:929-947. [PMID: 33776467 PMCID: PMC7988341 DOI: 10.2147/jir.s303750] [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: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
In an earlier publication a binary model for chronic diseases classification has been proposed. According to the model, chronic diseases were classified as “high Treg” or “low Treg” diseases, depending on whether the immune response is anti- or pro-inflammatory and assuming that regulatory T cells are major determinants of the response. It turned out that most cancers are “high Treg” diseases, while autoimmune diseases are “low Treg”. This paper proposes a molecular cause for this binary response. The mechanism proposed depends on the effect of protein kinases on the immune system. Thus, protein kinases are classified as anti- or pro-inflammatory kinases depending on whether they drive “high Treg” or “low Treg” diseases. Observations reported in the earlier publication can be described in terms of anti-inflammatory kinase (AIK) or pro-inflammatory kinase (PIK) activity. Analysis of literature data reveals that the two classes of kinases display distinctive properties relating to their interactions with pathogens and environmental factors. Pathogens that promote Treg activity (“high Treg” pathogens) activate AIKs, while pathogens that suppress Treg activity (“low Treg” pathogens) activate PIKs. Diseases driven by AIKs are associated with “high Treg” pathogens while those diseases driven by PIKs are associated with “low Treg” pathogens. By promoting the activity of AIKs, alcohol consumption increases the risk of “high Treg” cancers but decreases the risk of some “low Treg” autoimmune diseases. JAK1 gain-of-function mutations are observed at high frequencies in autoimmune diseases while JAK1 loss-of-function mutations are observed at high frequencies in cancers with high tumor-infiltrating Tregs. It should also be noted that the corresponding two classes of protein kinase inhibitors are mutually exclusive in terms of their approved therapeutic indications. There is no protein kinase inhibitor that is approved for the treatment of both autoimmune diseases and “high Treg” cancers. Although there are exceptions to the conclusions presented above, these conclusions are supported by the great bulk of published data. It therefore seems that the binary division of protein kinases is a useful tool for elucidating (at the molecular level) many distinctive properties of cancers and autoimmune diseases.
Collapse
Affiliation(s)
- Zeev Elkoshi
- Research and Development Department, Taro Pharmaceutical Industries Ltd, Haifa, Israel
| |
Collapse
|
36
|
Ashrafizadeh M, Delfi M, Hashemi F, Zabolian A, Saleki H, Bagherian M, Azami N, Farahani MV, Sharifzadeh SO, Hamzehlou S, Hushmandi K, Makvandi P, Zarrabi A, Hamblin MR, Varma RS. Biomedical application of chitosan-based nanoscale delivery systems: Potential usefulness in siRNA delivery for cancer therapy. Carbohydr Polym 2021; 260:117809. [PMID: 33712155 DOI: 10.1016/j.carbpol.2021.117809] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022]
Abstract
Gene therapy is an emerging and promising strategy in cancer therapy where small interfering RNA (siRNA) system has been deployed for down-regulation of targeted gene and subsequent inhibition in cancer progression; some issues with siRNA, however, linger namely, its off-targeting property and degradation by enzymes. Nanoparticles can be applied for the encapsulation of siRNA thus enhancing its efficacy in gene silencing where chitosan (CS), a linear alkaline polysaccharide derived from chitin, with superb properties such as biodegradability, biocompatibility, stability and solubility, can play a vital role. Herein, the potential of CS nanoparticles has been discussed for the delivery of siRNA in cancer therapy; proliferation, metastasis and chemoresistance are suppressed by siRNA-loaded CS nanoparticles, especially the usage of pH-sensitive CS nanoparticles. CS nanoparticles can provide a platform for the co-delivery of siRNA and anti-tumor agents with their enhanced stability via chemical modifications. As pre-clinical experiments are in agreement with potential of CS-based nanoparticles for siRNA delivery, and these carriers possess biocompatibiliy and are safe, further studies can focus on evaluating their utilization in cancer patients.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey; Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Masoud Delfi
- Department of Chemical Sciences, University of Naples "Federico II", Complesso Universitario Monte S. Angelo, Via Cintia, 80126 Naples, Italy
| | - Farid Hashemi
- PhD Student of Pharmacology, Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Morteza Bagherian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negar Azami
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Seyed Omid Sharifzadeh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Soodeh Hamzehlou
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Pooyan Makvandi
- Centre for Materials Interface, Istituto Italiano di Tecnologia, Pontedera 56025, Pisa, Italy
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Palacky University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
37
|
Razavi A, Keshavarz-Fathi M, Pawelek J, Rezaei N. Chimeric antigen receptor T-cell therapy for melanoma. Expert Rev Clin Immunol 2021; 17:209-223. [PMID: 33481629 DOI: 10.1080/1744666x.2021.1880895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION In recent years, chimeric antigen receptor (CAR) T cell therapy has emerged as a cancer treatment. After initial therapeutic success for hematologic malignancies, this approach has been extended for the treatment of solid tumors including melanoma. AREAS COVERED T cells need to be reprogramed to recognize specific antigens expressed only in tumor cells, a difficult problem since cancer cells are simply transformed normal cells. Tumor antigens, namely, CSPG4, CD70, and GD2 have been targeted by CAR-T cells for melanoma. Moreover, different co-stimulatory signaling domains need to be selected to direct T cell fate. In this review, various approaches for the treatment of melanoma and their effectiveness are comprehensively reviewed and the current status, challenges, and future perspective of CAR-T cell therapy for melanoma are discussed. Literature search was accomplished in three databases (PubMed, Google scholar, and Clinicaltrials.gov). Published papers and clinical trials were screened and relevant documents were included by checking pre-defined eligibility criteria. EXPERT OPINION Despite obstacles and the risk of adverse events, CAR T cell therapy could be used for patients with treatment-resistant cancer. Clinical trials are underway to determine the efficacy of this approach for the treatment of melanoma.
Collapse
Affiliation(s)
- Azadehsadat Razavi
- Department of Animal Biology, Faculty of Biology Sciences, University of Kharazmi, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - John Pawelek
- Department of Dermatology and the Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden
| |
Collapse
|
38
|
Gene Expression Alterations Associated with Oleuropein-Induced Antiproliferative Effects and S-Phase Cell Cycle Arrest in Triple-Negative Breast Cancer Cells. Nutrients 2020; 12:nu12123755. [PMID: 33297339 PMCID: PMC7762327 DOI: 10.3390/nu12123755] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/18/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
It is known that the Mediterranean diet is effective in reducing the risk of several chronic diseases, including cancer. A critical component of the Mediterranean diet is olive oil, and the relationship between olive oil consumption and the reduced risk of cancer has been established. Oleuropein (OL) is the most prominent polyphenol component of olive fruits and leaves. This compound has been shown to have potent properties in various types of cancers, including breast cancer. In the present study, the molecular mechanism of OL was examined in two racially different triple-negative breast cancer (TNBC) cell lines-African American (AA, MDA-MB-468) and Caucasian American (CA, MDA-MB-231). The data obtained showed that OL effectively inhibits cell growth in both cell lines, concomitant with S-phase cell cycle arrest-mediated apoptosis. The results also showed that OL-treated MDA-MB-468 cells were two-fold more sensitive to OL antiproliferative effect than MDA-MB-231 cells were. At lower concentrations, OL modified the expression of many apoptosis-involved genes. OL was more effective in MDA-MB-468, compared to MDA-MB-231 cells, in terms of the number and the fold-change of the altered genes. In MDA-MB-468 cells, OL induced a noticeable transcription activation in fourteen genes, including two members of the caspase family: caspase 1 (CASP1) and caspase 14 (CASP14); two members of the TNF receptor superfamily: Fas-associated via death domain (FADD) and TNF receptor superfamily 21 (TNFRSF21); six other proapoptotic genes: growth arrest and DNA damage-inducible 45 alpha (GADD45A), cytochrome c somatic (CYCS), BCL-2 interacting protein 2 (BNIP2), BCL-2 interacting protein 3 (BNIP3), BH3 interacting domain death agonist (BID), and B-cell lymphoma/leukemia 10 (BCL10); and the CASP8 and FADD-like apoptosis regulator (CFLAR) gene. Moreover, in MDA-MB-468 cells, OL induced a significant upregulation in two antiapoptotic genes: bifunctional apoptosis regulator (BFAR) and B-Raf proto-oncogene (BRAF) and a baculoviral inhibitor of apoptosis (IAP) repeat-containing 3 (BIRC3). On the contrary, in MDA-MB-231 cells, OL showed mixed impacts on gene expression. OL significantly upregulated the mRNA expression of four genes: BIRC3, receptor-interacting serine/threonine kinase 2 (RIPK2), TNF receptor superfamily 10A (TNFRSF10A), and caspase 4 (CASP4). Additionally, another four genes were repressed, including caspase 6 (CASP6), pyrin domain (PYD), and caspase recruitment domain (CARD)-containing (PAYCARD), baculoviral IAP repeat-containing 5 (BIRC5), and the most downregulated TNF receptor superfamily member 11B (TNFRSF11B, 16.34-fold). In conclusion, the data obtained indicate that the two cell lines were markedly different in the anticancer effect and mechanisms of oleuropein's ability to alter apoptosis-related gene expressions. The results obtained from this study should also guide the potential utilization of oleuropein as an adjunct therapy for TNBC to increase chemotherapy effectiveness and prevent cancer progression.
Collapse
|
39
|
Bellei B, Migliano E, Picardo M. A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue. Cancers (Basel) 2020; 12:cancers12113400. [PMID: 33212834 PMCID: PMC7697272 DOI: 10.3390/cancers12113400] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Melanoma cells reside in a complex stromal microenvironment, which is a critical component of disease onset and progression. Mesenchymal or fibroblastic cell type are the most abundant cellular element of tumor stroma. Factors secreted by melanoma cells can activate non-malignant associated fibroblasts to become melanoma associate fibroblasts (MAFs). MAFs promote tumorigenic features by remodeling the extracellular matrix, supporting tumor cells proliferation, neo-angiogenesis and drug resistance. Additionally, environmental factors may contribute to the acquisition of pro-tumorigenic phenotype of fibroblasts. Overall, in melanoma, perturbed tissue homeostasis contributes to modulation of major oncogenic intracellular signaling pathways not only in tumor cells but also in neighboring cells. Thus, targeted molecular therapies need to be considered from the reciprocal point of view of melanoma and stromal cells. Abstract The development of a modified stromal microenvironment in response to neoplastic onset is a common feature of many tumors including cutaneous melanoma. At all stages, melanoma cells are embedded in a complex tissue composed by extracellular matrix components and several different cell populations. Thus, melanomagenesis is not only driven by malignant melanocytes, but also by the altered communication between melanocytes and non-malignant cell populations, including fibroblasts, endothelial and immune cells. In particular, cancer-associated fibroblasts (CAFs), also referred as melanoma-associated fibroblasts (MAFs) in the case of melanoma, are the most abundant stromal cells and play a significant contextual role in melanoma initiation, progression and metastasis. As a result of dynamic intercellular molecular dialogue between tumor and the stroma, non-neoplastic cells gain specific phenotypes and functions that are pro-tumorigenic. Targeting MAFs is thus considered a promising avenue to improve melanoma therapy. Growing evidence demonstrates that aberrant regulation of oncogenic signaling is not restricted to transformed cells but also occurs in MAFs. However, in some cases, signaling pathways present opposite regulation in melanoma and surrounding area, suggesting that therapeutic strategies need to carefully consider the tumor–stroma equilibrium. In this novel review, we analyze four major signaling pathways implicated in melanomagenesis, TGF-β, MAPK, Wnt/β-catenin and Hyppo signaling, from the complementary point of view of tumor cells and the microenvironment.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy;
- Correspondence: ; Tel.: +39-0652666246
| | - Emilia Migliano
- Department of Plastic and Regenerative Surgery, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy;
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy;
| |
Collapse
|
40
|
Quaresmini D, Guida M. Neoangiogenesis in Melanoma: An Issue in Biology and Systemic Treatment. Front Immunol 2020; 11:584903. [PMID: 33193402 PMCID: PMC7658002 DOI: 10.3389/fimmu.2020.584903] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
Neoangiogenesis is a recognized hallmark of cancer, granting tumor cells to dispose of metabolic substrates through a newly created vascular supply. Neoangiogenesis was also confirmed in melanoma, where vascular proliferation is associated with increased aggressiveness and poorer prognosis. Furthermore, melanoma cells show the so-called vascular mimicry, consisting in the assumption of endothelial-like features inducing the expression of pro-angiogenic receptors and ligands, which take part in the interplay with extracellular matrix (ECM) components and are potentiated by the ECM remodeling and the barrier molecule junction alterations that characterize the metastatic phase. Although neoangiogenesis was biologically proven and clinically associated with worse outcomes in melanoma patients, in the past anti-angiogenic therapies were employed with poor improvement of the already unsatisfactory results associated with chemotherapic agents. Among the novel therapies of melanoma, immunotherapy has led to previously unexpected outcomes of treatment, yet there is a still strong need for potentiating the results, possibly by new regimens of combination therapies. Molecular models in many cancer types showed mutual influences between immune responses and vascular normalization. Recently, clinical trials are investigating the efficacy of the association between anti-angiogenetic agents and immune-checkpoint inhibitors to treat advanced stage melanoma. This paper reviews the biological bases of angiogenesis in melanoma and summarizes the currently available clinical data on the use of anti-angiogenetic compounds in melanoma.
Collapse
Affiliation(s)
- Davide Quaresmini
- Rare Tumors and Melanoma Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Michele Guida
- Rare Tumors and Melanoma Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
41
|
Abstract
Erdheim-Chester disease (ECD) is characterized by the infiltration of tissues by foamy CD68+CD1a- histiocytes, with 1500 known cases since 1930. Mutations activating the MAPK pathway are found in more than 80% of patients with ECD, mainly the BRAFV600E activating mutation in 57% to 70% of cases, followed by MAP2K1 in close to 20%. The discovery of BRAF mutations and of other MAP kinase pathway alterations, as well as the co-occurrence of ECD with LCH in 15% of patients with ECD, led to the 2016 revision of the classification of histiocytoses in which LCH and ECD belong to the "L" group. Both conditions are considered inflammatory myeloid neoplasms. Ten percent of ECD cases are associated with myeloproliferative neoplasms and/or myelodysplastic syndromes. Some of the most striking signs of ECD are the long bone involvement (80%-95%), as well as the hairy kidney appearance on computed tomography scan (63%), the coated aorta (40%), and the right atrium pseudo-tumoral infiltration (36%). Central nervous system involvement is a strong prognostic factor and independent predictor of death. Interferon-α seems to be the best initial treatment of ECD. Since 2012, more than 200 patients worldwide with multisystem or refractory ECD have benefitted from highly effective therapy with BRAF and MEK inhibitors. Targeted therapies have an overall, robust, and reproducible efficacy in ECD, with no acquired resistance to date, but their use may be best reserved for the most severe manifestations of the disease, as they may be associated with serious adverse effects and as-yet-unknown long-term consequences.
Collapse
|
42
|
Wei R, Wirkus J, Yang Z, Machuca J, Esparza Y, Mackenzie GG. EGCG sensitizes chemotherapeutic-induced cytotoxicity by targeting the ERK pathway in multiple cancer cell lines. Arch Biochem Biophys 2020; 692:108546. [PMID: 32818507 DOI: 10.1016/j.abb.2020.108546] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/07/2020] [Accepted: 08/09/2020] [Indexed: 02/06/2023]
Abstract
Epigallocatechin-3-gallate (EGCG), a major polyphenol component of green tea, presents anticancer efficacy. However, its exact mechanism of action is not known. In this study, we evaluated the effect of EGCG alone or in combination with current chemotherapeutics [gemcitabine, 5-flourouracil (5-FU), and doxorubicin] on pancreatic, colon, and lung cancer cell growth, as well as the mechanisms involved in the combined action. EGCG reduced pancreatic, colon, and lung cancer cell growth in a concentration and time-dependent manner. EGCG strongly induced apoptosis and blocked cell cycle progression. Moreover, EGCG enhanced the growth inhibitory effect of 5-FU and doxorubicin. Of note, EGCG enhanced 5-FU's and doxorubicin's effect on apoptosis, but not on cell cycle. Mechanistically, EGCG reduced ERK phosphorylation concentration-dependently, and sensitized gemcitabine, 5-FU, and doxorubicin to further suppress ERK phosphorylation in multiple cancer cell lines. In conclusion, EGCG presents a strong anticancer effect in pancreatic, colon, and lung cancer cells and is a robust combination partner for multiple chemotherapeutics as evidenced by reducing cancer cell growth, in part, by inhibiting the ERK pathway.
Collapse
Affiliation(s)
- Ran Wei
- Department of Nutrition, University of California, Davis, CA, 95616, USA; Zhejiang Agriculture and Forestry University, Laboratory of Tea and Human Health, Hangzhou, China
| | - Joanna Wirkus
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Zixuan Yang
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Jazmin Machuca
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Yasmin Esparza
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Gerardo G Mackenzie
- Department of Nutrition, University of California, Davis, CA, 95616, USA; University of California, Comprehensive Cancer Center, Sacramento, CA, USA.
| |
Collapse
|
43
|
Yao L, Jia G, Lu L, Bao Y, Ma W. Factors affecting tumor responders and predictive biomarkers of toxicities in cancer patients treated with immune checkpoint inhibitors. Int Immunopharmacol 2020; 85:106628. [PMID: 32474388 DOI: 10.1016/j.intimp.2020.106628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/24/2020] [Accepted: 05/20/2020] [Indexed: 12/20/2022]
|
44
|
Proietti I, Skroza N, Michelini S, Mambrin A, Balduzzi V, Bernardini N, Marchesiello A, Tolino E, Volpe S, Maddalena P, Di Fraia M, Mangino G, Romeo G, Potenza C. BRAF Inhibitors: Molecular Targeting and Immunomodulatory Actions. Cancers (Basel) 2020; 12:cancers12071823. [PMID: 32645969 PMCID: PMC7408709 DOI: 10.3390/cancers12071823] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
The BRAF inhibitors vemurafenib, dabrafenib and encorafenib are used in the treatment of patients with BRAF-mutant melanoma. They selectively target BRAF kinase and thus interfere with the mitogen-activated protein kinase (MAPK) signalling pathway that regulates the proliferation and survival of melanoma cells. In addition to their molecularly targeted activity, BRAF inhibitors have immunomodulatory effects. The MAPK pathway is involved in T-cell receptor signalling, and interference in the pathway by BRAF inhibitors has beneficial effects on the tumour microenvironment and anti-tumour immune response in BRAF-mutant melanoma, including increased immune-stimulatory cytokine levels, decreased immunosuppressive cytokine levels, enhanced melanoma differentiation antigen expression and presentation of tumour antigens by HLA 1, and increased intra-tumoral T-cell infiltration and activity. These effects promote recognition of the tumour by the immune system and enhance anti-tumour T-cell responses. Combining BRAF inhibitors with MEK inhibitors provides more complete blockade of the MAPK pathway. The immunomodulatory effects of BRAF inhibition alone or in combination with MEK inhibition provide a rationale for combining these targeted therapies with immune checkpoint inhibitors. Available data support the synergy between these treatment approaches, indicating such combinations provide an additional beneficial effect on the tumour microenvironment and immune response in BRAF-mutant melanoma.
Collapse
Affiliation(s)
- Ilaria Proietti
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
- Correspondence: ; Tel.: +39-3334684342 or +39-0773708811
| | - Nevena Skroza
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Simone Michelini
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Alessandra Mambrin
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Veronica Balduzzi
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Nicoletta Bernardini
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Anna Marchesiello
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Ersilia Tolino
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Salvatore Volpe
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Patrizia Maddalena
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Marco Di Fraia
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| | - Giorgio Mangino
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.M.); (G.R.)
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00100 Rome, Italy
| | - Giovanna Romeo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.M.); (G.R.)
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00100 Rome, Italy
| | - Concetta Potenza
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy; (N.S.); (S.M.); (A.M.); (V.B.); (N.B.); (A.M.); (E.T.); (S.V.); (P.M.); (M.D.F.); (C.P.)
| |
Collapse
|
45
|
Yin H, Tang Y, Guo Y, Wen S. Immune Microenvironment of Thyroid Cancer. J Cancer 2020; 11:4884-4896. [PMID: 32626535 PMCID: PMC7330689 DOI: 10.7150/jca.44506] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Thyroid cancer (TC) is a highly heterogeneous endocrine malignancy with an increased incidence in women than in men. Previous studies regarding the pathogenesis of TC focused on the pathological changes of the tumor cells while ignoring the importance of the mesenchymal cells in tumor microenvironment. However, more recently, the stable environment provided by the interaction of thyroid cancer cells with the peri-tumoral stroma has been widely studied. Studies have shown that components of an individual's immune system are closely related to the occurrence, invasion, and metastasis of TC, which may affect response to treatment and prognosis of the patients. This article presents a comprehensive review of the immune cells, secreted soluble mediators and immune checkpoints in the immune microenvironment, mechanisms that promoting TC cells immune evasion and existing immunotherapy strategies. Besides it provides new strategies for TC prognosis prediction and immunotherapy.
Collapse
Affiliation(s)
- Hongyu Yin
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Yemei Tang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Yujia Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,Department of Otolaryngology Head & Neck Surgery, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Shuxin Wen
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,General Hospital, Shenzhen University, Shenzhen 518061, Guangdong, P.R. China
| |
Collapse
|
46
|
Expression of FAM83H and ZNF16 are associated with shorter survival of patients with gallbladder carcinoma. Diagn Pathol 2020; 15:63. [PMID: 32460791 PMCID: PMC7254718 DOI: 10.1186/s13000-020-00985-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/20/2020] [Indexed: 02/03/2023] Open
Abstract
Background Recently, FAM83H was reported to have roles in cancer progression in conjunction with oncogenic molecules such as MYC and b-catenin. Moreover, the data from the public database indicates a molecular relationship between FAM83H and zinc finger proteins, especially between FAM83H and ZNF16. However, studies on FAM83H and ZNF16 in gallbladder cancer have been limited. Methods This study investigated the expression of FAM83H and ZNF16 in 105 gallbladder carcinomas. Results In human gallbladder carcinomas, immunohistochemical expression of FAM83H was significantly associated with ZNF16 expression. In univariate analysis, nuclear and cytoplasmic expression of FAM83H or ZNF16 were significantly associated with shorter survival of gallbladder carcinoma patients. Multivariate analysis revealed the nuclear expression of FAM83H as an independent indicator of poor prognosis of overall survival (p = 0.005) and relapse-free survival (p = 0.005) of gallbladder carcinoma patients. Moreover, co-expression patterns of nuclear FAM83H and ZNF16 were also independent indicators of shorter survival of gallbladder carcinoma patients (overall survival; p < 0.001, relapse-free survival; p < 0.001). Conclusions This study suggests FAM83H and ZNF16 are associated with the progression of gallbladder carcinoma, and the expressions of FAM83H and ZNF16 might be novel prognostic indicators of gallbladder carcinoma patients.
Collapse
|