1
|
Batalha S, Gomes CM, Brito C. Immune microenvironment dynamics of HER2 overexpressing breast cancer under dual anti-HER2 blockade. Front Immunol 2023; 14:1267621. [PMID: 38022643 PMCID: PMC10643871 DOI: 10.3389/fimmu.2023.1267621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The clinical prognosis of the HER2-overexpressing (HER2-OE) subtype of breast cancer (BC) is influenced by the immune infiltrate of the tumor. Specifically, monocytic cells, which are promoters of pro-tumoral immunosuppression, and NK cells, whose basal cytotoxic function may be enhanced with therapeutic antibodies. One of the standards of care for HER2+ BC patients includes the combination of the anti-HER2 antibodies trastuzumab and pertuzumab. This dual combination was a breakthrough against trastuzumab resistance; however, this regimen does not yield complete clinical benefit for a large fraction of patients. Further therapy refinement is still hampered by the lack of knowledge on the immune mechanism of action of this antibody-based dual HER2 blockade. Methods To explore how the dual antibody challenge influences the phenotype and function of immune cells infiltrating the HER2-OE BC microenvironment, we developed in vitro 3D heterotypic cell models of this subtype. The models comprised aggregates of HER2+ BC cell lines and human peripheral blood mononuclear cells. Cells were co-encapsulated in a chemically inert alginate hydrogel and maintained in agitation-based culture system for up to 7 days. Results The 3D models of the HER2-OE immune microenvironment retained original BC molecular features; the preservation of the NK cell compartment was achieved upon optimization of culture time and cytokine supplementation. Challenging the models with the standard-of-care combination of trastuzumab and pertuzumab resulted in enhanced immune cytotoxicity compared with trastuzumab alone. Features of the response to therapy within the immune tumor microenvironment were recapitulated, including induction of an immune effector state with NK cell activation, enhanced cell apoptosis and decline of immunosuppressive PD-L1+ immune cells. Conclusions This work presents a unique human 3D model for the study of immune effects of anti-HER2 biologicals, which can be used to test novel therapy regimens and improve anti-tumor immune function.
Collapse
Affiliation(s)
- Sofia Batalha
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Monteiro Gomes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
2
|
Novel scFv against Notch Ligand JAG1 Suitable for Development of Cell Therapies toward JAG1-Positive Tumors. Biomolecules 2023; 13:biom13030459. [PMID: 36979394 PMCID: PMC10046313 DOI: 10.3390/biom13030459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
The Notch signaling ligand JAG1 is overexpressed in various aggressive tumors and is associated with poor clinical prognosis. Hence, therapies targeting oncogenic JAG1 hold great potential for the treatment of certain tumors. Here, we report the identification of specific anti-JAG1 single-chain variable fragments (scFvs), one of them endowing chimeric antigen receptor (CAR) T cells with cytotoxicity against JAG1-positive cells. Anti-JAG1 scFvs were identified from human phage display libraries, reformatted into full-length monoclonal antibodies (Abs), and produced in mammalian cells. The characterization of these Abs identified two specific anti-JAG1 Abs (J1.B5 and J1.F1) with nanomolar affinities. Cloning the respective scFv sequences in our second- and third-generation CAR backbones resulted in six anti-JAG1 CAR constructs, which were screened for JAG1-mediated T-cell activation in Jurkat T cells in coculture assays with JAG1-positive cell lines. Studies in primary T cells demonstrated that one CAR harboring the J1.B5 scFv significantly induced effective T-cell activation in the presence of JAG1-positive, but not in JAG1-knockout, cancer cells, and enabled specific killing of JAG1-positive cells. Thus, this new anti-JAG1 scFv represents a promising candidate for the development of cell therapies against JAG1-positive tumors.
Collapse
|
3
|
Huang B, Miao L, Liu J, Zhang J, Li Y. A promising antitumor method: Targeting CSC with immune cells modified with CAR. Front Immunol 2022; 13:937327. [PMID: 36032145 PMCID: PMC9403009 DOI: 10.3389/fimmu.2022.937327] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
Tumors pose a great threat to human health; as a subgroup of tumor cells, cancer stem cells (CSCs) contribute to the genesis, development, metastasis, and recurrence of tumors because of their enhanced proliferation and multidirectional differentiation. Thus, a critical step in tumor treatment is to inhibit CSCs. Researchers have proposed many methods to inhibit or reduce CSCs, including monoclonal antibodies targeting specific surface molecules of CSCs, signal pathway inhibitors, and energy metabolic enzyme inhibitors and inducing differentiation therapy. Additionally, immunotherapy with immune cells engineered with a chimeric antigen receptor (CAR) showed favorable results. However, there are few comprehensive reviews in this area. In this review, we summarize the recent CSC targets used for CSC inhibition and the different immune effector cells (T cells, natural killer (NK) cells, and macrophages) which are engineered with CAR used for CSC therapy. Finally, we list the main challenges and options in targeting CSC with CAR-based immunotherapy. The design targeting two tumor antigens (one CSC antigen and one mature common tumor antigen) should be more reasonable and practical; meanwhile, we highlight the potential of CAR-NK in tumor treatment.
Collapse
Affiliation(s)
- Binjie Huang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Jie Liu
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Jiaxing Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Yumin Li,
| |
Collapse
|
4
|
Wang L, Jin Z, Master RP, Maharjan CK, Carelock ME, Reccoppa TBA, Kim MC, Kolb R, Zhang W. Breast Cancer Stem Cells: Signaling Pathways, Cellular Interactions, and Therapeutic Implications. Cancers (Basel) 2022; 14:3287. [PMID: 35805056 PMCID: PMC9265870 DOI: 10.3390/cancers14133287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/02/2022] [Accepted: 07/02/2022] [Indexed: 02/01/2023] Open
Abstract
Breast cancer stem cells (BCSCs) constitute a small population of cells within breast cancer and are characterized by their ability to self-renew, differentiate, and recapitulate the heterogeneity of the tumor. Clinically, BCSCs have been correlated with cancer progression, metastasis, relapse, and drug resistance. The tumorigenic roles of BCSCs have been extensively reviewed and will not be the major focus of the current review. Here, we aim to highlight how the crucial intrinsic signaling pathways regulate the fate of BCSCs, including the Wnt, Notch, Hedgehog, and NF-κB signaling pathways, as well as how different cell populations crosstalk with BCSCs within the TME, including adipocytes, endothelial cells, fibroblasts, and immune cells. Based on the molecular and cellular activities of BCSCs, we will also summarize the targeting strategies for BCSCs and related clinical trials. This review will highlight that BCSC development in breast cancer is impacted by both BCSC endogenous signaling and external factors in the TME, which provides an insight into how to establish a comprehensively therapeutic strategy to target BCSCs for breast cancer treatments.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Immunology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zeng Jin
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rohan P. Master
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Chandra K. Maharjan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Madison E. Carelock
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tiffany B. A. Reccoppa
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Department of Biology, College of Liberal Arts & Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Myung-Chul Kim
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
5
|
Mayoral-Peña K, González Peña OI, Orrantia Clark AM, Flores-Vallejo RDC, Oza G, Sharma A, De Donato M. Biorecognition Engineering Technologies for Cancer Diagnosis: A Systematic Literature Review of Non-Conventional and Plausible Sensor Development Methods. Cancers (Basel) 2022; 14:1867. [PMID: 35454775 PMCID: PMC9030888 DOI: 10.3390/cancers14081867] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 12/21/2022] Open
Abstract
Cancer is the second cause of mortality worldwide. Early diagnosis of this multifactorial disease is challenging, especially in populations with limited access to healthcare services. A vast repertoire of cancer biomarkers has been studied to facilitate early diagnosis; particularly, the use of antibodies against these biomarkers has been of interest to detect them through biorecognition. However, there are certain limitations to this approach. Emerging biorecognition engineering technologies are alternative methods to generate molecules and molecule-based scaffolds with similar properties to those presented by antibodies. Molecularly imprinted polymers, recombinant antibodies, and antibody mimetic molecules are three novel technologies commonly used in scientific studies. This review aimed to present the fundamentals of these technologies and address questions about how they are implemented for cancer detection in recent scientific studies. A systematic analysis of the scientific peer-reviewed literature regarding the use of these technologies on cancer detection was carried out starting from the year 2000 up to 2021 to answer these questions. In total, 131 scientific articles indexed in the Web of Science from the last three years were included in this analysis. The results showed that antibody mimetic molecules technology was the biorecognition technology with the highest number of reports. The most studied cancer types were: multiple, breast, leukemia, colorectal, and lung. Electrochemical and optical detection methods were the most frequently used. Finally, the most analyzed biomarkers and cancer entities in the studies were carcinoembryonic antigen, MCF-7 cells, and exosomes. These technologies are emerging tools with adequate performance for developing biosensors useful in cancer detection, which can be used to improve cancer diagnosis in developing countries.
Collapse
Affiliation(s)
- Kalaumari Mayoral-Peña
- School of Engineering and Sciences, Campus Queretaro, Tecnologico de Monterrey, Av. Epigmenio González No. 500, San Pablo, Queretaro 76130, Mexico; (K.M.-P.); (A.S.)
| | - Omar Israel González Peña
- School of Engineering and Sciences, Campus Monterrey, Tecnologico de Monterrey, Av. Eugenio Garza Sada Sur No. 2501, Tecnológico, Monterrey 64849, Mexico
- Institute for the Future of Education, Tecnologico de Monterrey, Av. Eugenio Garza Sada Sur No. 2501, Tecnológico, Monterrey 64849, Mexico
| | - Alexia María Orrantia Clark
- School of Engineering and Sciences, Campus Mexico City, Tecnologico de Monterrey, C. Puente 222, Ejidos de Huipulco, Tlalpan, Mexico City 14380, Mexico;
| | - Rosario del Carmen Flores-Vallejo
- Department of Biomedical Engineering and Mechatronics, Campus Toluca, Universidad del Valle de México (UVM), C. De Las Palmas Poniente 439, San Jorge Pueblo Nuevo, Metepec 52164, Mexico;
| | - Goldie Oza
- Laboratorio Nacional de Micro y Nanofluídica (LABMyN), Centro de Investigación y Desarrollo Tecnológico en Electroquímica (CIDETEQ), Parque San Fandila, Pedro Escobedo, Queretaro 76703, Mexico;
| | - Ashutosh Sharma
- School of Engineering and Sciences, Campus Queretaro, Tecnologico de Monterrey, Av. Epigmenio González No. 500, San Pablo, Queretaro 76130, Mexico; (K.M.-P.); (A.S.)
| | - Marcos De Donato
- School of Engineering and Sciences, Campus Queretaro, Tecnologico de Monterrey, Av. Epigmenio González No. 500, San Pablo, Queretaro 76130, Mexico; (K.M.-P.); (A.S.)
| |
Collapse
|
6
|
Silva G, Sales-Dias J, Casal D, Alves S, Domenici G, Barreto C, Matos C, Lemos AR, Matias AT, Kucheryava K, Ferreira A, Moita MR, Braga S, Brito C, Cabral MG, Casalou C, Barral DC, Sousa PMF, Videira PA, Bandeiras TM, Barbas A. Development of Dl1.72, a Novel Anti-DLL1 Antibody with Anti-Tumor Efficacy against Estrogen Receptor-Positive Breast Cancer. Cancers (Basel) 2021; 13:cancers13164074. [PMID: 34439228 PMCID: PMC8392387 DOI: 10.3390/cancers13164074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Over 70% of breast cancers (BCs) are estrogen receptor-positive (ER+). The development of endocrine therapy has considerably improved patient outcomes. However, there is a clinical need for novel effective therapies against ER+ BCs, since many of these do not respond to standard therapy, and more than one-third of responders acquire resistance, experience relapse and metastasize. The Notch ligand Delta-like 1 (DLL1) is a key player in ER+ BC development and aggressiveness. Contrary to complete Notch pharmacological inhibitors, antibody-targeting of individual Notch components is expected to have superior therapeutic efficacy and be better tolerated. In this study, we developed and characterized a novel specific anti-DLL1 antibody with efficacy in inhibiting BC cell proliferation, mammosphere formation and angiogenesis, as well as anti-tumor and anti-metastatic efficacy in an ER+ BC mouse model without side effects. Thus, our data suggest that this anti-DLL1 antibody is a promising candidate for ER+ BC treatment. Abstract The Notch-signaling ligand DLL1 has emerged as an important player and promising therapeutic target in breast cancer (BC). DLL1-induced Notch activation promotes tumor cell proliferation, survival, migration, angiogenesis and BC stem cell maintenance. In BC, DLL1 overexpression is associated with poor prognosis, particularly in estrogen receptor-positive (ER+) subtypes. Directed therapy in early and advanced BC has dramatically changed the natural course of ER+ BC; however, relapse is a major clinical issue, and new therapeutic strategies are needed. Here, we report the development and characterization of a novel monoclonal antibody specific to DLL1. Using phage display technology, we selected an anti-DLL1 antibody fragment, which was converted into a full human IgG1 (Dl1.72). The Dl1.72 antibody exhibited DLL1 specificity and affinity in the low nanomolar range and significantly impaired DLL1-Notch signaling and expression of Notch target genes in ER+ BC cells. Functionally, in vitro treatment with Dl1.72 reduced MCF-7 cell proliferation, migration, mammosphere formation and endothelial tube formation. In vivo, Dl1.72 significantly inhibited tumor growth, reducing both tumor cell proliferation and liver metastases in a xenograft mouse model, without apparent toxicity. These findings suggest that anti-DLL1 Dl1.72 could be an attractive agent against ER+ BC, warranting further preclinical investigation.
Collapse
Affiliation(s)
- Gabriela Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Correspondence: ; Tel.: +351-214-469-419
| | - Joana Sales-Dias
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Diogo Casal
- Departamento de Anatomia, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (D.C.); (S.A.)
- iNOVA4Health, CEDOC, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (A.T.M.); (A.F.); (S.B.); (M.G.C.); (C.C.); (D.C.B.)
| | - Sara Alves
- Departamento de Anatomia, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (D.C.); (S.A.)
- Serviço de Anatomia Patológica, Centro Hospitalar de Lisboa Central-Hospital de São José, 1150-199 Lisbon, Portugal
| | - Giacomo Domenici
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Clara Barreto
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
| | - Carolina Matos
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
| | - Ana R. Lemos
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana T. Matias
- iNOVA4Health, CEDOC, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (A.T.M.); (A.F.); (S.B.); (M.G.C.); (C.C.); (D.C.B.)
| | - Khrystyna Kucheryava
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
| | - Andreia Ferreira
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
| | - Maria Raquel Moita
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Sofia Braga
- iNOVA4Health, CEDOC, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (A.T.M.); (A.F.); (S.B.); (M.G.C.); (C.C.); (D.C.B.)
- Unidade de Mama, Instituto CUF de Oncologia, 1998-018 Lisbon, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - M. Guadalupe Cabral
- iNOVA4Health, CEDOC, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (A.T.M.); (A.F.); (S.B.); (M.G.C.); (C.C.); (D.C.B.)
| | - Cristina Casalou
- iNOVA4Health, CEDOC, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (A.T.M.); (A.F.); (S.B.); (M.G.C.); (C.C.); (D.C.B.)
| | - Duarte C. Barral
- iNOVA4Health, CEDOC, NOVA Medical School (NMS), Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (A.T.M.); (A.F.); (S.B.); (M.G.C.); (C.C.); (D.C.B.)
| | - Pedro M. F. Sousa
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula A. Videira
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal;
| | - Tiago M. Bandeiras
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana Barbas
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (J.S.-D.); (G.D.); (C.B.); (C.M.); (A.R.L.); (K.K.); (M.R.M.); (C.B.); (P.M.F.S.); (T.M.B.); (A.B.)
| |
Collapse
|