1
|
Chen P, Tian W, Zeng A, Gu H, Zeng J. Regulating Intratumoral Fungi With Hydrogels: A Novel Approach to Modulating the Tumor Microbiome for Cancer Therapy. Cancer Med 2025; 14:e70900. [PMID: 40304214 PMCID: PMC12041943 DOI: 10.1002/cam4.70900] [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: 01/17/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Fungi in tumors act as a double-edged sword, potentially worsening or alleviating malignancy based on the ecological balance within the tumor microenvironment (TME). Hydrogels, as innovative drug delivery systems, are poised to redefine treatment paradigms. As advanced biomaterials, they offer a versatile platform for encapsulating and releasing antifungal agents and immunomodulators, responding to the TME's unique demands. METHODS We have conducted and collated numerous relevant reviews and studies in recent years from three aspects: Hydrogels, intra-tumoral fungi, and tumor microbe microenvironment, in the hope of identifying the connections between hydrogels and intra-tumoral microbes. RESULTS This review underscores the crucial role of intra-tumoral microbes, particularly fungi, in tumorigenesis, progression, and treatment efficacy. At the same time, we concentrated on the findings of hydrogels investigations, with their remarkable adaptability to the tumor microenvironment emerge as intelligent drug delivery systems. CONCLUSIONS Hydrogels unique ability to precisely target and modulate the tumor microflora, including fungi, endows them with a significant edge in enhancing treatment efficacy. This innovative approach not only holds great promise for improving cancer therapy outcomes but also paves the way for developing novel strategies to control metastasis and prevent cancer recurrence.
Collapse
Affiliation(s)
- Ping Chen
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| | - Weiwei Tian
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| | - Huan Gu
- College of Pharmacy and FoodSouthwest Minzu UniversityChengduChina
| | - Jin Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan ProvinceSichuan‐Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese MedicineSichuan Academy of Chinese Medicine SciencesChengduChina
| |
Collapse
|
2
|
Zhang C, Wang H, Li X, Jiang Y, Sun G, Yu H. Enhancing antitumor immunity: the role of immune checkpoint inhibitors, anti-angiogenic therapy, and macrophage reprogramming. Front Oncol 2025; 15:1526407. [PMID: 40260303 PMCID: PMC12009726 DOI: 10.3389/fonc.2025.1526407] [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: 11/11/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer treatment has long been hindered by the complexity of the tumor microenvironment (TME) and the mechanisms that tumors employ to evade immune detection. Recently, the combination of immune checkpoint inhibitors (ICIs) and anti-angiogenic therapies has emerged as a promising approach to improve cancer treatment outcomes. This review delves into the role of immunostimulatory molecules and ICIs in enhancing anti-tumor immunity, while also discussing the therapeutic potential of anti-angiogenic strategies in cancer. In particular, we highlight the critical role of endoplasmic reticulum (ER) stress in angiogenesis. Moreover, we explore the potential of macrophage reprogramming to bolster anti-tumor immunity, with a focus on restoring macrophage phagocytic function, modulating hypoxic tumor environments, and targeting cytokines and chemokines that shape immune responses. By examining the underlying mechanisms of combining ICIs with anti-angiogenic therapies, we also review recent clinical trials and discuss the potential of biomarkers to guide and predict treatment efficacy.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xinying Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuxin Jiang
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hanqing Yu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Zhou Z, Zhang R, Zhou A, Lv J, Chen S, Zou H, Zhang G, Lin T, Wang Z, Zhang Y, Weng S, Han X, Liu Z. Proteomics appending a complementary dimension to precision oncotherapy. Comput Struct Biotechnol J 2024; 23:1725-1739. [PMID: 38689716 PMCID: PMC11058087 DOI: 10.1016/j.csbj.2024.04.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Recent advances in high-throughput proteomic profiling technologies have facilitated the precise quantification of numerous proteins across multiple specimens concurrently. Researchers have the opportunity to comprehensively analyze the molecular signatures in plentiful medical specimens or disease pattern cell lines. Along with advances in data analysis and integration, proteomics data could be efficiently consolidated and employed to recognize precise elementary molecular mechanisms and decode individual biomarkers, guiding the precision treatment of tumors. Herein, we review a broad array of proteomics technologies and the progress and methods for the integration of proteomics data and further discuss how to better merge proteomics in precision medicine and clinical settings.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan 450052, China
| | - Ruiqi Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Aoyang Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinxiang Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Haijiao Zou
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ting Lin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
4
|
Desigaux T, Comperat L, Dusserre N, Stachowicz ML, Lea M, Dupuy JW, Vial A, Molinari M, Fricain JC, Paris F, Oliveira H. 3D bioprinted breast cancer model reveals stroma-mediated modulation of extracellular matrix and radiosensitivity. Bioact Mater 2024; 42:316-327. [PMID: 39290339 PMCID: PMC11405629 DOI: 10.1016/j.bioactmat.2024.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Deciphering breast cancer treatment resistance remains hindered by the lack of models that can successfully capture the four-dimensional dynamics of the tumor microenvironment. Here, we show that microextrusion bioprinting can reproducibly generate distinct cancer and stromal compartments integrating cells relevant to human pathology. Our findings unveil the functional maturation of this millimeter-sized model, showcasing the development of a hypoxic cancer core and an increased surface proliferation. Maturation was also driven by the presence of cancer-associated fibroblasts (CAF) that induced elevated microvascular-like structures complexity. Such modulation was concomitant to extracellular matrix remodeling, with high levels of collagen and matricellular proteins deposition by CAF, simultaneously increasing tumor stiffness and recapitulating breast cancer fibrotic development. Importantly, our bioprinted model faithfully reproduced response to treatment, further modulated by CAF. Notably, CAF played a protective role for cancer cells against radiotherapy, facilitating increased paracrine communications. This model holds promise as a platform to decipher interactions within the microenvironment and evaluate stroma-targeted drugs in a context relevant to human pathology.
Collapse
Affiliation(s)
- Theo Desigaux
- Univ. Bordeaux, Tissue Bioengineering INSERM U1026, F-33000, Bordeaux, France
- INSERM U1026, ART BioPrint, F-33000, Bordeaux, France
| | - Leo Comperat
- Univ. Bordeaux, Tissue Bioengineering INSERM U1026, F-33000, Bordeaux, France
- INSERM U1026, ART BioPrint, F-33000, Bordeaux, France
| | - Nathalie Dusserre
- Univ. Bordeaux, Tissue Bioengineering INSERM U1026, F-33000, Bordeaux, France
- INSERM U1026, ART BioPrint, F-33000, Bordeaux, France
| | - Marie-Laure Stachowicz
- Univ. Bordeaux, Tissue Bioengineering INSERM U1026, F-33000, Bordeaux, France
- INSERM U1026, ART BioPrint, F-33000, Bordeaux, France
| | - Malou Lea
- Univ. Bordeaux, Tissue Bioengineering INSERM U1026, F-33000, Bordeaux, France
- INSERM U1026, ART BioPrint, F-33000, Bordeaux, France
| | - Jean-William Dupuy
- Univ. Bordeaux, Bordeaux Proteome, F-33000, Bordeaux, France
- Univ. Bordeaux, CNRS, INSERM, TBM-Core, US5, UAR 3427, OncoProt, F-33000, Bordeaux, France
| | - Anthony Vial
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600, Pessac, France
| | - Michael Molinari
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600, Pessac, France
| | - Jean-Christophe Fricain
- Univ. Bordeaux, Tissue Bioengineering INSERM U1026, F-33000, Bordeaux, France
- INSERM U1026, ART BioPrint, F-33000, Bordeaux, France
- Services d'Odontologie et de Santé Buccale, CHU Bordeaux, F-33000, Bordeaux, France
| | - François Paris
- CRCINA, INSERM, CNRS, Univ. Nantes, F-44000, Nantes, France
- Institut de Cancérologie de l'Ouest, F-44800, Saint Herblain, France
| | - Hugo Oliveira
- Univ. Bordeaux, Tissue Bioengineering INSERM U1026, F-33000, Bordeaux, France
- INSERM U1026, ART BioPrint, F-33000, Bordeaux, France
| |
Collapse
|
5
|
Fleischer T, Haugen MH, Ankill J, Silwal‐Pandit L, Børresen‐Dale A, Hedenfalk I, Hatschek T, Tost J, Engebraaten O, Kristensen VN. An integrated omics approach highlights how epigenetic events can explain and predict response to neoadjuvant chemotherapy and bevacizumab in breast cancer. Mol Oncol 2024; 18:2042-2059. [PMID: 38671580 PMCID: PMC11306529 DOI: 10.1002/1878-0261.13656] [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: 07/11/2023] [Revised: 02/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Treatment with the anti-angiogenic drug bevacizumab in addition to chemotherapy has shown efficacy for breast cancer in some clinical trials, but better biomarkers are needed to optimally select patients for treatment. Here, we present an omics approach where DNA methylation profiles are integrated with gene expression and results from proteomic data in breast cancer patients to predict response to therapy and pinpoint response-related epigenetic events. Fresh-frozen tumor biopsies taken before, during, and after treatment from human epidermal growth factor receptor 2 negative non-metastatic patients receiving neoadjuvant chemotherapy with or without bevacizumab were subjected to molecular profiling. Here, we report that DNA methylation at enhancer CpGs related to cell cycle regulation can predict response to chemotherapy and bevacizumab for the estrogen receptor positive subset of patients (AUC = 0.874), and we validated this observation in an independent patient cohort with a similar treatment regimen (AUC = 0.762). Combining the DNA methylation scores with the scores from a previously published protein signature resulted in a slight increase in the prediction performance (AUC = 0.784). We also show that tumors receiving the combination treatment underwent more extensive epigenetic alterations. Finally, we performed an integrative expression-methylation quantitative trait loci analysis on alterations in DNA methylation and gene expression levels, showing that the epigenetic alterations that occur during treatment are different between responders and non-responders and that these differences may be explained by the proliferation-epithelial-to-mesenchymal transition axis through the activity of grainyhead like transcription factor 2. Using tumor purity computed from copy number data, we developed a method for estimating cancer cell-specific methylation to confirm that the association to response reflects DNA methylation in cancer cells. Taken together, these results support the potential for clinical benefit of the addition of bevacizumab to chemotherapy when administered to the correct patients.
Collapse
Affiliation(s)
- Thomas Fleischer
- Department of Cancer Genetics, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Mads Haugland Haugen
- Department of Tumor Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Jørgen Ankill
- Department of Cancer Genetics, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Laxmi Silwal‐Pandit
- Department of Cancer Genetics, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Anne‐Lise Børresen‐Dale
- Department of Cancer Genetics, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences LundLund UniversityLundSweden
| | - Thomas Hatschek
- Breast Cancer CenterKarolinska University HospitalStockholmSweden
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| | - Jörg Tost
- Laboratory for Epigenetics and Environment, Centre National de Recherche en Génomique Humaine, CEA – Institut de Biologie François JacobUniversité Paris SaclayEvryFrance
| | - Olav Engebraaten
- Department of Tumor Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway
- Division of Cancer Medicine, Department of OncologyOslo University HospitalOsloNorway
| | - Vessela N. Kristensen
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway
- Department of Medical GeneticsOslo University HospitalOsloNorway
| |
Collapse
|
6
|
Hashemi M, Khosroshahi EM, Chegini MK, Asadi S, Hamyani Z, Jafari YA, Rezaei F, Eskadehi RK, Kojoori KK, Jamshidian F, Nabavi N, Alimohammadi M, Rashidi M, Mahmoodieh B, Khorrami R, Taheriazam A, Entezari M. Mechanistic insights into cisplatin response in breast tumors: Molecular determinants and drug/nanotechnology-based therapeutic opportunities. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108513. [PMID: 39216513 DOI: 10.1016/j.mrrev.2024.108513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer continues to be a major global health challenge, driving the need for effective therapeutic strategies. Cisplatin, a powerful chemotherapeutic agent, is widely used in breast cancer treatment. However, its effectiveness is often limited by systemic toxicity and the development of drug resistance. This review examines the molecular factors that influence cisplatin response and resistance, offering crucial insights for the scientific community. It highlights the significance of understanding cisplatin resistance's genetic and epigenetic contributors, which could lead to more personalized treatment approaches. Additionally, the review explores innovative strategies to counteract cisplatin resistance, including combination therapies, nanoparticle-based drug delivery systems, and targeted therapies. These approaches are under intensive investigation and promise to enhance breast cancer treatment outcomes. This comprehensive discussion is a valuable resource to advance breast cancer therapeutics and address the challenge of cisplatin resistance.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Kalhor Chegini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Hamyani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Medicine, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Yasamin Alsadat Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rezaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ramtin Khodaparast Eskadehi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kimia Kia Kojoori
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Faranak Jamshidian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Rashidi
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
7
|
Mo T, Brandal SHB, Geier OM, Engebråten O, Nilsen LB, Kristensen VN, Hole KH, Hompland T, Fleischer T, Seierstad T. MRI Assessment of Changes in Tumor Vascularization during Neoadjuvant Anti-Angiogenic Treatment in Locally Advanced Breast Cancer Patients. Cancers (Basel) 2023; 15:4662. [PMID: 37760629 PMCID: PMC10526130 DOI: 10.3390/cancers15184662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/16/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Anti-VEGF (vascular endothelial growth factor) treatment improves response rates, but not progression-free or overall survival in advanced breast cancer. It has been suggested that subgroups of patients may benefit from this treatment; however, the effects of adding anti-VEGF treatment to a standard chemotherapy regimen in breast cancer patients are not well studied. Understanding the effects of the anti-vascular treatment on tumor vasculature may provide a selection of patients that can benefit. The aim of this study was to study the vascular effect of bevacizumab using clinical dynamic contrast-enhanced MRI (DCE-MRI). A total of 70 women were randomized to receive either chemotherapy alone or chemotherapy with bevacizumab for 25 weeks. DCE-MRI was performed at baseline and at 12 and 25 weeks, and in addition 25 of 70 patients agreed to participate in an early MRI after one week. Voxel-wise pharmacokinetic analysis was performed using semi-quantitative methods and the extended Tofts model. Vascular architecture was assessed by calculating the fractal dimension of the contrast-enhanced images. Changes during treatment were compared with baseline and between the treatment groups. There was no significant difference in tumor volume at any point; however, DCE-MRI parameters revealed differences in vascular function and vessel architecture. Adding bevacizumab to chemotherapy led to a pronounced reduction in vascular DCE-MRI parameters, indicating decreased vascularity. At 12 and 25 weeks, the difference between the treatment groups is severely reduced.
Collapse
Affiliation(s)
- Torgeir Mo
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 4950 Oslo, Norway;
| | - Siri Helene Bertelsen Brandal
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Breast Diagnostic, Oslo University Hospital, 0379 Oslo, Norway
| | - Oliver Marcel Geier
- Department of Diagnostic Physics, Oslo University Hospital, 0379 Oslo, Norway;
| | - Olav Engebråten
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Oncology, Oslo University Hospital, 0379 Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 4950 Oslo, Norway
| | | | - Vessela N. Kristensen
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Knut Håkon Hole
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Oncologic Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0379 Oslo, Norway
| | - Tord Hompland
- Department of Radiation Biology, Oslo University Hospital, 4950 Oslo, Norway;
| | - Thomas Fleischer
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 4950 Oslo, Norway;
| | - Therese Seierstad
- Department of Research and Development, Division for Radiology and Nuclear Medicine, Oslo University Hospital, 0379 Oslo, Norway
| |
Collapse
|
8
|
Brogowska KK, Zajkowska M, Mroczko B. Vascular Endothelial Growth Factor Ligands and Receptors in Breast Cancer. J Clin Med 2023; 12:jcm12062412. [PMID: 36983412 PMCID: PMC10056253 DOI: 10.3390/jcm12062412] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy responsible for the largest number of deaths in women worldwide. The risk of developing BC is predisposed by many factors such as age, presence of genetic mutations or body weight. The diagnosis is mostly made relatively late, which is why patients are exposed to radical surgical treatments, long-term chemotherapy and lower survival rates. There are no sufficiently sensitive and specific screening tests; therefore, researchers are still looking for new diagnostic biomarkers that would indicate the appearance of neoplastic changes in the initial stage of neoplasm. The VEGF family of proteins (VEGF-A, VEGF-B, VEGF-C, VEGF-D, EG-VEGF, PlGF) and their receptors are significant factors in the pathogenesis of BC. They play a significant role in the process of angiogenesis and lymphangiogenesis in both physiological and pathological conditions. The usefulness of these proteins as potential diagnostic biomarkers has been initially proven. Moreover, the blockage of VEGF-related pathways seems to be a valid therapeutic target. Recent studies have tried to describe novel strategies, including targeting pericytes, use of miRNAs and extracellular tumor-associated vesicles, immunotherapeutic drugs and nanotechnology. This indicates their possible contribution to the formation of breast cancer and their usefulness as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | - Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
9
|
Margalit O, Harmsen WS, Shacham-Shmueli E, Voss MM, Boursi B, Wagner AD, Cohen R, Olswold CL, Saltz LB, Goldstein DA, Hurwitz H, Tebbutt NC, Kabbinavar FF, Adams RA, Chibaudel B, Grothey A, Yoshino T, Zalcberg J, de Gramont A, Shi Q, Lenz HJ. Evaluating sex as a predictive marker for response to bevacizumab in metastatic colorectal carcinoma: Pooled analysis of 3,369 patients in the ARCAD database. Eur J Cancer 2023; 178:162-170. [PMID: 36446161 DOI: 10.1016/j.ejca.2022.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Previous studies suggest a possible sex-specific response to bevacizumab in metastatic colorectal carcinoma (mCRC), showing a benefit in males, while the effect in females is less significant. METHODS Data from 3369 patients with mCRC enrolled on four first-line randomised trials testing chemotherapy with or without bevacizumab (2000-2007) were pooled. Association between sex and progression-free survival and overall survival (OS) was evaluated by stratified Cox regression model, adjusted for potential confounders. Predictive value was evaluated by interaction effect between sex and treatment. In a pre-planned secondary analysis, analyses were stratified using an age cut point of 60 years to evaluate the possible role of menopausal-related effects. RESULTS Bevacizumab was associated with an improved median OS in males and females, with a 2.3- and 0.6-months benefit, respectively. Stratified by age, bevacizumab resulted in improved OS in males at both age categories. In females at or above the age of 60 (n = 731), bevacizumab resulted in improved OS. However, in females below the age of 60 (n = 634), OS benefit did not reach statistical significance (adjusted hazard ratio = 0.94, 95% confidence interval 0.74-1.20). CONCLUSIONS Our results confirmed the OS benefit from the addition of bevacizumab to first-line chemotherapy in mCRC in both sexes. Among females, the benefit was less than 1 month. For females under the age of 60, there was no survival benefit. These findings could be used to relieve financial toxicity or be redistributed within healthcare systems for other health-related purposes.
Collapse
Affiliation(s)
- Ofer Margalit
- Sheba Medical Center, Ramat-Gan, Israel; Tel-Aviv University, Tel-Aviv, Israel.
| | - William S Harmsen
- Department of Quantitative Science Research, Mayo Clinic, Rochester, MN, USA
| | | | - Molly M Voss
- Department of Quantitative Science Research, Mayo Clinic, Scottsdale, AZ, USA
| | - Ben Boursi
- Sheba Medical Center, Ramat-Gan, Israel; Tel-Aviv University, Tel-Aviv, Israel
| | - Anna D Wagner
- Department of Oncology, Division of Medical Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Romain Cohen
- Department of Quantitative Science Research, Mayo Clinic, Rochester, MN, USA; Sorbonne University, Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, F-75012 Paris, France; Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, F-75012 Paris, France
| | - Curtis L Olswold
- Department of Quantitative Science Research, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Niall C Tebbutt
- University of Melbourne, Australia; Austin Health, Heidelberg, Victoria, Australia
| | - Fairooz F Kabbinavar
- David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, USA
| | | | - Benoist Chibaudel
- Department of Medical Oncology, Franco-British Institute, Levallois-Perret, France
| | | | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Japan
| | - John Zalcberg
- Department of Medical Oncology, Alfred Health and School of Public Health, Monash University, Melbourne, Australia
| | - Aimery de Gramont
- Department of Medical Oncology, Franco-British Institute, Levallois-Perret, France
| | - Qian Shi
- Department of Quantitative Science Research, Mayo Clinic, Rochester, MN, USA
| | - Heinz-Josef Lenz
- Department of Gastrointestinal Oncology, Keck School of Medicine at USC, Los Angeles, CA, USA
| |
Collapse
|
10
|
Fomchenko EI, Bayley JC, Alvarez-Breckenridge C, Rhines LD, Tatsui CE. Spinal Metastases and the Evolving Role of Molecular Targeted Therapy, Chemotherapy, and Immunotherapy. Neurospine 2022; 19:978-993. [PMID: 36597635 PMCID: PMC9816609 DOI: 10.14245/ns.2244290.145] [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: 04/07/2022] [Accepted: 07/13/2022] [Indexed: 12/27/2022] Open
Abstract
Metastatic involvement of the spine is a common complication of systemic cancer progression. Surgery and external beam radiotherapy are palliative treatment modalities aiming to preserve neurological function, control pain and maintain functional status. More recently, with development of image guidance and stereotactic delivery of high doses of conformal radiation, local tumor control has improved; however recurrent or radiation refractory disease remains a significant clinical problem with limited treatment options. This manuscript represents a narrative overview of novel targeted molecular therapies, chemotherapies, and immunotherapy treatments for patients with breast, lung, melanoma, renal cell, prostate, and thyroid cancers, which resulted in improved responses compared to standard chemotherapy. We present clinical examples of excellent responses in spinal metastatic disease which have not been specifically documented in the literature, as most clinical trials evaluate treatment response based on visceral disease. This review is useful for the spine surgeons treating patients with metastatic disease as knowledge of these responses could help with timing and planning of surgical interventions, as well as promote multidisciplinary discussions, allowing development of an individualized treatment strategy to patients presenting with widespread multifocal progressive disease, where surgery could lead to suboptimal results.
Collapse
Affiliation(s)
| | - James C. Bayley
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Claudio E. Tatsui
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA,Corresponding Author Claudio E. Tatsui Department of Neurosurgery, MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd, Houston, TX, USA
| |
Collapse
|
11
|
von der Lippe Gythfeldt H, Lien T, Tekpli X, Silwal-Pandit L, Borgen E, Garred Ø, Skjerven H, Schlichting E, Lundgren S, Wist E, Naume B, Kristensen V, Børresen-Dale AL, Lingjaerde OC, Engebraaten O. Immune phenotype of tumor microenvironment predicts response to bevacizumab in neoadjuvant treatment of ER-positive breast cancer. Int J Cancer 2020; 147:2515-2525. [PMID: 32488909 DOI: 10.1002/ijc.33108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/02/2020] [Accepted: 04/30/2020] [Indexed: 12/24/2022]
Abstract
Antiangiogenic drugs are potentially a useful supplement to neoadjuvant chemotherapy for a subgroup of patients with human epidermal growth factor receptor 2 (HER2) negative breast cancer, but reliable biomarkers for improved response are lacking. Here, we report on a randomized phase II clinical trial to study the added effect of bevacizumab in neoadjuvant chemotherapy with FEC100 (5-fluorouracil, epirubicin and cyclophosphamide) and taxanes (n = 132 patients). Gene expression from the tumors was obtained before neoadjuvant treatment, and treatment response was evaluated by residual cancer burden (RCB) at time of surgery. Bevacizumab increased the proportion of complete responders (RCB class 0) from 5% to 20% among patients with estrogen receptor (ER) positive tumors (P = .02). Treatment with bevacizumab was associated with improved 8-year disease-free survival (P = .03) among the good responders (RCB class 0 or I). Patients treated with paclitaxel (n = 45) responded better than those treated with docetaxel (n = 21; P = .03). Improved treatment response was associated with higher proliferation rate and an immune phenotype characterized by high presence of classically activated M1 macrophages, activated NK cells and memory activated CD4 T cells. Treatment with bevacizumab increased the number of adverse events, including hemorrhage, hypertension, infection and febrile neutropenia, but despite this, the ECOG status was not affected.
Collapse
Affiliation(s)
- Hedda von der Lippe Gythfeldt
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Tonje Lien
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Xavier Tekpli
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Laxmi Silwal-Pandit
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Helle Skjerven
- Department of Breast and Endocrine Surgery, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Ellen Schlichting
- Department of Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway
| | - Steinar Lundgren
- Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Erik Wist
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Naume
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vessela Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Christian Lingjaerde
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Informatics, University of Oslo, Oslo, Norway.,KG Jebsen Centre for B-Cell Malignancies, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav Engebraaten
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Guo F, Ji G, Li Q, Yang Y, Shui L, Shen Y, Yang H. Bacterial particles retard tumor growth as a novel vascular disrupting agent. Biomed Pharmacother 2020; 122:109757. [DOI: 10.1016/j.biopha.2019.109757] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/07/2019] [Accepted: 11/29/2019] [Indexed: 02/08/2023] Open
|
13
|
Toxicity of locoregional radiotherapy in combination with bevacizumab in patients with non-metastatic breast cancer (TOLERAB): Final long-term evaluation. PLoS One 2019; 14:e0221816. [PMID: 31469859 PMCID: PMC6716668 DOI: 10.1371/journal.pone.0221816] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/15/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Few data are available concerning the safety of bevacizumab (B) in combination with locoregional radiation therapy (RT). The objective of this study was to evaluate the 5-year late toxicity of concurrent B and RT in non-metastatic breast cancer. MATERIALS AND METHODS This multicentre prospective study included non-metastatic breast cancer patients enrolled in phase 3 clinical trials evaluating B with concurrent RT versus RT alone. All patients received neoadjuvant or adjuvant chemotherapy and normofractionated breast or chest wall RT, with or without regional lymph node RT. B was administered at an equivalent dose of 5 mg/kg once a week for 1 year. The safety profile was evaluated 1, 3 and 5 years after completion of radiotherapy. RESULTS A total of 64 patients were included between November 2007 and April 2010. Median follow-up was 60 months (12-73) and 5-year late toxicity data were available for 46 patients. The majority of tumours were triple-negative (68.8%), tumour size <2cm (41.3%) with negative nodal status (50.8%). Median total dose of B was 15,000mg and median duration was 11.2 months. No grade ≥3 toxicity was observed. Only 8 patients experienced grade 1-2 toxicities: n = 3 (6.5%) grade 1 lymphedema, n = 2 (4.3%) grade 1 pain, n = 1 (2.2%) grade 2 lymphedema, n = 1 (2.2%) grade 1 fibrosis. Five-year overall survival was 93.8%, disease-free survival was 89% and locoregional recurrence-free survival was 93.1%. CONCLUSION Concurrent B and locoregional RT are associated with acceptable 5-year toxicity in patients with non-metastatic breast cancer. No grade ≥3 toxicity was observed.
Collapse
|
14
|
Márquez-Garbán DC, Gorrín-Rivas M, Chen HW, Sterling C, Elashoff D, Hamilton N, Pietras RJ. Squalamine blocks tumor-associated angiogenesis and growth of human breast cancer cells with or without HER-2/neu overexpression. Cancer Lett 2019; 449:66-75. [PMID: 30771431 DOI: 10.1016/j.canlet.2019.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/07/2019] [Accepted: 02/10/2019] [Indexed: 12/22/2022]
Abstract
Angiogenesis is critical for breast cancer progression. Overexpression of HER-2/neu receptors occur in 25-30% of breast cancers, and treatment with trastuzumab inhibits HER-2-overexpressing tumor growth. Notably, HER-2-mediated signaling enhances vascular endothelial growth factor (VEGF) secretion to increase tumor-associated angiogenesis. Squalamine (aminosterol compound) suppresses VEGF-induced activation of kinases in vascular endothelial cells and inhibits tumor-associated angiogenesis. We assessed antitumor effects of squalamine either alone or with trastuzumab in nude mice bearing breast tumor xenografts without (MCF-7) or with HER2-overexpression (MCF-7/HER-2). Squalamine alone inhibited progression of MCF-7 tumors lacking HER2 overexpression, and squalamine combined with trastuzumab elicited marked inhibition of MCF-7/HER2 growth exceeding that of trastuzumab alone. MCF-7/HER-2 cells secrete higher levels of VEGF than MCF-7 cells, but squalamine elicited no growth inhibition of either MCF-7/HER-2 or MCF-7 cells in vitro. However, squalamine did stop growth of human umbilical vein endothelial cells (HUVECs) and reduced VEGF-induced endothelial tube-like formations in vitro. These effects correlated with blockade of focal adhesion kinase phosphorylation and stress fiber assembly in HUVECs. Thus, squalamine effectively inhibits growth of breast cancers with or without HER-2-overexpression, an effect due in part to blockade of tumor-associated angiogenesis.
Collapse
Affiliation(s)
- Diana C Márquez-Garbán
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA.
| | - Manuel Gorrín-Rivas
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA.
| | - Hsiao-Wang Chen
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA.
| | - Colin Sterling
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA
| | - David Elashoff
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA; Department of Medicine, Division of General Internal Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA.
| | - Nalo Hamilton
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA; UCLA School of Nursing, Los Angeles, CA, 90095, USA.
| | - Richard J Pietras
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA.
| |
Collapse
|
15
|
Systemic Treatment of HER2-Negative Metastatic Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
Sen F, Aydiner A. Endocrine Therapy of Metastatic Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
17
|
Wan G, Cao F, Wang X, Sun X. Bevacizumab Added to Neoadjuvant Chemotherapy in HER2-Negative Non-Metastatic Breast Cancer. J Cancer 2019; 10:416-417. [PMID: 30719135 PMCID: PMC6360292 DOI: 10.7150/jca.29461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 11/01/2018] [Indexed: 12/02/2022] Open
Affiliation(s)
- Guoxing Wan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Fengjun Cao
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xuanbin Wang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xue Sun
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| |
Collapse
|
18
|
Symonds L, Linden H, Gadi V, Korde L, Rodler E, Gralow J, Redman M, Baker K, Wu QV, Jenkins I, Kurland B, Garrison M, Smith J, Anderson J, Van Haelst C, Specht J. Combined Targeted Therapies for First-line Treatment of Metastatic Triple Negative Breast Cancer-A Phase II Trial of Weekly Nab-Paclitaxel and Bevacizumab Followed by Maintenance Targeted Therapy With Bevacizumab and Erlotinib. Clin Breast Cancer 2018; 19:e283-e296. [PMID: 30737173 DOI: 10.1016/j.clbc.2018.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/04/2018] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Angiogenesis and epidermal growth factor receptor signaling are potential therapeutic targets in triple negative breast cancer (TNBC). We hypothesized that targeting these critical pathways would prolong progression-free survival with first-line therapy for metastatic TNBC. PATIENTS AND METHODS We conducted a phase II trial of nab-paclitaxel and bevacizumab, followed by maintenance therapy with bevacizumab and erlotinib, for patients with metastatic TNBC. During induction, the patients received nab-paclitaxel 100 mg/m2 intravenously (days 1, 8, and 15) and bevacizumab 10 mg/kg intravenously (days 1 and 15) every 28 days for 6 cycles. Patients free of progression at 24 weeks received maintenance therapy with bevacizumab 10 mg/kg intravenously every 2 weeks and oral erlotinib 150 mg/d until disease progression. The primary endpoint was progression-free survival (PFS). The secondary endpoints were best overall response, overall survival (OS), and adverse events. We explored the measurement of circulating tumor cells as a prognostic marker. RESULTS A total of 55 evaluable patients were enrolled. The median PFS and OS for the cohort was 9.1 months (95% confidence interval, 7.2-11.1) and 18.1 months (95% confidence interval, 15.6-21.7), respectively. Of the 53 patients with measurable disease, 39 (74%) had experienced a partial response and 10 (19%) had stable disease using the Response Evaluation Criteria In Solid Tumors. The most common toxicities were uncomplicated neutropenia, fatigue, and neuropathy. Decreased circulating tumor cells from baseline to the first assessment correlated with longer PFS and OS. CONCLUSION Nab-paclitaxel and bevacizumab, followed by maintenance targeted therapy with bevacizumab and erlotinib, resulted in PFS similar to that of other trials. Most patients experienced a partial response (74%). Most patients received maintenance therapy (55%), providing a break from cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Lynn Symonds
- Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Hannah Linden
- Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Vijayakrishna Gadi
- Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Eve Rodler
- Division of Oncology and Hematology, Department of Internal Medicine, UC Davis Health, Sacramento, CA
| | - Julie Gralow
- Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Mary Redman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Kelsey Baker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Quan Vicky Wu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Isaac Jenkins
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | - Julie Smith
- Confluence Health at Wenatchee Valley, Wenatchee, WA
| | | | - Carol Van Haelst
- Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Jennifer Specht
- Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA.
| |
Collapse
|
19
|
Basho RK, Yam C, Gilcrease M, Murthy RK, Helgason T, Karp DD, Meric-Bernstam F, Hess KR, Valero V, Albarracin C, Litton JK, Chavez-MacGregor M, Hong D, Kurzrock R, Hortobagyi GN, Janku F, Moulder SL. Comparative Effectiveness of an mTOR-Based Systemic Therapy Regimen in Advanced, Metaplastic and Nonmetaplastic Triple-Negative Breast Cancer. Oncologist 2018; 23:1300-1309. [PMID: 30139837 DOI: 10.1634/theoncologist.2017-0498] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 06/25/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a heterogeneous disease with subtypes having different "targetable" molecular aberrations. Metaplastic breast cancers (MpBCs) are typically TNBCs and commonly have alterations in the PI3K/Akt/mTOR pathway. We previously reported efficacy for an mTOR-based chemotherapy regimen in MpBC. To determine if tumor subtype influences prognosis, we compared treatment outcomes of patients with MpBC with those of patients with nonmetaplastic TNBC receiving an mTOR-based systemic therapy regimen. PATIENTS AND METHODS Patients with advanced MpBC and nonmetaplastic TNBC were treated at our institution from April 16, 2009, through November 4, 2014, using mTOR inhibition (temsirolimus or everolimus) with liposomal doxorubicin and bevacizumab (DAT/DAE). Median progression-free survival (PFS) and overall survival (OS) were estimated by the Kaplan-Meier method. Cox regression analyses were used to evaluate associations between tumor histology and outcomes. Multivariable models were adjusted for all covariates. RESULTS Fourteen patients with nonmetaplastic TNBC and 59 patients with advanced MpBC were treated with DAT/DAE. MpBC patients were older (p = .002) and less likely to have a history of bevacizumab use (p = .023). Median PFS for the nonmetaplastic TNBC and MpBC patients was 2.5 months and 4.8 months, respectively. This difference in PFS was statistically significant on univariable (p = .006) but not multivariable analysis (p = .087). Median OS for the nonmetaplastic TNBC and MpBC patients was 3.7 months and 10.0 months, respectively (p = .0003). MpBC remained significantly associated with improved OS on multivariable analysis (p < .0001). CONCLUSION In our study, DAT/DAE appeared to be more effective in MpBC compared with nonmetaplastic TNBC. These data support patient selection for targeted therapy in TNBC. IMPLICATIONS FOR PRACTICE Metaplastic breast cancers (MpBCs) represent <1% of all breast cancers, demonstrate mesenchymal differentiation, and are typically resistant to chemotherapy. Patients with advanced MpBC treated with an mTOR-based systemic therapy regimen had better long-term outcomes compared with patients with nonmetaplastic triple-negative breast cancer treated with the same regimen, suggesting that metaplastic histology may predict benefit from agents targeting the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Reva K Basho
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Clinton Yam
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael Gilcrease
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rashmi K Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Thorunn Helgason
- Department of Investigational Cancer Therapeutics (Phase I Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (Phase I Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kenneth R Hess
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Constance Albarracin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mariana Chavez-MacGregor
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David Hong
- Department of Investigational Cancer Therapeutics (Phase I Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Razelle Kurzrock
- Department of Investigational Cancer Therapeutics (Phase I Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Hematology and Oncology, The University of California San Diego Moores Cancer Center, San Diego, California, USA
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stacy L Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Investigational Cancer Therapeutics (Phase I Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
20
|
Malo CS, Khadka RH, Ayasoufi K, Jin F, AbouChehade JE, Hansen MJ, Iezzi R, Pavelko KD, Johnson AJ. Immunomodulation Mediated by Anti-angiogenic Therapy Improves CD8 T Cell Immunity Against Experimental Glioma. Front Oncol 2018; 8:320. [PMID: 30211113 PMCID: PMC6124655 DOI: 10.3389/fonc.2018.00320] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/26/2018] [Indexed: 01/13/2023] Open
Abstract
Glioblastoma (GBM) is a lethal cancer of the central nervous system with a median survival rate of 15 months with treatment. Thus, there is a critical need to develop novel therapies for GBM. Immunotherapy is emerging as a promising therapeutic strategy. However, current therapies for GBM, in particular anti-angiogenic therapies that block vascular endothelial growth factor (VEGF), may have undefined consequences on the efficacy of immunotherapy. While this treatment is primarily prescribed to reduce tumor vascularization, multiple immune cell types also express VEGF receptors, including the most potent antigen-presenting cell, the dendritic cell (DC). Therefore, we assessed the role of anti-VEGF therapy in modifying DC function. We found that VEGF blockade results in a more mature DC phenotype in the brain, as demonstrated by an increase in the expression of the co-stimulatory molecules B7-1, B7-2, and MHC II. Furthermore, we observed reduced levels of the exhaustion markers PD-1 and Tim-3 on brain-infiltrating CD8 T cells, indicating improved functionality. Thus, anti-angiogenic therapy has the potential to be used in conjunction with and enhance immunotherapy for GBM.
Collapse
Affiliation(s)
- Courtney S Malo
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Roman H Khadka
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | | | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Michael J Hansen
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Raymond Iezzi
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Department of Neurology, Mayo Clinic, Rochester, MN, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
21
|
Harris AR, Perez MJ, Munson JM. Docetaxel facilitates lymphatic-tumor crosstalk to promote lymphangiogenesis and cancer progression. BMC Cancer 2018; 18:718. [PMID: 29976154 PMCID: PMC6034223 DOI: 10.1186/s12885-018-4619-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 06/20/2018] [Indexed: 12/20/2022] Open
Abstract
Background Infiltration into lymphatic vessels is a critical step in breast cancer metastasis. Lymphatics undergo changes that facilitate metastasis as a result of activation of the cells lining lymphatic vessels, lymphatic endothelial cells (LECs). Inhibition of activation by targeting VEGFR3 can reduce invasion toward lymphatics. To best benefit patients, this approach should be coupled with standard of care that slows tumor growth, such as chemotherapy. Little is known about how chemotherapies, like docetaxel, may influence lymphatics and conversely, how lymphatics can alter responses to therapy. Methods A novel 3D in vitro co-culture model of the human breast tumor microenvironment was employed to examine the contribution of LECs to tumor invasion and viability with docetaxel and anti-VEGFR3, using three cell lines, MDA-MB-231, HCC38, and HCC1806. In vivo, the 4T1 mouse model of breast carcinoma was used to examine the efficacy of combinatorial therapy with docetaxel and anti-VEGFR3 on lymph node metastasis and tumor growth. Lymphangiogenesis in these mice was analyzed by immunohistochemistry and flow cytometry. Luminex analysis was used to measure expression of lymphangiogenic cytokines. Results In vitro, tumor cell invasion significantly increased with docetaxel when LECs were present; this effect was attenuated by inhibition of VEGFR3. LECs reduced docetaxel-induced cell death independent of VEGFR3. In vivo, docetaxel significantly increased breast cancer metastasis to the lymph node. Docetaxel and anti-VEGFR3 combination therapy reduced lymph node and lung metastasis in 4T1 and synergized to reduce tumor growth. Docetaxel induced VEGFR3-dependent vessel enlargement, lymphangiogenesis, and expansion of the LEC population in the peritumoral microenvironment, but not tumor-free stroma. Docetaxel caused an upregulation in pro-lymphangiogenic factors including VEGFC and TNF-α in the tumor microenvironment in vivo. Conclusions Here we present a counter-therapeutic effect of docetaxel chemotherapy that triggers cancer cells to elicit lymphangiogenesis. In turn, lymphatics reduce cancer response to docetaxel by altering the cytokine milieu in breast cancer. These changes lead to an increase in tumor cell invasion and survival under docetaxel treatment, ultimately reducing docetaxel efficacy. These docetaxel-induced effects can be mitigated by anti-VEGFR3 therapy, resulting in a synergism between these treatments that reduces tumor growth and metastasis. Electronic supplementary material The online version of this article (10.1186/s12885-018-4619-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexandra R Harris
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Matthew J Perez
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jennifer M Munson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA. .,Department of Biomedical Engineering & Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Polytechnic Institute & State University, Blacksburg, VA, 24061, USA.
| |
Collapse
|
22
|
Lodola F, Laforenza U, Cattaneo F, Ruffinatti FA, Poletto V, Massa M, Tancredi R, Zuccolo E, Khdar DA, Riccardi A, Biggiogera M, Rosti V, Guerra G, Moccia F. VEGF-induced intracellular Ca 2+ oscillations are down-regulated and do not stimulate angiogenesis in breast cancer-derived endothelial colony forming cells. Oncotarget 2017; 8:95223-95246. [PMID: 29221123 PMCID: PMC5707017 DOI: 10.18632/oncotarget.20255] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023] Open
Abstract
Endothelial colony forming cells (ECFCs) represent a population of truly endothelial precursors that promote the angiogenic switch in solid tumors, such as breast cancer (BC). The intracellular Ca2+ toolkit, which drives the pro-angiogenic response to VEGF, is remodelled in tumor-associated ECFCs such that they are seemingly insensitive to this growth factor. This feature could underlie the relative failure of anti-VEGF therapies in cancer patients. Herein, we investigated whether and how VEGF uses Ca2+ signalling to control angiogenesis in BC-derived ECFCs (BC-ECFCs). Although VEGFR-2 was normally expressed, VEGF failed to induce proliferation and in vitro tubulogenesis in BC-ECFCs. Likewise, VEGF did not trigger robust Ca2+ oscillations in these cells. Similar to normal cells, VEGF-induced intracellular Ca2+ oscillations were triggered by inositol-1,4,5-trisphosphate-dependent Ca2+ release from the endoplasmic reticulum (ER) and maintained by store-operated Ca2+ entry (SOCE). However, InsP3-dependent Ca2+ release was significantly lower in BC-ECFCs due to the down-regulation of ER Ca2+ levels, while there was no remarkable difference in the amplitude, pharmacological profile and molecular composition of SOCE. Thus, the attenuation of the pro-angiogenic Ca2+ response to VEGF was seemingly due to the reduction in ER Ca2+ concentration, which prevents VEGF from triggering robust intracellular Ca2+ oscillations. However, the pharmacological inhibition of SOCE prevented BC-ECFC proliferation and in vitro tubulogenesis. These findings demonstrate for the first time that BC-ECFCs are insensitive to VEGF, which might explain at cellular and molecular levels the failure of anti-VEGF therapies in BC patients, and hint at SOCE as a novel molecular target for this disease.
Collapse
Affiliation(s)
- Francesco Lodola
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy.,Current address: Italian Institute of Technology, Center for Nano Science and Technology, Milano 20133, Italy
| | - Umberto Laforenza
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy
| | | | - Valentina Poletto
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Foundation IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Margherita Massa
- Laboratory of Immunology Transplantation, Foundation IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Richard Tancredi
- Medical Oncology Unit, Foundation IRCCS Salvatore Maugeri, Pavia 27100, Italy
| | - Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| | - Dlzar Alì Khdar
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| | - Alberto Riccardi
- Medical Oncology Unit, Foundation IRCCS Salvatore Maugeri, Pavia 27100, Italy.,Department of Internal Medicine, University of Pavia, Pavia 27100, Italy
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| | - Vittorio Rosti
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Foundation IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso 86100, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia 27100, Italy
| |
Collapse
|
23
|
Abu-Jamous B, Buffa FM, Harris AL, Nandi AK. In vitro downregulated hypoxia transcriptome is associated with poor prognosis in breast cancer. Mol Cancer 2017; 16:105. [PMID: 28619028 PMCID: PMC5472949 DOI: 10.1186/s12943-017-0673-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hypoxia is a characteristic of breast tumours indicating poor prognosis. Based on the assumption that those genes which are up-regulated under hypoxia in cell-lines are expected to be predictors of poor prognosis in clinical data, many signatures of poor prognosis were identified. However, it was observed that cell line data do not always concur with clinical data, and therefore conclusions from cell line analysis should be considered with caution. As many transcriptomic cell-line datasets from hypoxia related contexts are available, integrative approaches which investigate these datasets collectively, while not ignoring clinical data, are required. RESULTS We analyse sixteen heterogeneous breast cancer cell-line transcriptomic datasets in hypoxia-related conditions collectively by employing the unique capabilities of the method, UNCLES, which integrates clustering results from multiple datasets and can address questions that cannot be answered by existing methods. This has been demonstrated by comparison with the state-of-the-art iCluster method. From this collection of genome-wide datasets include 15,588 genes, UNCLES identified a relatively high number of genes (>1000 overall) which are consistently co-regulated over all of the datasets, and some of which are still poorly understood and represent new potential HIF targets, such as RSBN1 and KIAA0195. Two main, anti-correlated, clusters were identified; the first is enriched with MYC targets participating in growth and proliferation, while the other is enriched with HIF targets directly participating in the hypoxia response. Surprisingly, in six clinical datasets, some sub-clusters of growth genes are found consistently positively correlated with hypoxia response genes, unlike the observation in cell lines. Moreover, the ability to predict bad prognosis by a combined signature of one sub-cluster of growth genes and one sub-cluster of hypoxia-induced genes appears to be comparable and perhaps greater than that of known hypoxia signatures. CONCLUSIONS We present a clustering approach suitable to integrate data from diverse experimental set-ups. Its application to breast cancer cell line datasets reveals new hypoxia-regulated signatures of genes which behave differently when in vitro (cell-line) data is compared with in vivo (clinical) data, and are of a prognostic value comparable or exceeding the state-of-the-art hypoxia signatures.
Collapse
Affiliation(s)
- Basel Abu-Jamous
- Department of Electronic and Computer Engineering, Brunel University London, Uxbridge, Middlesex, UB8 3PH UK
- Department of Plant Sciences, University of Oxford, Oxford, OX1 3RB UK
| | - Francesca M. Buffa
- Cancer Research UK, Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS UK
| | - Adrian L. Harris
- Cancer Research UK, Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS UK
| | - Asoke K. Nandi
- Department of Electronic and Computer Engineering, Brunel University London, Uxbridge, Middlesex, UB8 3PH UK
- The Key Laboratory of Embedded Systems and Service Computing, College of Electronic and Information Engineering, Tongji University, Shanghai, Peoples, Republic of China
| |
Collapse
|
24
|
Gamucci T, Mentuccia L, Natoli C, Sperduti I, Cassano A, Michelotti A, Di Lauro L, Sergi D, Fabi A, Sarobba MG, Marchetti P, Barba M, Magnolfi E, Maugeri‐Saccà M, Rossi E, Sini V, Grassadonia A, Pellegrini D, Astone A, Nisticò C, Angelini F, Vaccaro A, Pellegrino A, De Angelis C, Palleschi M, Moscetti L, Bertolini I, Buglioni S, Giordano A, Pizzuti L, Vici P. A Real-World Multicentre Retrospective Study of Paclitaxel-Bevacizumab and Maintenance Therapy as First-Line for HER2-Negative Metastatic Breast Cancer. J Cell Physiol 2017; 232:1571-1578. [PMID: 27861874 PMCID: PMC6220933 DOI: 10.1002/jcp.25685] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 11/10/2016] [Indexed: 01/04/2023]
Abstract
Bevacizumab in combination with taxanes in HER2-negative metastatic breast cancer (MBC) patients has shown improved progression-free survival (PFS), despite the lack of clear overall survival (OS) benefit. We performed a retrospective analysis to evaluate the impact of paclitaxel-bevacizumab and of maintenance therapy with bevacizumab (BM) and endocrine therapy (ET) in the real-world practice. We identified 314 HER2-negative MBC patients treated in 12 cancer centers. Overall, the median PFS and OS were 14 and 40 months, respectively. Among the 254 patients potentially eligible for BM, 183 received BM after paclitaxel discontinuation until progression/toxicity. PFS and OS were improved in patients who had received BM in comparison with those potentially eligible but who did not receive BM (P< 0.0001 and P = 0.001, respectively). Results were confirmed when adjusting for propensity score. Among the 216 hormone-receptor positive patients eligible for BM, a more favorable PFS and OS were observed when maintenance ET was administered (P < 0.0001). Multivariate analysis showed that PS, BM, number of disease sites and maintenance ET were related to PFS, while response and maintenance ET were related to OS. In hormone-receptor positive patients, BM produced a significant PFS and a trend towards OS benefit only in absence of maintenance ET (P = 0.0007 and P = 0.06, respectively). In the triple-negative subgroup, we observed a trend towards a better OS for patients who received BM (P = 0.06), without differences in PFS (P = 0.21). Our results confirmed the efficacy of first-line paclitaxel-bevacizumab in real-world practice; both BM and maintenance ET significantly improved PFS and OS compared to no maintenance therapies. J. Cell. Physiol. 232: 1571-1578, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Clara Natoli
- Department of Medical, Oral and Biotechnological SciencesCentro Scienze dell’ Invecchiamento e Medicina Traslazionale ‐ CeSI‐MeTChietiItaly
| | - Isabella Sperduti
- Bio‐Statistics Unit, Regina Elena National Cancer InstituteRomeItaly
| | - Alessandra Cassano
- Department of Medical OncologyPoliclinico Universitario A. GemelliRomeItaly
| | | | - Luigi Di Lauro
- Division of Medical Oncology 2Regina Elena National Cancer InstituteRomeItaly
| | - Domenico Sergi
- Division of Medical Oncology 2Regina Elena National Cancer InstituteRomeItaly
| | - Alessandra Fabi
- Division of Medical Oncology 1Regina Elena National Cancer InstituteRomeItaly
| | | | - Paolo Marchetti
- Department of Clinical and Molecular Medicine“Sapienza” University of RomeAzienda Ospedaliera Sant'AndreaRomeItaly
| | - Maddalena Barba
- Division of Medical Oncology 2Regina Elena National Cancer InstituteRomeItaly
- Scientific DirectionRegina Elena National Cancer InstituteRomeItaly
| | | | - Marcello Maugeri‐Saccà
- Division of Medical Oncology 2Regina Elena National Cancer InstituteRomeItaly
- Scientific DirectionRegina Elena National Cancer InstituteRomeItaly
| | - Ernesto Rossi
- Department of Medical OncologyPoliclinico Universitario A. GemelliRomeItaly
| | - Valentina Sini
- Department of Clinical and Molecular Medicine“Sapienza” University of RomeAzienda Ospedaliera Sant'AndreaRomeItaly
| | - Antonino Grassadonia
- Department of Medical, Oral and Biotechnological SciencesCentro Scienze dell’ Invecchiamento e Medicina Traslazionale ‐ CeSI‐MeTChietiItaly
| | - Domenica Pellegrini
- Division of Medical Oncology 1Regina Elena National Cancer InstituteRomeItaly
| | - Antonino Astone
- Department of Medical OncologyPoliclinico Universitario A. GemelliRomeItaly
| | - Cecilia Nisticò
- Division of Medical Oncology 1Regina Elena National Cancer InstituteRomeItaly
| | - Franco Angelini
- Medical Oncology UnitRegina Apostolorum HospitalAlbano, RomeItaly
| | | | | | | | | | - Luca Moscetti
- Medical Oncology Unit, Belcolle HospitalViterboItaly
- Department of Medical and Surgical Sciences for Children and AdultsAzienda Ospedaliero‐Universitaria Policlinico di ModenaModenaItaly
| | - Ilaria Bertolini
- Oncology Unit IAzienda Ospedaliera Universitaria PisanaPisaItaly
| | | | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for BiotechnologyCollege of Science and TechnologyTemple UniversityPhiladelphiaPennsylvania
| | - Laura Pizzuti
- Division of Medical Oncology 2Regina Elena National Cancer InstituteRomeItaly
| | - Patrizia Vici
- Division of Medical Oncology 2Regina Elena National Cancer InstituteRomeItaly
| |
Collapse
|
25
|
Silwal-Pandit L, Nord S, von der Lippe Gythfeldt H, Møller EK, Fleischer T, Rødland E, Krohn M, Borgen E, Garred Ø, Olsen T, Vu P, Skjerven H, Fangberget A, Holmen MM, Schlitchting E, Wille E, Nordberg Stokke M, Moen Vollan HK, Kristensen V, Langerød A, Lundgren S, Wist E, Naume B, Lingjærde OC, Børresen-Dale AL, Engebraaten O. The Longitudinal Transcriptional Response to Neoadjuvant Chemotherapy with and without Bevacizumab in Breast Cancer. Clin Cancer Res 2017; 23:4662-4670. [PMID: 28487444 DOI: 10.1158/1078-0432.ccr-17-0160] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/31/2017] [Accepted: 05/03/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Chemotherapy-induced alterations to gene expression are due to transcriptional reprogramming of tumor cells or subclonal adaptations to treatment. The effect on whole-transcriptome mRNA expression was investigated in a randomized phase II clinical trial to assess the effect of neoadjuvant chemotherapy with the addition of bevacizumab.Experimental Design: Tumor biopsies and whole-transcriptome mRNA profiles were obtained at three fixed time points with 66 patients in each arm. Altogether, 358 specimens from 132 patients were available, representing the transcriptional state before treatment start, at 12 weeks and after treatment (25 weeks). Pathologic complete response (pCR) in breast and axillary nodes was the primary endpoint.Results: pCR was observed in 15 patients (23%) receiving bevacizumab and chemotherapy and 8 patients (12%) receiving only chemotherapy. In the estrogen receptor-positive patients, 11 of 54 (20%) treated with bevacizumab and chemotherapy achieved pCR, while only 3 of 57 (5%) treated with chemotherapy reached pCR. In patients with estrogen receptor-positive tumors treated with combination therapy, an elevated immune activity was associated with good response. Proliferation was reduced after treatment in both treatment arms and most pronounced in the combination therapy arm, where the reduction in proliferation accelerated during treatment. Transcriptional alterations during therapy were subtype specific, and the effect of adding bevacizumab was most evident for luminal-B tumors.Conclusions: Clinical response and gene expression response differed between patients receiving combination therapy and chemotherapy alone. The results may guide identification of patients likely to benefit from antiangiogenic therapy. Clin Cancer Res; 23(16); 4662-70. ©2017 AACR.
Collapse
Affiliation(s)
- Laxmi Silwal-Pandit
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Silje Nord
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Hedda von der Lippe Gythfeldt
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Elen K Møller
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Thomas Fleischer
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Einar Rødland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Marit Krohn
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Tone Olsen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Phuong Vu
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Helle Skjerven
- Department of Research, Vestre Viken Hospital Trust, Drammen, Norway
| | - Anne Fangberget
- Department of Radiology and Nuclear Medicine, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Marit M Holmen
- Department of Radiology and Nuclear Medicine, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Ellen Schlitchting
- Department of Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Wille
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | | | - Hans Kristian Moen Vollan
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Vessela Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Anita Langerød
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Steinar Lundgren
- Department of Oncology, St. Olavs University Hospital, Trondheim, Norway.,Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Erik Wist
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Insitute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Naume
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Insitute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Christian Lingjærde
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Department of Computer Science, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Insitute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav Engebraaten
- Department of Oncology, Oslo University Hospital, Oslo, Norway. .,Insitute for Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
26
|
Zambonin V, De Toma A, Carbognin L, Nortilli R, Fiorio E, Parolin V, Pilotto S, Cuppone F, Pellini F, Lombardi D, Pollini GP, Tortora G, Bria E. Clinical results of randomized trials and 'real-world' data exploring the impact of Bevacizumab for breast cancer: opportunities for clinical practice and perspectives for research. Expert Opin Biol Ther 2017; 17:497-506. [PMID: 28133971 DOI: 10.1080/14712598.2017.1289171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Angiogenesis plays a fundamental role in breast cancer (BC) growth, progression and metastatic spread. After the promising introduction of bevacizumab for the treatment of advanced BC, the initial enthusiasm decreased when the FDA withdrew its approval in 2011. Nevertheless, several clinical studies exploring the role of bevacizumab have been subsequently published. Areas covered: The aim of this study is to review the available clinical trials exploring the potential effectiveness of bevacizumab in BC, regardless of the disease setting. Expert opinion: Even if the evidence suggests that bevacizumab must be ruled out from the HER2-positive and adjuvant setting, bevacizumab's benefit remains uncertain in the neoadjuvant setting and in the advanced treatment of HER2-negative patients. In the first setting, the addition of bevacizumab to chemotherapy increased the pathological complete response (pCR) rate in most clinical trials. However, the current absence of evidence that pCR is a trial-level surrogate for survival requires waiting for long-term results. In the advanced setting, all trials showed a benefit in progression-free survival, but not in overall survival, highlighting an increase of adverse events. The lack of predictors of response represents the main unmet need in which future clinical research will undoubtedly invest.
Collapse
Affiliation(s)
- Valentina Zambonin
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Alessandro De Toma
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Luisa Carbognin
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Rolando Nortilli
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Elena Fiorio
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Veronica Parolin
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Sara Pilotto
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | | | - Francesca Pellini
- b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,d Chirurgia Senologica, Azienda Ospedaliera Universitaria Integrata, A.O.U.I. Breast Surgery Verona , Verona , Italy
| | - Davide Lombardi
- b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,d Chirurgia Senologica, Azienda Ospedaliera Universitaria Integrata, A.O.U.I. Breast Surgery Verona , Verona , Italy
| | - Giovanni Paolo Pollini
- b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,d Chirurgia Senologica, Azienda Ospedaliera Universitaria Integrata, A.O.U.I. Breast Surgery Verona , Verona , Italy
| | - Giampaolo Tortora
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Emilio Bria
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| |
Collapse
|
27
|
da Motta LL, Ledaki I, Purshouse K, Haider S, De Bastiani MA, Baban D, Morotti M, Steers G, Wigfield S, Bridges E, Li JL, Knapp S, Ebner D, Klamt F, Harris AL, McIntyre A. The BET inhibitor JQ1 selectively impairs tumour response to hypoxia and downregulates CA9 and angiogenesis in triple negative breast cancer. Oncogene 2017; 36:122-132. [PMID: 27292261 PMCID: PMC5061082 DOI: 10.1038/onc.2016.184] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 03/22/2016] [Accepted: 04/15/2016] [Indexed: 12/16/2022]
Abstract
The availability of bromodomain and extra-terminal inhibitors (BETi) has enabled translational epigenetic studies in cancer. BET proteins regulate transcription by selectively recognizing acetylated lysine residues on chromatin. BETi compete with this process leading to both downregulation and upregulation of gene expression. Hypoxia enables progression of triple negative breast cancer (TNBC), the most aggressive form of breast cancer, partly by driving metabolic adaptation, angiogenesis and metastasis through upregulation of hypoxia-regulated genes (for example, carbonic anhydrase 9 (CA9) and vascular endothelial growth factor A (VEGF-A). Responses to hypoxia can be mediated epigenetically, thus we investigated whether BETi JQ1 could impair the TNBC response induced by hypoxia and exert anti-tumour effects. JQ1 significantly modulated 44% of hypoxia-induced genes, of which two-thirds were downregulated including CA9 and VEGF-A. JQ1 prevented HIF binding to the hypoxia response element in CA9 promoter, but did not alter HIF expression or activity, suggesting some HIF targets are BET-dependent. JQ1 reduced TNBC growth in vitro and in vivo and inhibited xenograft vascularization. These findings identify that BETi dually targets angiogenesis and the hypoxic response, an effective combination at reducing tumour growth in preclinical studies.
Collapse
Affiliation(s)
- L L da Motta
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Biochemistry/UFRGS, Porto Alegre, Brazil
- CAPES Foundation, Ministry of Education of Brazil, Brasilia, Brazil
| | - I Ledaki
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - K Purshouse
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - S Haider
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | - D Baban
- High Throughput Genomics, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - M Morotti
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - G Steers
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - S Wigfield
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - E Bridges
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - J-L Li
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Institute of Translational and Stratified Medicine, Plymouth University, Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - S Knapp
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium, University of Oxford, Oxford, UK
- Goethe University Frankfurt, Institute for Pharmaceutical Chemistry and Buchmann Institute for Life Sciences, Campus Riedberg, Frankfurt, Germany
| | - D Ebner
- Nuffield Department of Medicine, Target Discovery Institute (TDI), University of Oxford, Oxford, UK
| | - F Klamt
- Department of Biochemistry/UFRGS, Porto Alegre, Brazil
| | - A L Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - A McIntyre
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Cancer Biology, Division of Cancer and Stem Cells, The University of Nottingham, Nottingham, UK
| |
Collapse
|
28
|
Makhoul I, Todorova VK, Siegel ER, Erickson SW, Dhakal I, Raj VR, Lee JY, Orloff MS, Griffin RJ, Henry-Tillman RS, Klimberg S, Hutchins LF, Kadlubar SA. Germline Genetic Variants in TEK, ANGPT1, ANGPT2, MMP9, FGF2 and VEGFA Are Associated with Pathologic Complete Response to Bevacizumab in Breast Cancer Patients. PLoS One 2017; 12:e0168550. [PMID: 28045923 PMCID: PMC5207665 DOI: 10.1371/journal.pone.0168550] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 12/02/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We previously reported improved pathologic complete response (pCR) in a prospective phase II study using neoadjuvant bevacizumab in combination with chemotherapy compared to chemotherapy alone in breast cancer patients (41% vs. 25%, p = 0.0291). In this study, we queried germline single-nucleotide polymorphisms (SNPs) in angiogenesis-related genes for their impact on pCR and overall survival (OS). METHODS DNA for genotyping was available from 34 subjects who received bevacizumab in addition to chemotherapy and 29 subjects who did not. Using Illumina® technology, we queried 504 SNPs with a minor allele frequency (MAF) of at least 5%, located in 10 angiogenesis-related genes, for their effect on pCR via logistic regression with an additive-inheritance model while adjusting for race and bevacizumab treatment. SNPs that showed significant associations with pCR were selected for additional characterization. RESULTS After adjusting for race and tumor type, patients who had bevacizumab added to their neoadjuvant therapy were found to experience a significantly improved rate of pCR compared to patients who did not (adjusted OR 8.40, 95% CI 1.90-37.1). When patients were analyzed for SNP effects via logistic regression with race and bevacizumab treatment included as covariates, two SNPs in angiopoietin 1 (ANGPT1), six in ANGPT2, three in fibroblast growth factor 2 (FGF2), four in matrix metalloproteinase 9 (MMP9), three in tyrosine kinase, endothelial (TEK) and two in vascular endothelial growth factor A (VEGFA) were associated with pCR (P<0.05). However, when overall survival was considered, there was no difference between treatment groups or between genotypes. CONCLUSION Genetic variability in TEK, ANGPT1, ANGPT2, FGF2, MMP9 and VEGFA is associated with pCR in bevacizumab-treated patients. Consistent with other studies, adding bevacizumab to standard chemotherapy did not impact OS, likely due to other factors and thus, while SNPs in TEK, ANGPT1, ANGPT2, FGF2, MMP9 and VEGFA were associated with pCR, they were not predictive of OS in this patient population. TRIAL REGISTRATION ClinicalTrials.gov NCT00203502.
Collapse
Affiliation(s)
- Issam Makhoul
- Division of Hematology/Oncology Division, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| | - Valentina K. Todorova
- Division of Medical Genetics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Eric R. Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Stephen W. Erickson
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Ishwori Dhakal
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Vinay R. Raj
- Division of Medical Genetics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jeannette Y. Lee
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Mohammed S. Orloff
- Department of Epidemiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Ronda S. Henry-Tillman
- Division of Breast Surgical Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Suzanne Klimberg
- Division of Breast Surgical Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Laura F. Hutchins
- Division of Hematology/Oncology Division, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Susan A. Kadlubar
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| |
Collapse
|
29
|
Predicting and Overcoming Chemotherapeutic Resistance in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:59-104. [PMID: 29282680 DOI: 10.1007/978-981-10-6020-5_4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our understanding of breast cancer and its therapeutic approach has improved greatly due to the advancement of molecular biology in recent years. Clinically, breast cancers are characterized into three basic types based on their immunohistochemical properties. They are triple-negative breast cancer, estrogen receptor (ER) and progesterone receptor (PR)-positive-HR positive breast cancer, and human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Even though these subtypes have been characterized, assessment of a breast cancer's receptor status is still widely used to determine whether or not a targeted therapy could be applied. Moreover, drug resistance is common in all breast cancer types despite the different treatment modalities applied. The development of resistance to different therapeutics is not mutually exclusive. It seems that tumor could be resistant to multiple treatment strategies, such as being both chemoresistant and monoclonal antibody resistant. However, the underlying mechanisms are complicated and need further investigation. In this chapter, we aim to provide a brief review of the different types of breast cancer and their respective treatment strategies. We also review the possible mechanisms of potential drug resistance associated with each treatment type. We believe that a better understanding of the drug resistance mechanisms can lead to a more effective and efficient therapeutic success.
Collapse
|
30
|
Strategies and Progress of Endocrine Therapy for Patients with Metastatic Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:403-418. [PMID: 29282695 DOI: 10.1007/978-981-10-6020-5_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Breast cancer is one of the most prevalent cancers and the leading causes of cancer mortality in women worldwide and in China. For hormone receptor-positive (HR+) breast cancer, accounting for approximately 60-80% of breast cancer, endocrine therapy (ET) is the primary treatment strategy. For patients with HR+ metastatic breast cancer (MBC), there are many endocrine-based treatment options that can improve long-term outcomes and optimize quality of life. With the emergence and availability of new and effective agents, the options for ET have expanded in the last two decades. Although hormone therapy has been a standard of care for many decades, treatment must be individualized based on tumor biology and extent of disease. For example, the patients with impending organ failure may be treated with induction chemotherapy to improve organ function, followed by ET. For the patients who develop metastatic disease while on adjuvant ET, particularly when associated with organ failure, or for those with low expression of hormone receptors or expression of HER2, chemotherapy again may be a preferred initial treatment. ET blocks estrogen-driven tumor growth through different mechanisms; however, HR+ MBC can be intrinsically resistant or may acquire resistance to the treatment. Several targeted agents have been approved to use in combination with ET to improve response and delay development of resistance.
Collapse
|
31
|
Butler CT, Reynolds AL, Tosetto M, Dillon ET, Guiry PJ, Cagney G, O'Sullivan J, Kennedy BN. A Quininib Analogue and Cysteinyl Leukotriene Receptor Antagonist Inhibits Vascular Endothelial Growth Factor (VEGF)-independent Angiogenesis and Exerts an Additive Antiangiogenic Response with Bevacizumab. J Biol Chem 2016; 292:3552-3567. [PMID: 28035003 DOI: 10.1074/jbc.m116.747766] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/19/2016] [Indexed: 12/31/2022] Open
Abstract
Excess blood vessel growth contributes to the pathology of metastatic cancers and age-related retinopathies. Despite development of improved treatments, these conditions are associated with high economic costs and drug resistance. Bevacizumab (Avastin®), a monoclonal antibody against vascular endothelial growth factor (VEGF), is used clinically to treat certain types of metastatic cancers. Unfortunately, many patients do not respond or inevitably become resistant to bevacizumab, highlighting the need for more effective antiangiogenic drugs with novel mechanisms of action. Previous studies discovered quininib, an antiangiogenic small molecule antagonist of cysteinyl leukotriene receptors 1 and 2 (CysLT1 and CysLT2). Here, we screened a series of quininib analogues and identified a more potent antiangiogenic novel chemical entity (IUPAC name (E)-2-(2-quinolin-2-yl-vinyl)-benzene-1,4-diol HCl) hereafter designated Q8. Q8 inhibits developmental angiogenesis in Tg(fli1:EGFP) zebrafish and inhibits human microvascular endothelial cell (HMEC-1) proliferation, tubule formation, and migration. Q8 elicits antiangiogenic effects in a VEGF-independent in vitro model of angiogenesis and exerts an additive antiangiogenic response with the anti-VEGF biologic bevacizumab. Cell-based receptor binding assays confirm that Q8 is a CysLT1 antagonist and is sufficient to reduce cellular levels of NF-κB and calpain-2 and secreted levels of the proangiogenic proteins intercellular adhesion molecule-1, vascular cell adhesion protein-1, and VEGF. Distinct reductions of VEGF by bevacizumab explain the additive antiangiogenic effects observed in combination with Q8. In summary, Q8 is a more effective antiangiogenic drug compared with quininib. The VEGF-independent activity coupled with the additive antiangiogenic response observed in combination with bevacizumab demonstrates that Q8 offers an alternative therapeutic strategy to combat resistance associated with conventional anti-VEGF therapies.
Collapse
Affiliation(s)
- Clare T Butler
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Alison L Reynolds
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Miriam Tosetto
- Centre for Colorectal Disease, St. Vincent's University Hospital, Dublin 4, Ireland, and
| | - Eugene T Dillon
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Patrick J Guiry
- UCD School of Chemistry, UCD Centre for Synthesis and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gerard Cagney
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Jacintha O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Breandán N Kennedy
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| |
Collapse
|
32
|
Bear HD, Tang G, Rastogi P, Geyer CE, Zoon CK, Kidwell KM, Robidoux A, Baez-Diaz L, Brufsky AM, Mehta RS, Fehrenbacher L, Young JA, Senecal FM, Gaur R, Margolese RG, Adams PT, Gross HM, Costantino JP, Paik S, Swain SM, Mamounas EP, Wolmark N. The Effect on Surgical Complications of Bevacizumab Added to Neoadjuvant Chemotherapy for Breast Cancer: NRG Oncology/NSABP Protocol B-40. Ann Surg Oncol 2016; 24:1853-1860. [PMID: 27864694 DOI: 10.1245/s10434-016-5662-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND NRG Oncology/NSABP trial B-40 tested the impact of adding bevacizumab (bev) to neoadjuvant chemotherapy for operable breast cancer. Secondary endpoints included rates of surgical complications after surgery in patients who did or did not receive bev. METHODS A total of 1206 women with HER2-negative operable breast cancer were randomly assigned to receive one of three different docetaxel-plus-anthracycline-based regimens, without or with bev (15 mg/kg every 3 weeks) for the first 6 of 8 cycles and for 10 doses postoperatively. Surgical complications were assessed from date of surgery through 24 months following study entry. RESULTS Early surgical complications were significantly more frequent in the bev group (25.4 vs. 18.9%; trend test p = 0.008), but most were grade 1-2. Early noninfectious wound dehiscences were infrequent and not significantly different (5.4 vs. 3.1%; trend test p = 0.15). Long-term noninfectious wound complications were significantly higher for patients receiving bev (11.8 vs. 5.1%; trend test p = 0.0007), but the incidence of grade ≥3 wound dehiscence was low in both groups (<1%). Among 193 patients undergoing expander or implant reconstructions, 19 (19.6%) of 97 in the bev-receiving group versus 10 (10.4%) of 96 in the non-bev group had grade ≥3 complications (Pearson, p = 0.11). CONCLUSIONS Overall, adding bev increased surgical complications, but most serious complications were not significantly increased. In particular, the need for surgical intervention in patients undergoing breast reconstruction with prosthetic implants was higher with bev but was not statistically significantly different. With precautions, bev can be used safely perioperatively in patients undergoing surgery for breast cancer.
Collapse
Affiliation(s)
- Harry D Bear
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA. .,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Gong Tang
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,University of Pittsburgh, Pittsburgh, PA, USA
| | - Priya Rastogi
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,University of Pittsburgh Cancer Institute School of Medicine, Pittsburgh, PA, USA
| | - Charles E Geyer
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Christine K Zoon
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Kelley M Kidwell
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,University of Pittsburgh, Pittsburgh, PA, USA.,Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - André Robidoux
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Luis Baez-Diaz
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,San Juan MBCCOP, San Juan, PR, USA
| | - Adam M Brufsky
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,University of Pittsburgh/Magee Womens Hospital, Pittsburgh, PA, USA
| | - Rita S Mehta
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,School of Medicine, Chao Family Comprehensive Cancer Center, University of California at Irvine, Orange, CA, USA
| | - Louis Fehrenbacher
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Kaiser Permanente Oncology Clinical Trials, Northern California, Vallejo, CA, USA
| | - James A Young
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,CCOP, Colorado Cancer Research Program, Colorado Springs, CO, USA
| | - Francis M Senecal
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,CCOP, North-West Medical Specialties, Tacoma, WA, USA
| | - Rakesh Gaur
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Kansas City Clinical Oncology Program, Kansas City, MO, USA
| | - Richard G Margolese
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Paul T Adams
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Genesys Regional Medical Center, Grand Blanc, MI, USA
| | - Howard M Gross
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Dayton CCOP, Dayton, OH, USA
| | - Joseph P Costantino
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,University of Pittsburgh, Pittsburgh, PA, USA
| | - Soonmyung Paik
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Severance Biomedical Science Institute and Department of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Sandra M Swain
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Washington Cancer Institute, Washington Hospital Center, Washington, DC, USA.,Georgetown University Medical Center, Washington, DC, USA
| | - Eleftherios P Mamounas
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,UF Health Cancer Center at Orlando Health, Orlando, FL, USA
| | - Norman Wolmark
- NRG Oncology and the National Surgical Adjuvant Breast and Bowel Project (NSABP) (NSABP legacy trials are now part of the NRG Oncology portfolio), Pittsburgh, PA, USA.,Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Molecular Markers and Targeted Therapeutics in Metastatic Tumors of the Spine: Changing the Treatment Paradigms. Spine (Phila Pa 1976) 2016; 41 Suppl 20:S218-S223. [PMID: 27488299 DOI: 10.1097/brs.0000000000001833] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY TYPE A review of the literature. OBJECTIVE The aim of this study was to discuss the evolution of molecular signatures and the history and development of targeted therapeutics in metastatic tumor types affecting the spinal column. SUMMARY OF BACKGROUND DATA Molecular characterization of metastatic spine tumors is expected to usher in a revolution in diagnostic and treatment paradigms. Molecular characterization will provide critical information that can be used for initial diagnosis, prognosticating the ideal treatment strategy, assessment of treatment efficacy, surveillance and monitoring recurrence, and predicting complications, clinical outcome, and overall survival in patients diagnosed with metastatic cancers to the spinal column. METHODS A review of the literature was performed focusing on illustrative examples of the role that molecular-based therapeutics have played in clinical outcomes for patients diagnosed with metastatic tumor types affecting the spinal column. RESULTS The impact of molecular therapeutics including receptor tyrosine kinases and immune checkpoint inhibitors and the ability of molecular signatures to provide prognostic information are discussed in metastatic breast cancer, lung cancer, prostate cancer, melanoma, and renal cell cancer affecting the spinal column. CONCLUSION For the providers who will ultimately counsel patients diagnosed with metastases to the spinal column, molecular advancements will radically alter the management/surgical paradigms utilized. Ultimately, the translation of these molecular advancements into routine clinical care will greatly improve the quality and quantity of life for patients diagnosed with spinal malignancies and provide better overall outcomes and counseling for treating physicians. LEVEL OF EVIDENCE N/A.
Collapse
|
34
|
New potential beneficial effects of actein, a triterpene glycoside isolated from Cimicifuga species, in breast cancer treatment. Sci Rep 2016; 6:35263. [PMID: 27731376 PMCID: PMC5059658 DOI: 10.1038/srep35263] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/27/2016] [Indexed: 12/03/2022] Open
Abstract
Actein is a triterpene glycoside isolated from the rhizomes of Cimicifuga foetida (Chinese herb “shengma”) which could inhibit the growth of breast cancer cells. Nevertheless, the effect of actein on angiogenesis, which is an essential step for tumor growth and metastasis, has never been reported. Hence, this study aimed to investigate the in vitro and in vivo effects of actein on angiogenesis using human microvascular endothelial cells (HMEC-1), matrigel plug and tumor-bearing mouse models. Our results showed that actein significantly inhibited the proliferation, reduced the migration and motility of endothelial cells, and it could suppress the protein expressions of VEGFR1, pJNK and pERK, suggesting that JNK/ERK pathways were involved. In vivo results showed that oral administration of actein at 10 mg/kg for 7 days inhibited blood vessel formation in the growth factor-containing matrigel plugs. Oral actein treatments (10–15 mg/kg) for 28 days resulted in decreasing mouse 4T1 breast tumor sizes and metastasis to lungs and livers. The apparent reduced angiogenic proteins (CD34 and Factor VIII) expressions and down-regulated metastasis-related VEGFR1 and CXCR4 gene expressions were observed in breast tumors. Our novel findings provide insights into the use of actein for development of anti-angiogenic agents for breast cancer.
Collapse
|
35
|
Liu M, Li Z, Yang J, Jiang Y, Chen Z, Ali Z, He N, Wang Z. Cell-specific biomarkers and targeted biopharmaceuticals for breast cancer treatment. Cell Prolif 2016; 49:409-20. [PMID: 27312135 PMCID: PMC6496337 DOI: 10.1111/cpr.12266] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 05/04/2016] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the second leading cause of cancer death among women, and its related treatment has been attracting significant attention over the past decades. Among the various treatments, targeted therapy has shown great promise as a precision treatment, by binding to cancer cell-specific biomarkers. So far, great achievements have been made in targeted therapy of breast cancer. In this review, we first discuss cell-specific biomarkers, which are not only useful for classification of breast cancer subtyping but also can be utilized as goals for targeted therapy. Then, the innovative and generic-targeted biopharmaceuticals for breast cancer, including monoclonal antibodies, non-antibody proteins and small molecule drugs, are reviewed. Finally, we provide our outlook on future developments of biopharmaceuticals, and provide solutions to problems in this field.
Collapse
Affiliation(s)
- Mei Liu
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Zhiyang Li
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
- Department of Laboratory MedicineNanjing Drum Tower Hospital Clinical CollegeNanjing UniversityNanjingChina
| | - Jingjing Yang
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| | - Yanyun Jiang
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| | - Zhongsi Chen
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Zeeshan Ali
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| | - Nongyue He
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Zhifei Wang
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
36
|
Elgebaly A, Menshawy A, El Ashal G, Osama O, Ghanem E, Omar A, Negida A. Sunitinib alone or in combination with chemotherapy for the treatment of advanced breast cancer: A systematic review and meta-analysis. Breast Dis 2016; 36:91-101. [PMID: 27612040 DOI: 10.3233/bd-160218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Sunitinib is an orally delivered tyrosine kinase inhibitor that exhibits antiangiogenic effects. FDA has approved sunitinib for the treatment of metastatic renal cell carcinoma. However, its efficacy for the treatment of advanced breast cancer (ABC) remains controversial. Therefore, we performed this systematic review and meta-analysis to synthesize evidence from published randomized controlled trials (RCTs) about the efficacy of sunitinib alone and in combination with chemotherapy for the treatment of ABC. METHODS We followed PRISMA statement guidelines during the preparation of this systematic review and meta-analysis. A computer literature search of PubMed, SCOPUS, web of knowledge, and Cochrane Central Register of Controlled Trials (CENTRAL) has been conducted using relevant keywords. Studies were screened for eligibility and data were extracted to an online data extraction form. Progression free survival (PFS) and overall survival (OS) were pooled as Hazard ratio (HR) in a meta-analysis model using generic inverse variance method. Objective response rate (ORR) and complications were pooled as relative risk (RR) in a random effect model meta-analysis using Mantel-Haenzel method. RESULTS Six RCTs, with a total sample size of 2273 patients, met our eligibility criteria and were included in this meta-analysis. Sunitinib monotherapy was not superior to chemotherapy in terms of PFS (HR = 1.00, 95% CI [0.86 to 1.16], P = 0.99), OS (HR = 1.07; 95% CI [0.87 to 1.32], P = 0.5), or ORR (RR = 0.70, 95% CI [0.74 to 1.03], P = 0.07). Sunitinib in combination with chemotherapy did not show superiority to chemotherapy in terms of PFS (HR = 0.99, 95% CI [0.86 to 1.14], P = 0.89) and OS (HR = 1.04, 95% CI [0.85 to 1.28], P = 0.69). However, the ORR favored sunitinib in combination with chemotherapy group (RR = 1.15, 95% CI [1.01 to 1.31]) with a statistically significant P value (P = 0.03). CONCLUSIONS Current evidence shows that sunitinib, either alone or in combination with chemotherapy, has no clinical benefit for patients with advanced breast cancer. However, previous studies did not considered patient stratification and outcome assessment based on molecular markers. In terms of safety, toxicity was common with sunitinib treatment.
Collapse
Affiliation(s)
- Ahmed Elgebaly
- Medical Research Group of Egypt, Egypt
- Faculty of Medicine, Al Azhar University, Cairo, Egypt
| | - Ahmed Menshawy
- Medical Research Group of Egypt, Egypt
- Faculty of Medicine, Al Azhar University, Cairo, Egypt
| | - Gehad El Ashal
- Medical Research Group of Egypt, Egypt
- Kasr Al ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Omnya Osama
- Medical Research Group of Egypt, Egypt
- Kasr Al ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Esraa Ghanem
- Medical Research Group of Egypt, Egypt
- Faculty of Medicine, Al Azhar University, Cairo, Egypt
| | - Ahmed Omar
- Medical Research Group of Egypt, Egypt
- Kasr Al ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Negida
- Medical Research Group of Egypt, Egypt
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- Student Research Unit, Zagazig University, Zagazig, El-Sharkia, Egypt
- CURE and MORE national research collaborative
| |
Collapse
|
37
|
Bevacizumab plus neoadjuvant chemotherapy in patients with HER2-negative inflammatory breast cancer (BEVERLY-1): a multicentre, single-arm, phase 2 study. Lancet Oncol 2016; 17:600-11. [DOI: 10.1016/s1470-2045(16)00011-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/04/2016] [Accepted: 01/04/2016] [Indexed: 01/24/2023]
|
38
|
Humbert O, Riedinger JM, Vrigneaud JM, Kanoun S, Dygai-Cochet I, Berriolo-Riedinger A, Toubeau M, Depardon E, Lassere M, Tisserand S, Fumoleau P, Brunotte F, Cochet A. 18F-FDG PET-Derived Tumor Blood Flow Changes After 1 Cycle of Neoadjuvant Chemotherapy Predicts Outcome in Triple-Negative Breast Cancer. J Nucl Med 2016; 57:1707-1712. [PMID: 27103025 DOI: 10.2967/jnumed.116.172759] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/14/2016] [Indexed: 12/20/2022] Open
Abstract
Previous studies have suggested that early changes in blood flow (BF) in response to neoadjuvant chemotherapy and evaluated with 15O-water are a surrogate biomarker of outcome in women with breast cancer. This study investigates, in the triple-negative breast cancer subtype, the prognostic relevance of tumor BF changes (ΔBF) in response to chemotherapy, assessed using a short dynamic 18F-FDG PET acquisition. METHODS Forty-six consecutive women with triple-negative breast cancer and an indication for neoadjuvant chemotherapy were prospectively included. Women benefited from a baseline 18F-FDG PET examination with a 2-min chest-centered dynamic acquisition, started at the time of 18F-FDG injection. Breast tumor perfusion was calculated from this short dynamic image using a first-pass model. This dynamic PET acquisition was repeated after the first cycle of chemotherapy to measure early ΔBF. Delayed static PET acquisitions were also performed (90 min after 18F-FDG injection) to measure changes in tumor glucose metabolism (ΔSUVmax). The association between tumor BF, clinicopathologic characteristics, and patients' overall survival (OS) was evaluated. RESULTS Median baseline tumor BF was 21 mL/min/100 g (range, 6-46 mL/min/100 g) and did not significantly differ according to tumor size, Scarf-Bloom-Richardson grade, or Ki-67 expression. Median tumor ∆BF was -30%, with highly scattered values (range, -93% to +118%). A weak correlation was observed between ΔBF and ∆SUVmax (r = +0.40, P = 0.01). The median follow-up was 30 mo (range, 6-73 mo). Eight women developed recurrent disease, 7 of whom died. Low OS was associated with menopausal history (P = 0.03), persistent or increased tumor vascularization on the interim PET (ΔBF cutoff = -30%; P = 0.03), non-breast-conserving surgery (P = 0.04), and the absence of a pathologic complete response (pCR) (P = 0.01). ΔBF and pCR provided incremental prognostic stratification: 3-y OS was 100% in pCR women, 87% in no-pCR women but achieving an early tumor BF response, and only 48% in no-pCR/no-BF-response women (ΔBF cutoff = -30%, P < 0.001). CONCLUSION This study suggests the clinical usefulness of an early user- and patient-friendly 2-min dynamic acquisition to monitor breast tumor ΔBF to neoadjuvant chemotherapy using 18F-FDG PET/CT. Monitoring tumor perfusion and angiogenesis response to treatment seems to be a promising target for PET tracers.
Collapse
Affiliation(s)
- Olivier Humbert
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France .,LE2I UMR 6306, CNRS, Arts et Métiers, Université de Bourgogne Franche-Comté, Besançon, France
| | - Jean-Marc Riedinger
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France.,Departments of Biology and Pathology, Centre GF Leclerc, Dijon, France
| | - Jean-Marc Vrigneaud
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France.,LE2I UMR 6306, CNRS, Arts et Métiers, Université de Bourgogne Franche-Comté, Besançon, France
| | - Salim Kanoun
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France.,LE2I UMR 6306, CNRS, Arts et Métiers, Université de Bourgogne Franche-Comté, Besançon, France.,Imaging Department, CHU Le Bocage, Dijon, France; and
| | | | | | - Michel Toubeau
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France
| | - Edouard Depardon
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France
| | - Maud Lassere
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France
| | - Simon Tisserand
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France
| | - Pierre Fumoleau
- Department of Medical Oncology, Centre GF Leclerc, Dijon, France
| | - François Brunotte
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France.,LE2I UMR 6306, CNRS, Arts et Métiers, Université de Bourgogne Franche-Comté, Besançon, France.,Imaging Department, CHU Le Bocage, Dijon, France; and
| | - Alexandre Cochet
- Department of Nuclear Medicine, Centre GF Leclerc, Dijon, France.,LE2I UMR 6306, CNRS, Arts et Métiers, Université de Bourgogne Franche-Comté, Besançon, France.,Imaging Department, CHU Le Bocage, Dijon, France; and
| |
Collapse
|
39
|
Arpino G, Marmé F, Cortés J, Ricevuto E, Leonard R, Llombart-Cussac A. Tailoring the dosing schedule of nab-paclitaxel in metastatic breast cancer according to patient and disease characteristics: Recommendations from a panel of experts. Crit Rev Oncol Hematol 2016; 99:81-90. [DOI: 10.1016/j.critrevonc.2015.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/03/2015] [Accepted: 10/20/2015] [Indexed: 02/08/2023] Open
|
40
|
Li X, Zhu S, Hong C, Cai H. Angiogenesis inhibitors for patients with ovarian cancer: a meta-analysis of 12 randomized controlled trials. Curr Med Res Opin 2016; 32:555-62. [PMID: 26652645 DOI: 10.1185/03007995.2015.1131152] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To investigate the effects of angiogenesis inhibitors in the treatment of patients with advanced or recurrent ovarian cancer, a meta-analysis was performed and overall survival (OS), progression-free survival (PFS), and toxicity were assessed. PATIENTS AND METHODS The PubMed and Embase databases, and the Cochrane Central Register of Controlled Trials were searched for publications between January 2000 and June 2015. Hazard ratios (HRs) with their 95% confidence intervals (CIs), or data for calculating HRs with 95% CIs were derived. RESULTS The 12 trials in this meta-analysis were divided into three groups: four trials with a VEGF inhibitor (the bevacizumab group), six trials with VEGFR inhibitors (the VEGFRIs group), and two trials with an angiopoietin inhibitor (the trebananib group). PFS improvement was seen in all groups (HR = 0.61, 95% CI 0.48 to 0.79, P < 0.001 for bevacizumab; HR = 0.71, 95% CI 0.59 to 0.87, P = 0.001 for VEGFRIs; and HR = 0.67, 95% CI 0.62 to 0.72, P < 0.001 for trebananib). Regarding OS, bevacizumab showed a trend of improvement (HR = 0.90, 95% CI 0.80 to 1.01, P = 0.079), VEGFRIs showed no improvement (HR = 0.92, 95% CI 0.75 to 1.11, P = 0.368), and trebananib demonstrated a significant prolongation (HR = 0.81, 95% CI 0.67 to 0.99, P = 0.036). Bevacizumab was associated with more class-specific adverse events (RR = 4.05, 95% CI 1.99 to 8.27, P < 0.001). Although the toxicity profiles differed, VEGFRIs developed common higher incidences of hypertension, diarrhea, and fatigue. A higher incidence of edema was reported in the trebananib group (RR = 2.60, 95% CI 0.84 to 8.00, P = 0.097). CONCLUSIONS Anti-angiogenic therapy showed clear PFS benefit with increased toxicity, but its role in OS was undefined for ovarian cancer which emphasized the need for patient selection.
Collapse
Affiliation(s)
- Xuyuan Li
- a a Department of Medical Oncology , Affiliated Shantou Hospital of Sun Yat-sen University , Shantou , Guangdong , China
| | - Sujuan Zhu
- b b Department of Good Clinical Practice , Cancer Hospital of Shantou University Medical College , Shantou , Guangdong , China
| | - Chaoqun Hong
- c c Cancer Research Lab, Cancer Hospital of Shantou University Medical College , Shantou , Guangdong , China
| | - Haoquan Cai
- d d Department of Medical Oncology , First affiliated Hospital of Shantou University Medical College , Shantou , Guangdong , China
| |
Collapse
|
41
|
Guo P, Yang J, Jia D, Moses MA, Auguste DT. ICAM-1-Targeted, Lcn2 siRNA-Encapsulating Liposomes are Potent Anti-angiogenic Agents for Triple Negative Breast Cancer. Theranostics 2016; 6:1-13. [PMID: 26722369 PMCID: PMC4679350 DOI: 10.7150/thno.12167] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/16/2015] [Indexed: 12/21/2022] Open
Abstract
Lipocalin 2 (Lcn2) is a promising therapeutic target as well as a potential diagnostic biomarker for breast cancer. It has been previously shown to promote breast cancer progression by inducing the epithelial to mesenchymal transition in breast cancer cells as well as by enhancing angiogenesis. Lcn2 levels in urine and tissue samples of breast cancer patients has also been correlated with breast cancer status and poor patient prognosis. In this study, we have engineered a novel liposomal small interfering RNA (siRNA) delivery system to target triple negative breast cancer (TNBC) via a recently identified molecular target, intercellular adhesion molecule-1 (ICAM-1). This ICAM-1-targeted, Lcn2 siRNA- encapsulating liposome (ICAM-Lcn2-LP) binds human TNBC MDA-MB-231cells significantly stronger than non-neoplastic MCF-10A cells. Efficient Lcn2 knockdown by ICAM-Lcn2-LPs led to a significant reduction in the production of vascular endothelial growth factor (VEGF) from MDA-MB-231 cells, which, in turn, led to reduced angiogenesis both in vitro and in vivo. Angiogenesis (neovascularization) is a requirement for solid tumor growth and progression, and its inhibition is an important therapeutic strategy for human cancers. Our results indicate that a tumor-specific strategy such as the TNBC-targeted, anti-angiogenic therapeutic approach developed here, may be clinically useful in inhibiting TNBC progression.
Collapse
Affiliation(s)
- Peng Guo
- 1. Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, United States
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Jiang Yang
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Di Jia
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Marsha A. Moses
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Debra T. Auguste
- 1. Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, New York, NY 10031, United States
- 2. Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
- 3. Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| |
Collapse
|
42
|
Kalimutho M, Parsons K, Mittal D, López JA, Srihari S, Khanna KK. Targeted Therapies for Triple-Negative Breast Cancer: Combating a Stubborn Disease. Trends Pharmacol Sci 2015; 36:822-846. [PMID: 26538316 DOI: 10.1016/j.tips.2015.08.009] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/14/2015] [Accepted: 08/17/2015] [Indexed: 11/17/2022]
Abstract
Triple-negative breast cancers (TNBCs) constitute a heterogeneous subtype of breast cancers that have a poor clinical outcome. Although no approved targeted therapy is available for TNBCs, molecular-profiling efforts have revealed promising molecular targets, with several candidate compounds having now entered clinical trials for TNBC patients. However, initial results remain modest, thereby highlighting challenges potentially involving intra- and intertumoral heterogeneity and acquisition of therapy resistance. We present a comprehensive review on emerging targeted therapies for treating TNBCs, including the promising approach of immunotherapy and the prognostic value of tumor-infiltrating lymphocytes. We discuss the impact of pathway rewiring in the acquisition of drug resistance, and the prospect of employing combination therapy strategies to overcome challenges towards identifying clinically-viable targeted treatment options for TNBC.
Collapse
Affiliation(s)
- Murugan Kalimutho
- Signal Transduction Laboratory, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia.
| | - Kate Parsons
- Signal Transduction Laboratory, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Natural Sciences, Griffith University, Nathan, QLD 411, Australia
| | - Deepak Mittal
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - J Alejandro López
- School of Natural Sciences, Griffith University, Nathan, QLD 411, Australia; Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Sriganesh Srihari
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Kum Kum Khanna
- Signal Transduction Laboratory, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Natural Sciences, Griffith University, Nathan, QLD 411, Australia.
| |
Collapse
|
43
|
Reinert T, Barrios CH. Optimal management of hormone receptor positive metastatic breast cancer in 2016. Ther Adv Med Oncol 2015; 7:304-20. [PMID: 26557899 PMCID: PMC4622303 DOI: 10.1177/1758834015608993] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hormone receptor positive tumors represent the most common form of breast cancer and account for most of the deaths from the disease. Endocrine therapy represents the main initial therapeutic strategy for these patients and has been associated with significant clinical benefits in a majority of patients. While in early stages endocrine therapy is administered as part of a curative approach once clinical metastases develop, the disease is considered incurable and the main management objectives are tumor control and quality of life. The two major clinical paradigms of always indicating endocrine therapy in the absence of visceral crises and sequencing endocrine treatments have been guiding our therapeutic approach to these patients. However, for many decades, we have delivered endocrine therapy with a 'one size fits all' approach by applying agents that interfere with hormone receptor signaling equally in every clinical patient scenario. We have been unable to incorporate the well-known biologic principle of different degrees of hormone receptor dependency in our therapeutic recommendations. Recent developments in the understanding of molecular interactions of hormone signaling with other important growth factor, metabolic and cell division pathways have opened the possibility of improving results by modulating hormone signaling and interfering with resistance mechanisms yet to be fully understood. Unfortunately, limitations in the design of trials conducted in this area have made it difficult to develop predictive biomarkers and most of the new combinations with targeted agents, even though showing improvements in clinical endpoints, have been directed to an unselected population of patients. In this review we explore some of the current and most relevant literature in the management of hormone receptor positive advance breast cancer.
Collapse
Affiliation(s)
- Tomas Reinert
- Instituto do Câncer, Sistema de Saúde Mãe de Deus, Porto Alegre, RS, Brazil
| | - Carlos H. Barrios
- PUCRS School of Medicine, Department of Medicine, Padre Chagas 66/203, CEP 90 570 080, Porto Alegre, RS, Brazil
| |
Collapse
|
44
|
Al-Abd AM, Aljehani ZK, Gazzaz RW, Fakhri SH, Jabbad AH, Alahdal AM, Torchilin VP. Pharmacokinetic strategies to improve drug penetration and entrapment within solid tumors. J Control Release 2015; 219:269-277. [PMID: 26342660 DOI: 10.1016/j.jconrel.2015.08.055] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/09/2015] [Accepted: 08/28/2015] [Indexed: 02/08/2023]
Abstract
Despite the discovery of a large number of anticancer agents, cancer still remains among the leading causes of death since the middle of the twentieth century. Solid tumors possess a high degree of genetic instability and emergence of treatment resistance. Tumor resistance has emerged for almost all approved anticancer drugs and will most probably emerge for newly discovered anticancer agents as well. The use of pharmacokinetic approaches to increase anticancer drug concentrations within the solid tumor compartment and prolong its entrapment might diminish the possibility of resistance emergence at the molecular pharmacodynamic level and might even reverse tumor resistance. Several novel treatment modalities such as metronomic therapy, angiogenesis inhibitors, vascular disrupting agents and tumor priming have been introduced to improve solid tumor treatment outcomes. In the current review we will discuss the pharmacokinetic aspect of these treatment modalities in addition to other older treatment modalities, such as extracellular matrix dissolving agents, extracellular matrix synthesis inhibitors, chemoembolization and cellular efflux pump inhibition. Many of these strategies showed variable degrees of success/failure; however, reallocating these modalities based on their influence on the intratumoral pharmacokinetics might improve their understanding and treatment outcomes.
Collapse
Affiliation(s)
- Ahmed M Al-Abd
- Department of Pharmacology, Medical Division, National Research Centre, Dokki, Giza, Egypt; Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA; Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zekra K Aljehani
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rana W Gazzaz
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sarah H Fakhri
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aisha H Jabbad
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
45
|
Bear HD, Tang G, Rastogi P, Geyer CE, Liu Q, Robidoux A, Baez-Diaz L, Brufsky AM, Mehta RS, Fehrenbacher L, Young JA, Senecal FM, Gaur R, Margolese RG, Adams PT, Gross HM, Costantino JP, Paik S, Swain SM, Mamounas EP, Wolmark N. Neoadjuvant plus adjuvant bevacizumab in early breast cancer (NSABP B-40 [NRG Oncology]): secondary outcomes of a phase 3, randomised controlled trial. Lancet Oncol 2015; 16:1037-1048. [PMID: 26272770 PMCID: PMC4624323 DOI: 10.1016/s1470-2045(15)00041-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND NSABP B-40 was a 3 × 2 factorial trial testing whether adding capecitabine or gemcitabine to docetaxel followed by doxorubicin plus cyclophosphamide neoadjuvant chemotherapy would improve outcomes in women with operable, HER2-negative breast cancer and whether adding neoadjuvant plus adjuvant bevacizumab to neoadjuvant chemotherapy regimens would also improve outcomes. As reported previously, addition of neoadjuvant bevacizumab increased the proportion of patients achieving a pathological complete response, which was the primary endpoint. We present secondary patient outcomes, including disease-free survival, a specified endpoint by protocol, and data for distant recurrence-free interval, and overall survival, which were not prespecified endpoints but were collected prospectively. METHODS In this randomised controlled trial (NSABP B-40), we enrolled women aged 18 years or older, with operable, HER2-non-amplified invasive adenocarcinoma of the breast, 2 cm or greater in diameter by palpation, clinical stage T1c-3, cN0, cN1, or cN2a, without metastatic disease and diagnosed by core needle biopsy. Patients received one of three docetaxel-based neoadjuvant regimens for four cycles: docetaxel alone (100 mg/m(2)) with addition of capecitabine (825 mg/m(2) oral twice daily days 1-14, 75 mg/m(2) docetaxel) or with addition of gemcitabine (1000 mg/m(2) days 1 and 8 intravenously, 75 mg/m(2) docetaxel), all followed by neoadjuvant doxorubicin and cyclophosphamide (60 mg/m(2) and 600 mg/m(2) intravenously) every 3 weeks for four cycles. Those randomly assigned to bevacizumab groups were to receive bevacizumab (15 mg/kg, every 3 weeks for six cycles) with neoadjuvant chemotherapy and postoperatively for ten doses. Randomisation was done (1:1:1:1:1:1) via a biased-coin minimisation procedure to balance the characteristics with respect to clinical nodal status, clinical tumour size, hormone receptor status, and age. Intent-to-treat analyses were done for disease-free survival and overall survival. This study is registered with ClinicalTrials.gov, number NCT00408408. FINDINGS Between Jan 5, 2007, and June 30, 2010, 1206 patients were enrolled in the study. Follow-up data were collected from Oct 31, 2007 to March 27, 2014, and were available for overall survival in 1186 patients, disease-free survival in 1184, and distant recurrence-free interval in 1181. Neither capecitabine nor gemcitabine increased disease-free survival or overall survival. Median follow-up was 4·7 years (IQR 4·0-5·2). The addition of bevacizumab significantly increased overall survival (hazard ratio 0·65 [95% CI 0·49-0·88]; p=0·004) but did not significantly increase disease-free survival (0·80 [0·63-1·01]; p=0·06). Four deaths occurred on treatment due to vascular disorder (docetaxel plus capecitabine followed by doxorubicin plus cyclophosphamide group), sudden death (docetaxel plus capecitabine followed by doxorubicin plus cyclophosphamide group), infective endocarditis (docetaxel plus bevacizumab followed by doxorubicin plus cyclophosphamide and bevacizumab group), and visceral arterial ischaemia (docetaxel followed by doxorubicin plus cyclophosphamide group). The most common grade 3-4 adverse events in the bevacizumab group were neutropenia (grade 3, 99 [17%]; grade 4, 37 [6%]), hand-foot syndrome (grade 3, 63 [11%]), and hypertension (grade 3, 60 [10%]; grade 4, two [<1%]) and in the non-bevacizumab group were neutropenia (grade 3, 98 [16%]; grade 4, 36 [6%]), fatigue (grade 3, 53 [9%]), and hand-foot syndrome (grade 3, 43 [7%]). INTERPRETATION The addition of gemcitabine or capecitabine to neoadjuvant docetaxel plus doxorubicin plus cyclophosphamide does not seem to provide any benefit to patients with operable breast cancer, and should not change clinical practice in the short term. The improved overall survival with bevacizumab contradicts the findings of other studies of bevacizumab in breast cancer and may indicate the need for additional investigation of this agent. FUNDING National Institutes of Health, Genentech, Roche Laboratories, Lilly Research Laboratories, and Precision Therapeutics.
Collapse
Affiliation(s)
- Harry D Bear
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Virginia Commonwealth University School of Medicine and the Massey Cancer Center, Richmond, VA, USA.
| | - Gong Tang
- NRG Oncology and the University of Pittsburgh, Pittsburgh, PA, USA
| | - Priya Rastogi
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles E Geyer
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Virginia Commonwealth University School of Medicine and the Massey Cancer Center, Richmond, VA, USA
| | - Qing Liu
- NRG Oncology and the University of Pittsburgh, Pittsburgh, PA, USA
| | - André Robidoux
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Luis Baez-Diaz
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Minority-Based CCOP, San Juan, Puerto Rico
| | - Adam M Brufsky
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; University of Pittsburgh Cancer Institute, Magee Womens Hospital, Pittsburgh, PA, USA
| | - Rita S Mehta
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; University of California at Irvine, Division of Hematology/Oncology, Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | - Louis Fehrenbacher
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Kaiser Permanente Oncology Clinical Trials, Northern California, Vallejo, CA, USA
| | - James A Young
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Colorado Cancer Research Program, Colorado Springs, CO, USA
| | - Francis M Senecal
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Northwest Medical Specialties, Tacoma, WA, USA
| | - Rakesh Gaur
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; CCOP, Kansas City, MO, USA
| | - Richard G Margolese
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Paul T Adams
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Genesys Regional Medical Center, Grand Blanc, MI, USA
| | - Howard M Gross
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; CCOP, Dayton, Dayton, OH, USA
| | | | - Soonmyung Paik
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Severance Biomedical Science Institute and Department of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Sandra M Swain
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Washington Cancer Institute, MedStar Washington Hospital Center, Washington, DC, USA
| | - Eleftherios P Mamounas
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; UF Health Cancer Center at Orlando Health, Orlando, FL, USA
| | - Norman Wolmark
- National Surgical Adjuvant Breast and Bowel Project (NSABP/NRG Oncology), Pittsburgh, PA, USA; Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA, USA
| |
Collapse
|
46
|
Di Salvatore M, Lo Giudice L, Rossi E, Santonocito C, Nazzicone G, Rodriquenz MG, Cappuccio S, Inno A, Fuso P, Orlandi A, Strippoli A, Capoluongo E, Astone A, Cassano A, Barone C. Association of IL-8 and eNOS polymorphisms with clinical outcomes in bevacizumab-treated breast cancer patients: an exploratory analysis. Clin Transl Oncol 2015; 18:40-6. [PMID: 26141413 DOI: 10.1007/s12094-015-1334-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 06/20/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND The role of bevacizumab in metastatic breast cancer is controversial. Identification of predictive biomarkers could help to select patients who really benefit from it. We evaluated the association of angiogenesis-related gene polymorphisms with the treatment outcome of bevacizumab in metastatic breast cancer patients. PATIENTS AND METHODS eNOS-786T/C and -894G/T, IL-8-251T/A genomic polymorphisms were assessed in 31 metastatic breast cancer patients treated with bevacizumab plus chemotherapy in the first-line setting. Testing for association between each polymorphism and treatment outcome was performed. RESULTS Patients with IL-8 251 AA genotype showed a significantly lower progression-free survival in each combination comparison: "TT" vs "AA" (13 vs 8 months; p = 0.008); TT vs TA vs AA (13 vs 11 vs 8 months; p = 0.02); TT vs TA +AA (13 vs 11 months; p = 0.01); TT + TA vs AA (12 vs 8 months; p = 0.01) and a lower overall survival when compared with TT +TA genotype (26 vs 51 months, p = 0.04). Patients carrying eNOS 894 TT genotype showed a statistically significant lower progression-free survival than patients with GG genotype (11.5 vs 26.5 months; p = 0.04) with no differences in the overall survival. No association with response rate was found with any of the polymorphisms analyzed. CONCLUSION These findings suggest that IL-8 251T/A and eNOS-894 G/T polymorphisms might have a role in predicting treatment outcome of bevacizumab in metastatic breast cancer. Our results are hypothesis generating and need to be confirmed in larger clinical trials.
Collapse
Affiliation(s)
- M Di Salvatore
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy.
| | - L Lo Giudice
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - E Rossi
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - C Santonocito
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - G Nazzicone
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - M G Rodriquenz
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - S Cappuccio
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Inno
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy.,Medical Oncology, Sacro Cuore Don Calabria Hospital, Via Don A. Sempreboni 5, 37024, Negrar, VR, Italy
| | - P Fuso
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Orlandi
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Strippoli
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - E Capoluongo
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Astone
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Cassano
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - C Barone
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| |
Collapse
|
47
|
Cunha SI, Bocci M, Lövrot J, Eleftheriou N, Roswall P, Cordero E, Lindström L, Bartoschek M, Haller BK, Pearsall RS, Mulivor AW, Kumar R, Larsson C, Bergh J, Pietras K. Endothelial ALK1 Is a Therapeutic Target to Block Metastatic Dissemination of Breast Cancer. Cancer Res 2015; 75:2445-56. [DOI: 10.1158/0008-5472.can-14-3706] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Santos LVD, Cruz MR, Lopes GDL, Lima JPDSN. VEGF-A levels in bevacizumab-treated breast cancer patients: a systematic review and meta-analysis. Breast Cancer Res Treat 2015; 151:481-9. [PMID: 25947646 DOI: 10.1007/s10549-015-3410-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/29/2015] [Indexed: 11/27/2022]
Abstract
Bevacizumab may improve outcomes of patients with breast cancer, but the absence of an established biomarker hampers patient selection and researchers´ ability to demonstrate a clear survival benefit. Its putative target, circulating VEGF-A, emerged as the main candidate and we sought to identify the relationship between VEGF-A levels and outcomes through systematic review. We searched electronic databases and meeting proceedings for randomized controlled trials (RCTs) comparing the addition of bevacizumab to standard chemotherapy for breast cancer. RCTs were included if outcomes were presented separately according to VEGF-A plasma levels. Random-effects model were applied to calculate the pooled hazard ratios for progression-free survival, event-free survival (EFS), comprising disease recurrence, progression or any-cause death, and overall survival (OS), with respective confidence intervals (95 % CI). High and low VEGF-A levels subgroups followed each trial definition, and results were compared using the interaction test. Heterogeneity was calculated using χ (2) test (I (2)). Three trials enrolled a total of 3748 patients. 1713 patients had baseline VEGF-A levels in plasma available for assessment and were included. One trial added bevacizumab in the adjuvant setting (N = 2591) and two on first-line metastatic disease with taxane-based therapy (N = 1160) There was no interaction between VEGF-A levels and study setting (adjuvant vs. first line therapy). Bevacizumab improved PFS of patients with above median VEGF-A plasma levels (HR 0.56; 95 % CI 0.43-0.73; P < 0.001; I (2) = 0 %), but not of those with below median VEGF-A levels (HR 0.89; 95 % CI 0.68-1.15; P = 0.37; I (2) = 0 %), with relevant differences between these two groups, P-for interaction = 0.02. The same happened with EFS (VEGF-A above median HR 0.62; 95 % CI 0.39-0.79; P < 0.001; I (2) = 11 %; below median HR 0.89; 95 % CI 0.71-1.14; P = 0.98; I (2) = 17 %; P-for interaction = 0.03). OS data were not available. VEGF-A level is a reasonable candidate biomarker for bevacizumab in the treatment of breast cancer. Further studies have to confirm its surrogacy in overall survival and in other scenarios including other anti-angiogenic therapies.
Collapse
Affiliation(s)
- Lucas Vieira dos Santos
- Departamento de Oncologia Clinica, Hemomed Instituto de Oncologia e Hematologia, São Paulo, Brazil
| | | | | | | |
Collapse
|
49
|
Diéras V, Wildiers H, Jassem J, Dirix LY, Guastalla JP, Bono P, Hurvitz SA, Gonçalves A, Romieu G, Limentani SA, Jerusalem G, Lakshmaiah KC, Roché H, Sánchez-Rovira P, Pienkowski T, Seguí Palmer MÁ, Li A, Sun YN, Pickett CA, Slamon DJ. Trebananib (AMG 386) plus weekly paclitaxel with or without bevacizumab as first-line therapy for HER2-negative locally recurrent or metastatic breast cancer: A phase 2 randomized study. Breast 2015; 24:182-90. [PMID: 25747197 DOI: 10.1016/j.breast.2014.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/31/2014] [Accepted: 11/05/2014] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION This phase 2 randomized study evaluated trebananib (AMG 386), a peptide-Fc fusion protein that inhibits angiogenesis by neutralizing the interaction of angiopoietin-1 and -2 with Tie2, in combination with paclitaxel with or without bevacizumab in previously untreated patients with HER2-negative locally recurrent/metastatic breast cancer. METHODS Patients received paclitaxel 90 mg/m(2) once weekly (3-weeks-on/1-week-off) and were randomly assigned 1:1:1:1 to also receive blinded bevacizumab 10 mg/kg once every 2 weeks plus either trebananib 10 mg/kg once weekly (Arm A) or 3 mg/kg once weekly (Arm B), or placebo (Arm C); or open-label trebananib 10 mg/kg once a week (Arm D). Progression-free survival was the primary endpoint. RESULTS In total, 228 patients were randomized. Median estimated progression-free survival for Arms A, B, C, and D was 11.3, 9.2, 12.2, and 10 months, respectively. Hazard ratios (95% CI) for Arms A, B, and D versus Arm C were 0.98 (0.61-1.59), 1.12 (0.70-1.80), and 1.28 (0.79-2.09), respectively. The objective response rate was 71% in Arm A, 51% in Arm B, 60% in Arm C, and 46% in Arm D. The incidence of grade 3/4/5 adverse events was 71/9/4%, 61/14/5%, 62/16/3%, and 52/4/7% in Arms A/B/C/D. In Arm D, median progression-free survival was 12.8 and 7.4 months for those with high and low trebananib exposure (AUCss ≥ 8.4 versus < 8.4 mg·h/mL), respectively. CONCLUSIONS There was no apparent prolongation of estimated progression-free survival with the addition of trebananib to paclitaxel and bevacizumab at the doses tested. Toxicity was manageable. Exposure-response analyses support evaluation of combinations incorporating trebananib at doses > 10 mg/kg in this setting. TRIAL REGISTRATION ClinicalTrials.gov, NCT00511459.
Collapse
Affiliation(s)
| | | | | | - Luc Y Dirix
- General Hospital Sint-Augustinus, Antwerp, Belgium.
| | | | - Petri Bono
- Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | | | | | | - K C Lakshmaiah
- Kidwai Memorial Institute of Oncology, Bangalore, India.
| | | | | | | | | | - Ai Li
- Amgen Inc., Thousand Oaks, CA, USA.
| | | | | | | |
Collapse
|
50
|
|