1
|
Kheraldine H, Hassan AF, Saeed S, Merhi M, Mateo JM, Ulamec M, Peric-Balja M, Vranic S, Al-Thawadi H, Moustafa AEA. Neratinib and metformin: A novel therapeutic approach against HER2-Positive Breast Cancer. Biomed Pharmacother 2025; 187:118034. [PMID: 40252335 DOI: 10.1016/j.biopha.2025.118034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND HER2-positive breast cancer (BC) is highly aggressive with a poor prognosis. It is driven by HER2 oncoprotein activation/crosstalk with other receptors like EGFR/(HER1), HER3, and HER4, in addition to IGF-1R, making these receptors ideal therapeutic targets as they are expressed/overexpressed in this subtype. We postulated that targeting HER2 and IGF-1R together is a promising therapy for HER2-positive BC. Thus, we explored the outcome of a novel combination treatment using neratinib, a pan-HER inhibitor, and metformin, an IGF-1R inhibitor, on HER2-positive BC cells. METHODS In this investigation, we used cellular and molecular biology techniques in addition to an angiogenesis model and tissue microarray analysis. RESULTS Our data revealed that this combination therapy significantly reduced cell viability compared to individual treatments and exhibited a synergistic effect in HER2-positive BC cells. Moreover, the combination disrupted cell cycle progression and inhibited colony formation, and invasion of HER2-positive BC cells; this is accompanied by the deregulation of HER1-3 and IGF-1R expression patterns, in addition to Caspase-3, BCL2, Fascin, and Vimentin. Moreover, key regulator molecular pathways, including, ERK1/2, AKT, p38 MAPK, and mTOR, were significantly downregulated upon treatment with neratinib and metformin combination. Additionally, our data pointed out that neratinib and metformin combination inhibited angiogenesis, in-ovo, an important biological event in cancer progression. Finally, using a cohort of 55 HER2-positive BC samples, we revealed that HER2 and IGF-1R are co-expressed in most of the cases. CONCLUSIONS These findings suggest that neratinib and metformin combination can present a promising strategy for targeting multiple pathways in HER2-positive BC.
Collapse
Affiliation(s)
- Hadeel Kheraldine
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Arij Fouzat Hassan
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Sumayyah Saeed
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Jericha Miles Mateo
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Monika Ulamec
- Clinical Department of Pathology and Cytology Ljudevit Jurak, Sister of Charity University Hospital Center, Zagreb, Croatia; Department of Pathology and Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Melita Peric-Balja
- Oncological Pathology Department, Ljudevit Jurak Clinical Department of Pathology and Cytology, Sister of Charity University Hospital Center, Zagreb, Croatia
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Hamda Al-Thawadi
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar; Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, Canada; ABS Research Review & Consultation, Montreal, QC, Canada.
| |
Collapse
|
2
|
Zhang X, Chen X, Fu Y, Zhou H, Lin Y. Study on heterogeneity of vascularity and cellularity via multiparametric MRI habitat imaging in breast cancer. BMC Med Imaging 2025; 25:159. [PMID: 40361010 PMCID: PMC12070691 DOI: 10.1186/s12880-025-01698-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND This study aimed to visually analyze the heterogeneity of vascularity and cellularity across different sub-regions of breast cancer using habitat imaging (HI) to predict human epidermal growth factor receptor 2 (HER2) expression and evaluate the effectiveness of neoadjuvant therapy (NAT) in breast cancer patients. METHODS A retrospective analysis was conducted on 76 patients diagnosed with breast cancer. Diffusion-weighted imaging (DWI) and dynamic contrast-enhanced MRI (DCE-MRI) sequences were utilized to acquire MR images. Apparent diffusion coefficient (ADC), Ktrans, Kep, and Ve values were measured for each sub-region, and the percentage of each sub-region relative to the total lesion was calculated. Statistical analyses, including t-tests, rank-sum tests, chi-square tests, and Spearman correlation, were performed. RESULTS Three distinct sub-regions within breast cancer lesions were identified through HI, characterized physiologically as: low vascularity-high cellularity (LV-HC), low vascularity-low cellularity (LV-LC), and high vascularity-low cellularity (HV-LC). Significant differences were observed in the proportions of these tumor sub-regions between HER2-positive and HER2-negative breast cancers. Additionally, HER2-low and HER2-zero breast cancers demonstrated statistical differences in the second sub-region (LV-LC). Furthermore, the proportion of the first sub-region (LV-HC) was negatively correlated with the efficacy of NAT in breast cancer patients. CONCLUSIONS Habitat imaging can identify distinct sub-regions within breast cancer lesions, providing a noninvasive imaging biomarker for predicting HER2 expression levels and assessing the efficacy of NAT in breast cancer patients.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, No. 69, Dongxiabei Road, Jinping District, Shantou, Guangdong Province, 515041, China
| | - Xiaoyan Chen
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, No. 69, Dongxiabei Road, Jinping District, Shantou, Guangdong Province, 515041, China
| | - Yao Fu
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, No. 69, Dongxiabei Road, Jinping District, Shantou, Guangdong Province, 515041, China
| | - Han Zhou
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, No. 69, Dongxiabei Road, Jinping District, Shantou, Guangdong Province, 515041, China
| | - Yan Lin
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, No. 69, Dongxiabei Road, Jinping District, Shantou, Guangdong Province, 515041, China.
| |
Collapse
|
3
|
Wang Y, Huang M, Zhou X, Li H, Ma X, Sun C. Potential of natural flavonoids to target breast cancer angiogenesis (review). Br J Pharmacol 2025; 182:2235-2258. [PMID: 37940117 DOI: 10.1111/bph.16275] [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: 07/03/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023] Open
Abstract
Angiogenesis is the process by which new blood vessels form and is required for tumour growth and metastasis. It helps in supplying oxygen and nutrients to tumour cells and plays a crucial role in the local progression and distant metastasis of, and development of treatment resistance in, breast cancer. Tumour angiogenesis is currently regarded as a critical therapeutic target; however, anti-angiogenic therapy for breast cancer fails to produce satisfactory results, owing to issues such as inconsistent efficacy and significant adverse reactions. As a result, new anti-angiogenic drugs are urgently needed. Flavonoids, a class of natural compounds found in many foods, are inexpensive, widely available, and exhibit a broad range of biological activities, low toxicity, and favourable safety profiles. Several studies find that various flavonoids inhibit angiogenesis in breast cancer, indicating great therapeutic potential. In this review, we summarize the role of angiogenesis in breast cancer and the potential of natural flavonoids as anti-angiogenic agents for breast cancer treatment. We discuss the value and significance of nanotechnology for improving flavonoid absorption and utilization and anti-angiogenic effects, as well as the challenges of using natural flavonoids as drugs. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Yuetong Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengge Huang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintong Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Xiaoran Ma
- Department of Oncology, Linyi People's Hospital, Linyi, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
4
|
Bach Y, Sharma D, Aoyama H, Ma LX, Barron CC, Wang X, Akhtar S, Yang Y, St Bernard A, McLaughlin R, Megid TBC, Farooq AR, Chen EX, Jang RWJ, Elimova E. Ramucirumab and paclitaxel in second or greater lines of therapy in patients with HER2-positive gastroesophageal cancer: a single center study. Oncologist 2025; 30:oyaf037. [PMID: 40152313 PMCID: PMC11950916 DOI: 10.1093/oncolo/oyaf037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2) overexpression is present in approximately 20-25 of patients with advanced gastroesophageal adenocarcinoma (GEA). Upon progression on 1st line therapy, ramucirumab and paclitaxel (rampac) is given in ≥2 line setting regardless of HER2 status. We aim to assess whether ramucirumab is associated with better survival in HER2 positive(+) pts compared to those with HER2(-) disease. METHODS We reviewed all consecutive adult patients with metastatic/unresectable GEA who were treated with rampac for ≥2nd line therapy at Princess Margaret Cancer Centre from 2010 to 2021. Progression free survival (PFS) and overall survival (OS) were defined as time from starting rampac to progression or death and estimated using the Kaplan-Meier method. RESULTS There were 126 patients who received rampac following progression of 1st line chemotherapy, 96(76%) were male. The age at time of presentation and starting rampac was 59.0 ± 10.3 years and 59.9 ± 10.3 years, respectively. At the time of diagnosis, 32(25%) patients were HER2+. The majority of patients (n = 99;78%) received rampac in the 2L line setting compared to 28(22%) patients who received it in the 3rd/4th line setting. The median PFS and OS for HER2 + pts were 3.6 months and 9.4 months, respectively, which were similar to HER2- patients (median PFS = 3.6 months; median OS = 8.2 months). There was no statistically significant association between HER2 positivity and PFS (adjusted hazards ratio (HR) = 0.76, 95% confidence interval (CI) 0.48-1.22, P = .26), nor OS (adjusted HR = 0.88, 95% CI, 0.55-1.41, P = .59). CONCLUSION Rampac remains a valid treatment option for patients who are unable to participate in trials or do not have access to further HER2-directed therapy beyond first line.
Collapse
Affiliation(s)
- Yvonne Bach
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Divya Sharma
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Hiroko Aoyama
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Lucy X Ma
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Carly C Barron
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Xin Wang
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Sokaina Akhtar
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Yahan Yang
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Alana St Bernard
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Ronan McLaughlin
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Thais B C Megid
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Abdul R Farooq
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Eric X Chen
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Raymond W J Jang
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| | - Elena Elimova
- Princess Margaret Cancer Centre, 700 University Avenue, Toronto, ON, Canada M5G 1X6
| |
Collapse
|
5
|
Qian X, Jin X, He J, Zhang J, Hu S. Exploring lipidomic profiles and their correlation with hormone receptor and HER2 status in breast cancer. Oncol Lett 2025; 29:34. [PMID: 39512509 PMCID: PMC11542162 DOI: 10.3892/ol.2024.14781] [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: 05/24/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024] Open
Abstract
Dysregulated lipid metabolism promotes the progression of various cancer types, including breast cancer. The present study aimed to explore the lipidomic profiles of patients with breast cancer, providing insights into the correlation between lipid compositions and tumor subtypes characterized by hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) status. Briefly, 30 patients with breast cancer were categorized into four groups based on their HR and HER2 status: HR+ no HER2 expression (HER2-0), HR+ HER2-low; HR+ HER2-positive (pos) and HR- HER2-pos. The lipidomic profiles of these patients were analyzed using high-throughput liquid chromatography-mass spectrometry. The data were processed through principal component analysis (PCA), partial least squares-discriminant analysis (PLS-DA) and random forest (RF) classification to assess the lipidomic variations and significant lipid features among these groups. The profiles of the lipids, particularly triglycerides (TG) such as TG(16:0-18:1-18:1)+NH4, were significantly different across the groups. PCA and PLS-DA identified unique lipid profiles in the HR+ HER2-pos and HR+ HER2-0 groups, while RF highlighted phosphatidylinositol-3,4,5-trisphosphate(21:2)+NH4 as a crucial lipid feature for accurate patient grouping. Advanced statistical analysis showed significant correlations between lipid carbon chain length and the number of double bonds within the classifications, providing insights into the role of structural lipid properties in tumor biology. Additionally, a clustering heatmap and network analysis indicated significant lipid-lipid interactions. Pathway enrichment analysis showed critical biological pathways, such as the 'Assembly of active LPL and LIPC lipase complexes', which has high enrichment ratio and statistical significance. In conclusion, the present study underscored that lipidomic profiling is crucial in understanding the metabolic alterations associated with different breast cancer subtypes. These findings highlighted specific lipid features and interactions that may serve as potential biomarkers for breast cancer classification and target pathways for therapeutic intervention. Furthermore, advanced lipidomic analyses can be integrated to decipher complex biological data, offering a foundation for further research into the role of lipid metabolism in cancer progression.
Collapse
Affiliation(s)
- Xiaojun Qian
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Xiaolin Jin
- Health Management Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Jiaying He
- Health Management Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Junjing Zhang
- Health Management Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Shan Hu
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
6
|
Chen X, Huang J, Xie X, Chen L, Lan X, Song L, Bai X, Du C. Apatinib and trastuzumab-based chemotherapy for heavily treated primary trastuzumab-resistant metastatic breast cancer. J Cancer Res Ther 2024; 20:1991-1996. [PMID: 39792408 DOI: 10.4103/jcrt.jcrt_979_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/28/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND The low incidence and poor prognosis primary trastuzumab resistance (PTR) in HER2-positive breast cancer has limited research into possible treatments. Thus, it remains unclear whether this group of patients could benefit from nontargeting HER2 antiangiogenic therapy. PATIENTS AND METHODS We collected the medical data for HER2-positive patients with PTR who received apatinib 250 mg and trastuzumab-based chemotherapy (ATBC) between March 18, 2017, and March 31, 2022. All patients had progressed on ≥2 anti-HER2 treatments, including trastuzumab and small molecular tyrosine kinase inhibitors. We evaluated tumor response and safety profiles to ATBC over a median follow-up time of 34.5 months. RESULTS A total of 198 consecutively HER2-positive metastatic breast cancer patients were reviewed; 20 were PTR and received ATBC. The clinical benefit rate of the total cohort was 55.0%. No patient showed a complete response. The median PFS and overall survival (OS) of the entire cohort was 5.7 months (95% CI 2.9-8.5) and 24.6 months (95% CI 6.9-42.4), respectively. The estimated 2-year survival rate was 46.7% (95% CI 38.4-81.6%). The most common nonhematologic adverse events were hypertension (70.0%), hand-foot skin reaction (55.0%), proteinuria (40.0%), and cardiovascular decrease of LVEF (20.0%). No new toxicities were observed. CONCLUSION ATBC had favorable effects for PTR breast cancer patients in later line treatment. The toxicity of the triple-combination regimen was tolerable; thus, further research should focus on identifying PTR patients who could benefit from ATBC.
Collapse
Affiliation(s)
- Xuelian Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Sarf EA, Dyachenko EI, Bel’skaya LV. The Role of Salivary Vascular Endothelial Growth Factor A, Cytokines, and Amino Acids in Immunomodulation and Angiogenesis in Breast Cancer. Biomedicines 2024; 12:1329. [PMID: 38927536 PMCID: PMC11201966 DOI: 10.3390/biomedicines12061329] [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: 05/25/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
In this work, we focused on the analysis of VEGF content in saliva and its relationship with pro-inflammatory cytokines and amino acids involved in immunomodulation and angiogenesis in breast cancer. The study included 230 breast cancer patients, 92 patients with benign breast disease, and 59 healthy controls. Before treatment, saliva samples were obtained from all participants, and the content of VEGF and cytokines in saliva was determined by an enzyme-linked immunosorbent assay, as well as the content of amino acids by high-performance liquid chromatography. It was found that VEGF was positively correlated with the level of pro-inflammatory cytokines IL-1β (r = 0.6367), IL-6 (r = 0.3813), IL-8 (r = 0.4370), and IL-18 (r = 0.4184). Weak correlations were shown for MCP-1 (r = 0.2663) and TNF-α (r = 0.2817). For the first time, we demonstrated changes in the concentration of VEGF and related cytokines in saliva in different molecular biological subtypes of breast cancer depending on the stage of the disease, differentiation, proliferation, and metastasis to the lymph nodes. A correlation was established between the expression of VEGF and the content of aspartic acid (r = -0.3050), citrulline (r = -0.2914), and tryptophan (r = 0.3382) in saliva. It has been suggested that aspartic acid and citrulline influence the expression of VEGF via the synthesis of the signaling molecule NO, and then tryptophan ensures tolerance of the immune system to tumor cells.
Collapse
Affiliation(s)
| | | | - Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 14, Tukhachevsky Str., 644099 Omsk, Russia; (E.A.S.); (E.I.D.)
| |
Collapse
|
8
|
Kawakami T, Yamazaki K. Recent Progress in Treatment for HER2-Positive Advanced Gastric Cancer. Cancers (Basel) 2024; 16:1747. [PMID: 38730700 PMCID: PMC11083522 DOI: 10.3390/cancers16091747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Human epidermal receptor (HER) 2-positive advanced gastric cancer is one of the major subtypes of gastric cancer, accounting for ~20% of all cases. Although combination therapy with trastuzumab and chemotherapy provides meaningful survival benefit, clinical trials targeting HER2 have failed to demonstrate clinical benefits in first- or subsequent-line treatment. Trastuzumab deruxtecan, an antibody-drug conjugate, has shown positive results even in later-line treatment and has become new standard treatment. In first-line therapy, combination therapy with pembrolizumab and trastuzumab plus chemotherapy demonstrated a dramatic response rate. Therefore, the FDA rapidly approved it without waiting for the results of survival time. The emergence of combination therapy including immunotherapy with HER2-targeting agents and the development of HER2 targeting agents with or without immunotherapy have been advancing for treating HER2-positive gastric cancer. In this review, we will discuss the current status of treatment development and future perspectives for HER2-positive gastric cancer.
Collapse
Affiliation(s)
- Takeshi Kawakami
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka 411-0934, Japan;
| | | |
Collapse
|
9
|
Aragón-Franco R, Ruiz-Manzano RA, Nava-Castro KE, Del Rìo Araiza VH, Garay-Canales CA, Pérez-Torres A, Chacón-Salinas R, Girón-Pérez MI, Morales-Montor J. Convergence between helminths and breast cancer: intratumoral injection of the excretory/secretory antigens of the human parasite Toxocara canis (EST) increase lung macro and micro metastasis. Front Immunol 2024; 15:1332933. [PMID: 38576624 PMCID: PMC10993691 DOI: 10.3389/fimmu.2024.1332933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/21/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Worldwide, breast cancer is the most important cancer in incidence and prevalence in women. Different risk factors interact to increase the probability of developing it. Biological agents such as helminth parasites, particularly their excretory/secretory antigens, may play a significant role in tumor development. Helminths and their antigens have been recognized as inducers or promoters of cancer due to their ability to regulate the host's immune response. Previously in our laboratory, we demonstrated that chronic infection by Toxocara canis increases the size of mammary tumors, affecting the systemic response to the parasite. However, the parasite does not invade the tumor, and we decided to study if the excretion/secretion of antigens from Toxocara canis (EST) can affect the progression of mammary tumors or the pathophysiology of cancer which is metastasis. Thus, this study aimed to determine whether excretion/secretion T. canis antigens, injected directly into the tumor, affect tumor growth and metastasis. Methods We evaluated these parameters through the monitoring of the intra-tumoral immune response. Results Mice injected intratumorally with EST did not show changes in the size and weight of the tumors; although the tumors showed an increased microvasculature, they did develop increased micro and macro-metastasis in the lung. The analysis of the immune tumor microenvironment revealed that EST antigens did not modulate the proportion of immune cells in the tumor, spleen, or peripheral lymph nodes. Macroscopic and microscopic analyses of the lungs showed increased metastasis in the EST-treated animals compared to controls, accompanied by an increase in VEGF systemic levels. Discussion Thus, these findings showed that intra-tumoral injection of T. canis EST antigens promote lung metastasis through modulation of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Raúl Aragón-Franco
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Ciudad de México, Mexico
| | - Rocío Alejandra Ruiz-Manzano
- Laboratorio de Neuroinmunoendocrinología, Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Karen Elizabeth Nava-Castro
- Laboratorio de Biología y Química Atmosférica, Departamento de Ciencias Ambientales, Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Víctor Hugo Del Rìo Araiza
- Laboratorio de Interacciones Endocrinoinmunitarias en Enfermedades Parasitarias, Facultad de Medicina Veterinaria y Zootecnia, Departamento de Parasitología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Claudia Angelica Garay-Canales
- Laboratorio de Neuroinmunoendocrinología, Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Armando Pérez-Torres
- Departamento de Biologia Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Romel Chacón-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Ciudad de México, Mexico
| | - Manuel Iván Girón-Pérez
- Laboratorio de Inmunotoxicología, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepic, Nayarit, Mexico
| | - Jorge Morales-Montor
- Laboratorio de Neuroinmunoendocrinología, Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
10
|
Kim IH. Emerging Targets for Systemic Treatment of Gastric Cancer: HER2 and Beyond. J Gastric Cancer 2024; 24:29-56. [PMID: 38225765 PMCID: PMC10774754 DOI: 10.5230/jgc.2024.24.e6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/17/2024] Open
Abstract
In recent years, remarkable progress has been made in the molecular profiling of gastric cancer. This progress has led to the development of various molecular classifications to uncover subtype-specific dependencies that can be targeted for therapeutic interventions. Human epidermal growth factor receptor 2 (HER2) is a crucial biomarker for advanced gastric cancer. The recent promising results of novel approaches, including combination therapies or newer potent agents such as antibody-drug conjugates, have once again brought attention to anti-HER2 targeted treatments. In HER2-negative diseases, the combination of cytotoxic chemotherapy and programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitors has become the established standard of care in first-line settings. In the context of gastric cancer, potential biomarkers such as PD-L1 expression, Epstein-Barr virus, microsatellite instability, and tumor mutational burden are being considered for immunotherapy. Recently, promising results have been reported in studies on anti-Claudin18.2 and fibroblast growth factor receptor 2 treatments. Currently, many ongoing trials are aimed at identifying potential targets using novel approaches. Further investigations will be conducted to enhance the progress of these therapies, addressing challenges such as primary and acquired resistance, tumor heterogeneity, and clonal evolution. We believe that these efforts will improve patient prognoses. Herein, we discuss the current evidence of potential targets for systemic treatment, clinical considerations, and future perspectives.
Collapse
Affiliation(s)
- In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Gastric Cancer Centre, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea,.
| |
Collapse
|
11
|
Kim CG, Jung M, Kim HS, Lee CK, Jeung HC, Koo DH, Bae WK, Zang DY, Kim BJ, Kim H, Yun UJ, Che J, Park S, Kim TS, Kwon WS, Park J, Cho SW, Nam CM, Chung HC, Rha SY. Trastuzumab Combined With Ramucirumab and Paclitaxel in Patients With Previously Treated Human Epidermal Growth Factor Receptor 2-Positive Advanced Gastric or Gastroesophageal Junction Cancer. J Clin Oncol 2023; 41:4394-4405. [PMID: 37364218 DOI: 10.1200/jco.22.02122] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/20/2023] [Accepted: 05/01/2023] [Indexed: 06/28/2023] Open
Abstract
PURPOSE Trastuzumab-containing chemotherapy is the recommended first-line regimen for human epidermal growth factor receptor 2 (HER2)-positive advanced gastric or gastroesophageal junction (G/GEJ) cancer. We evaluated the safety and efficacy of trastuzumab combined with ramucirumab and paclitaxel as second-line treatment for HER2-positive G/GEJ cancer. PATIENTS AND METHODS Patients with HER2-positive advanced G/GEJ cancer who progressed after first-line treatment with trastuzumab-containing chemotherapy were enrolled from five centers in the Republic of Korea. Patients were administered a 28-day cycle of trastuzumab (once on days 1, 8, 15, and 22: 2 mg/kg followed by 4 mg/kg loading dose), ramucirumab (once on days 1 and 15: 8 mg/kg), and paclitaxel (once on days 1, 8, and 15: dose level 1, 80 mg/m2; or dose level -1, 70 mg/m2). Phase II was conducted with the recommended phase II dose (RP2D). Primary end points were determination of RP2D during phase Ib and investigator-assessed progression-free survival (PFS) in patients treated with RP2D. RESULTS Dose-limiting toxicity at dose level 1 was not documented during phase Ib, and a full dose combination was selected as the RP2D. Among 50 patients with a median follow-up duration of 27.5 months (95% CI, 17.4 to 37.6), median PFS and overall survival were 7.1 months (95% CI, 4.8 to 9.4) and 13.6 months (95% CI, 9.4 to 17.7), respectively. Objective response rate was 54% (27 of 50, including one complete response), and disease control rate was 96% (48 of 50). Loss of HER2 expression was observed in 34.8% (8 of 23) patients after first-line treatment, and no definite association between HER2 expression and the outcome was revealed. Safety profiles were consistent with previous reports. CONCLUSION Trastuzumab combined with ramucirumab and paclitaxel showed appreciable efficacy with manageable safety profiles in patients with previously treated HER2-positive G/GEJ cancer.
Collapse
Affiliation(s)
- Chang Gon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Song Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hei-Cheul Jeung
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong-Hoe Koo
- Divison of Hematology-Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Woo Kyun Bae
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Dae Young Zang
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Bum Jun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Un-Jung Yun
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jingmin Che
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- MD Biolab Co, Ltd, Seoul, Republic of Korea
| | - Sejung Park
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae Soo Kim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woo Sun Kwon
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Juin Park
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Woo Cho
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Young JR, Ressler JA, Shiroishi MS, Mortimer JE, Schmolze D, Fitzgibbons M, Chen BT. Association of Relative Cerebral Blood Volume from Dynamic Susceptibility Contrast-Enhanced Perfusion MR with HER2 Status in Breast Cancer Brain Metastases. Acad Radiol 2023; 30:1816-1822. [PMID: 36549990 DOI: 10.1016/j.acra.2022.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/28/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
RATIONALE AND OBJECTIVES With the development of HER2-directed therapies, identifying non-invasive imaging biomarkers of HER2 expression in breast cancer brain metastases has become increasingly important. The purpose of this study was to investigate whether relative cerebral blood volume (rCBV) from dynamic susceptibility contrast-enhanced (DSC) perfusion MR could help identify the HER2 status of breast cancer brain metastases. MATERIALS AND METHODS With IRB approval for this HIPAA-compliant cross-sectional study and a waiver of informed consent, we queried our institution's electronic medical record to derive a cohort of 14 histologically proven breast cancer brain metastases with preoperative DSC perfusion MR and HER2 analyses of the resected/biopsied brain specimens from 2011-2021. The rCBV of the lesions was measured and compared using Mann-Whitney tests. Receiver operating characteristic analyses were performed to evaluate the performance of rCBV in identifying HER2 status. RESULTS The study cohort was comprised of 14 women with a mean age of 56 years (range: 32-81 years) with a total of 14 distinct lesions. The rCBV of HER2-positive breast cancer brain metastases was significantly greater than the rCBV of HER2-negative lesions (8.02 vs 3.97, U=48.00, p=0.001). rCBV differentiated HER2-positive lesions from HER2-negative lesions with an area under the curve of 0.98 (standard error=0.032, p<0.001). The accuracy-maximizing rCBV threshold (4.8) was associated with an accuracy of 93% (13/14), a sensitivity of 100% (7/7), and a specificity of 86% (6/7). CONCLUSION rCBV may assist in identifying the HER2 status of breast cancer brain metastases, if validated in a large prospective trial.
Collapse
Affiliation(s)
- Jonathan R Young
- Division of Neuroradiology, Department of Radiology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd, Duarte, California, 91010.
| | - Julie A Ressler
- Division of Neuroradiology, Department of Radiology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd, Duarte, California, 91010
| | - Mark S Shiroishi
- Division of Neuroradiology, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Joanne E Mortimer
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Daniel Schmolze
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Mariko Fitzgibbons
- Division of Neuroradiology, Department of Radiology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd, Duarte, California, 91010
| | - Bihong T Chen
- Division of Neuroradiology, Department of Radiology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd, Duarte, California, 91010
| |
Collapse
|
13
|
Qu F, Liu Q, Lu R, Li W. Disitamab Vedotin (RC48) combined with bevacizumab for treatment of HR-negative/HER2-positive metastatic breast cancer with liver and brain involvement: A case report. Front Oncol 2023; 13:1245701. [PMID: 37711199 PMCID: PMC10498115 DOI: 10.3389/fonc.2023.1245701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Background The overexpression of human epidermal growth factor receptor 2 (HER2) is strongly correlated with an elevated risk of developing distant metastases, particularly brain metastases, in breast cancer (BC) cases. RC48 (also known as Disitamab vedotin), represents a promising antibody-drug conjugate (ADC), that comprises three well-defined components: hertuzumab against the prominent tumor target-HER2, monomethyl auristatin E (MMAE) and a cleavable linker. Preclinical studies have demonstrated its robust antitumor activity in BC patient-derived xenograft models with HER2-positive or HER2-low expression. Additionally, antiangiogenic drugs like bevacizumab have shown potential efficacy on advanced BC via inhibiting pathological neovascularizationits. Case presentation Here, we will share our experience in treating a 49-year-old woman initially diagnosed with stage IV breast cancer characterized by hormone receptor (HR)-negativity and HER2-positivity. This complex case entailed brain and liver metastases, and the patient exhibited resistance to various HER2-targeted treatment regimens. Finally, the patient received RC48 plus bevacizumab as the advanced forth-line treatment, which was well tolerated with no observed toxicities. Subsequent radiological assessments revealed remarkable regression in the brain metastatic lesions, classified as having partial response based on the RECIST 1.1 system. The period of progression-free survival (PFS) was 7 months. Conclusion The present study underscores the efficacy of systemic treatment with RC48 in conjunction, showcasing substantial enhancement in both radiographic indicators and clinical symptomatology among patients with brain metastatic breast cancer (BMBC). More specifically, the sequential application of ADCs in combination with antiangiogenics presents a novel avenue for advancing the treatment landscape of metastatic BC.
Collapse
Affiliation(s)
- Fei Qu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Qian Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Rongrong Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Radford M, Abushukair H, Hentzen S, Cavalcante L, Saeed A. Targeted and Immunotherapy Approaches in HER2-Positive Gastric and Gastroesophageal Junction Adenocarcinoma: A New Era. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2023; 6:150-157. [PMID: 37637236 PMCID: PMC10448730 DOI: 10.36401/jipo-22-36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/28/2023] [Accepted: 05/05/2023] [Indexed: 08/29/2023]
Abstract
HER2-targeted therapy with the HER2 monoclonal antibody trastuzumab has achieved impressive outcomes in the first-line settings of patients with advanced gastric and gastroesophageal junction (GEJ) adenocarcinoma overexpressing HER2. However, considering that a substantial proportion of those patients eventually relapses, as well as the relatively limited performance of those agents in second-line settings, a deeper understanding of resistance mechanisms is needed for enhanced guidance for patients' therapeutic selection in the second-line setting and beyond. In this review, we highlight trastuzumab's (HER2-targeting agent) performance in patients with gastric or GEJ cancer, with insight into mechanisms of resistance. We also discuss the new integration of PD-1 inhibitor pembrolizumab into the trastuzumab for gastric cancer frontline regimen, the latest addition of trastuzumab deruxtecan to the treatment armamentarium, and the potential of pipeline HER2-targeting approaches and combinations in patients with gastric or GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Maluki Radford
- Department of Medicine, Division of Medical Oncology, Kansas University Medical Center, Kansas City, KS, USA
| | - Hassan Abushukair
- Department of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Stijn Hentzen
- Department of Medicine, Division of Medical Oncology, Kansas University Medical Center, Kansas City, KS, USA
| | - Ludimila Cavalcante
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Han MR, Park AY, Seo BK, Bae MS, Kim JS, Son GS, Lee HY, Chang YW, Cho KR, Song SE, Woo OH, Ju HY, Oh H. Association between vascular ultrasound features and DNA sequencing in breast cancer: a preliminary study. Discov Oncol 2023; 14:52. [PMID: 37120792 PMCID: PMC10149538 DOI: 10.1007/s12672-023-00657-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/14/2023] [Indexed: 05/01/2023] Open
Abstract
There are few radiogenomic studies to correlate ultrasound features of breast cancer with genomic changes. We investigated whether vascular ultrasound phenotypes are associated with breast cancer gene profiles for predicting angiogenesis and prognosis. We prospectively correlated quantitative and qualitative features of microvascular ultrasound (vascular index, vessel morphology, distribution, and penetrating vessel) and contrast-enhanced ultrasound (time-intensity curve parameters and enhancement pattern) with genomic characteristics in 31 breast cancers. DNA obtained from breast tumors and normal tissues were analyzed using targeted next-generation sequencing of 105 genes. The single-variant association test was used to identify correlations between vascular ultrasound features and genomic profiles. Chi-square analysis was used to detect single nucleotide polymorphisms (SNPs) associated with ultrasound features by estimating p values and odds ratios (ORs). Eight ultrasound features were significantly associated with 9 SNPs (p < 0.05). Among them, four ultrasound features were positively associated with 5 SNPs: high vascular index with rs1136201 in ERBB2 (p = 0.04, OR = 7.75); large area under the curve on contrast-enhanced ultrasound with rs35597368 in PDGFRA (p = 0.04, OR = 4.07); high peak intensity with rs35597368 in PDGFRA (p = 0.049, OR = 4.05) and rs2305948 in KDR (p = 0.04, OR = 5.10); and long mean transit time with rs2275237 in ARNT (p = 0.02, OR = 10.25) and rs755793 in FGFR2 (p = 0.02, OR = 10.25). We identified 198 non-silent SNPs in 71 various cancer-related genes. Vascular ultrasound features can reflect genomic changes associated with angiogenesis and prognosis in breast cancer.
Collapse
Affiliation(s)
- Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Ah Young Park
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Bo Kyoung Seo
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan, Gyeonggi-do 15355 Republic of Korea
| | - Min Sun Bae
- Department of Radiology, Inha University Hospital and College of Medicine, Inhang-ro 27, Jung-gu, Incheon, 22332 Republic of Korea
| | - Jung Sun Kim
- Division of Hematology/Oncology, Department of Internal medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi-do Republic of Korea
| | - Gil Soo Son
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi-do Republic of Korea
| | - Hye Yoon Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi-do Republic of Korea
| | - Young Woo Chang
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi-do Republic of Korea
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sung Eun Song
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ok Hee Woo
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hye-Yeon Ju
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Hyunseung Oh
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi-do Republic of Korea
| |
Collapse
|
16
|
Huang J, Chen X, Guo J, Song L, Mu Y, Zhao H, Du C. Apatinib combined with trastuzumab and albumin-bound paclitaxel for treatment of HER2+ breast cancer with brain metastases resistant to anti-HER2 TKIs: A case report. Oncol Lett 2022; 25:56. [PMID: 36644147 PMCID: PMC9827463 DOI: 10.3892/ol.2022.13642] [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: 10/19/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Although human epidermal growth factor receptor 2 (HER2)-targeted therapy significantly improves the prognosis of patients with HER2-positive breast cancer, most patients with advanced breast cancer eventually progress due to drug resistance. At present, there is no standard treatment after patients become resistant to HER2-targeted therapy. Previous studies have indicated that anti-angiogenesis drugs have potential efficacy in the treatment of advanced breast cancer. The present study reported on a case of a pretreated patient with HER2-positive advanced breast cancer with brain metastases who developed resistance to multiple lines of HER2-targeted treatment. The patient was treated with apatinib combined with trastuzumab and albumin-bound paclitaxel. The patient achieved partial response to the third-line treatment with a progression-free survival of 9 months. After combination treatment, the symptoms of headache and vomiting were relieved and all the brain metastases were significantly reduced. The present case indicated that apatinib may have anti-tumor activity in patients with HER2-positive breast cancer with HER2-targeted drug resistance. The present case provides valuable information and may offer a new possibility for the treatment of patients with breast cancer with brain metastases who progressed after clinical treatment with small-molecule anti-HER2 tyrosine kinase inhibitor drugs.
Collapse
Affiliation(s)
- Jiayi Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, P.R. China
| | - Xiao Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, P.R. China
| | - Jinfeng Guo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, P.R. China
| | - Lin Song
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, P.R. China
| | - Yanxi Mu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, P.R. China
| | - Han Zhao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, P.R. China
| | - Caiwen Du
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518116, P.R. China,Correspondence to: Professor Caiwen Du, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 113 Baohe Road, Shenzhen, Guangdong 518116, P.R. China, E-mail:
| |
Collapse
|
17
|
He L, Shen X, Liu Y, Gao L, Wu J, Yu C, Li G, Wang X, Shao X. The reversal of anti-HER2 resistance in advanced HER2-positive breast cancer using apatinib: two cases reports and literature review. Transl Cancer Res 2022; 11:4206-4217. [PMID: 36523304 PMCID: PMC9745359 DOI: 10.21037/tcr-22-2483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/15/2022] [Indexed: 09/29/2024]
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2)-targeted treatment has yielded a notable clinical benefit in patients with HER2-positive breast cancer. However, nearly 50% of patients still suffer disease progression due to resistance to HER2-targeted therapy. After the failure of macromolecular monoclonal antibodies (mAbs) therapy, we can choose small molecule tyrosine kinase inhibitors (TKIs) to reverse HER2 resistance. When small molecule TKIs resistance, we can use mAb combined with small molecule TKI, or antibody-drug conjugates (ADCs) to reverse HER2 resistance. However, then due to the availability and price of ADCs, patients may not use them. Consequently, new therapeutic approaches are required to overcome HER2-targeted therapy resistance. Vascular endothelial growth factor and its receptors (VEGF/VEGFRs) promote tumor angiogenesis. They can also activate downstream signaling pathways to promote tumorigenesis. VEGFR is a key regulator of the tyrosine kinase signaling pathway and may be a potential target in HER2-positive breast cancer. Apatinib is a small molecule TKI that specifically binds to VEGFR2 and thus exerts an antitumor effect. Although there is no definite indication for apatinib in breast cancer, it has a good benefit in advanced gastric cancer. CASE DESCRIPTION The two patients we reported were both HER2-positive breast cancer who we followed for more than 10 years. After the failure of multi-line anti-HER2 treatment, apatinib combined with anti-HER2 treatment had PFS of 8.4 months and 10.6 months, respectively. One patient had grade 2 hand-foot syndrome. The other had grade 2 leukopenia and grade 2 thrombocytopenia, both of them improved after control. And the best response of them were PR and SD, respectively. CONCLUSIONS Our cases demonstrate that, in HER2-positive breast cancer patients with HER2-targeted resistance, apatinib may be able to reverse HER2 resistance. These two cases suggest an alternative method for clinical HER2-targeted treatment of drug-resistant breast cancer patients and provide new insights for future research.
Collapse
Affiliation(s)
- Libin He
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiabo Shen
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yiyuan Liu
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Gao
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiayi Wu
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Chang Yu
- Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Guangliang Li
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiying Shao
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
18
|
Kilinç E, Bahar AY. The Value of Intratumoral and Extratumoral Microvessel Density for the Tumor-dominated Area in the Endometrial Carcinoma. Appl Immunohistochem Mol Morphol 2022; 30:501-508. [PMID: 35731561 DOI: 10.1097/pai.0000000000001044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Microvessel density (MVD) measuring angiogenesis can influence clinicopathologic variables in endometrial carcinoma (EC). MVD is usually assessed in the densest vascular area the tumor, but the distinction between intratumoral and extratumoral MVD is not reported, and tumor-dominated area is not examined for MVD in the EC in the literature. MATERIALS AND METHODS A total of 151 cases with EC, which had hysterectomy from 2005 to 2020, were included. All histopathologic parameters were re-evaluated blindly. MVD was counted in the intratumoral (densest tumoral and vascular area) and extratumoral (periphery of the tumor, tumor invasion tip, densest tumoral, and vascular area) areas using immunohistochemical CD31 expression. RESULTS Large tumor size, deep myometrial invasion, high grade, nonendometrioid tumor type, cervix invasion, lymph node metastasis, human epidermal growth factor receptor 2 positivity, stage III to IV, substantial lymphovascular invasion, and overall survival had significant relations with intratumoral and extratumoral MVD ( P <0.05). Age and lymphoplasmacytic inflammation showed marginal significance for extratumoral MVD. Extratumoral and intratumoral MVD had high (near-perfect) agreement (κ=0.870, P =0.001). Cervix invasion, stage III to IV, high grade, intratumoral, and extratumoral MVD were correlated with lower overall survival in the multivariate analyses. CONCLUSION High MVD had an impact on the behavior and prognosis of EC and may be a potential indicator for antiangiogenic treatments for aggressive tumors. Although intratumoral and extratumoral MVD in the tumor-dominated area have mostly similar effects in our study, this situation can be cleared more by further investigations.
Collapse
Affiliation(s)
- Emine Kilinç
- Department of Pathology, Kahramanmaraş Sütçü İmam University School of Medicine, Kahramanmaraş, Turkey
| | | |
Collapse
|
19
|
Kim BJ, Jee HJ, Rha SY, Han HS, Ryu MH, Park SH, Kim JG, Bae WK, Lee KW, Oh DY, Byun JH, Kim DS, Suh YJ, An H, Zang DY. Ramucirumab plus paclitaxel as a second-line treatment in HER2-positive gastric cancer: subgroup analysis of a nationwide, real-world study in Korea (KCSG-ST19-16). Gastric Cancer 2022; 25:609-618. [PMID: 35015188 DOI: 10.1007/s10120-021-01276-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND A subgroup analysis of data from a nationwide study (KCSG-ST19-16) was performed to evaluate the efficacy and safety of second-line ramucirumab plus paclitaxel treatment in patients with human epidermal growth factor receptor 2 (HER2)-positive advanced gastric or gastro-esophageal junction (GEJ) adenocarcinoma. METHODS The KCSG-ST19-16 study enrolled a total of 1063 patients from 56 hospitals in South Korea with advanced gastric or GEJ adenocarcinoma, who had received second-line treatment with ramucirumab plus paclitaxel. HER2 status was known for 994 (93.5%) of these patients, who were thus included in the subgroup analysis. RESULTS In total, 163 of 994 patients (16.4%), had HER2-positive gastric or GEJ adenocarcinoma. The objective response rate to ramucirumab plus paclitaxel treatment was significantly higher in patients with HER2-positive disease compared to those with HER2-negative disease (23.0% [95% confidence interval (CI), 15.9-30.1] vs. 15.1% [95% CI, 12.3-17.9], p = 0.025). The median progression-free survival was longer in patients with HER2-positive versus HER2-negative disease, but the difference was not statistically significant (4.3 months [95% CI, 3.7-5.3] vs 3.7 months [95% CI, 3.4-4.0], p = 0.054). There was no statistically significant difference in median overall survival (OS) between the groups (9.8 months [95% CI, 8.9-12.3] vs 10.1 months [95% CI, 9.2-10.9], p = 0.564). CONCLUSIONS In patients with HER2-positive gastric or GEJ adenocarcinoma, the objective response rate to second-line treatment with ramucirumab plus paclitaxel was significantly higher compared to patients with HER2-negative disease. However, an increased response to treatment was not associated with an improvement in OS.
Collapse
Affiliation(s)
- Bum Jun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do, 14068, Republic of Korea
| | - Hee-Jung Jee
- Department of Biostatistics, Chungnam National University Sejong Hospital, Sejong, Republic of Korea
| | - Sun Young Rha
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Se Hoon Park
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong Gwang Kim
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea
| | - Woo Kyun Bae
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Hwasun, Republic of Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 Beon-gil Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji-Hye Byun
- Innovation Research Department, Health Insurance Review and Assessment Service, Wonju, Republic of Korea
| | - Dong Sook Kim
- Review and Assessment Research Department, Health Insurance Review and Assessment Service, Wonju, Republic of Korea
| | - Young Ju Suh
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hyonggin An
- Department of Biostatistics, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Dae Young Zang
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do, 14068, Republic of Korea.
| |
Collapse
|
20
|
Population pharmacokinetics and exposure-response relationship of trastuzumab and bevacizumab in early-stage breast cancer. Eur J Clin Pharmacol 2021; 77:1861-1873. [PMID: 34245336 DOI: 10.1007/s00228-021-03179-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 06/21/2021] [Indexed: 10/20/2022]
Abstract
AIMS To describe the sources of interindividual variability of bevacizumab and trastuzumab pharmacokinetics in early-stage breast cancer, and to study the relationship between exposure and both early clinical response and specific adverse events. PATIENTS AND METHODS Patients (n = 86) received 6 cycles of docetaxel + trastuzumab. Early tumour response was assessed by determination of the maximum standard uptake value (SUVmax) variation (ΔSUVmax) after 1 cycle using [18F]-fluorodeoxyglucose (FDG) PET. Early poor responders (ΔSUVmax < 70%) also received bevacizumab from cycle 3 to cycle 6. Sources of interindividual variability in pharmacokinetics of both antibodies were studied by population compartment modelling. Exposure as assessed by area under the concentration-versus-time curve (AUC) was compared between responders and non-responders and between patients experiencing specific adverse events or not. RESULTS A two-compartment model described the pharmacokinetics of both antibodies satisfactorily. Their central volume of distributions (Vc) increased with body surface area and their elimination half-lives were shorter (~14 days) than previously reported (~26-28 days). There was a time-dependent increase in trastuzumab Vc, positively correlated to baseline SUVmax. Bevacizumab elimination rate (k10) was positively correlated with ΔSUVmax measured at the end of the first cycle. Bevacizumab had no significantly influence on trastuzumab pharmacokinetics. No relationship between exposure and clinical response or occurrence of adverse events was found. CONCLUSION Tumour uptake as assessed by SUVmax influences the pharmacokinetics of bevacizumab and trastuzumab. In early-stage breast cancer, elimination half-lives of these therapeutic monoclonal antibodies may be shorter than those previously reported in more advanced disease.
Collapse
|
21
|
Roviello G, Catalano M, D’Angelo A, Palmieri VE. Second line of treatment for HER2-positive gastric cancer: an evolving issue. Rep Pract Oncol Radiother 2021; 26:316-317. [PMID: 34211782 PMCID: PMC8241291 DOI: 10.5603/rpor.a2021.0024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 01/30/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
| | - Martina Catalano
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Alberto D’Angelo
- Department of Biology and Biochemistry, University of Bath, Bath, England, United Kingdom
| | | |
Collapse
|
22
|
Ruiz-Manzano RA, Palacios-Arreola MI, Hernández-Cervantes R, Del Río-Araiza VH, Nava-Castro KE, Ostoa-Saloma P, Muñoz-Cruz S, Morales-Montor J. Potential Novel Risk Factor for Breast Cancer: Toxocara canis Infection Increases Tumor Size Due to Modulation of the Tumor Immune Microenvironment. Front Oncol 2020; 10:736. [PMID: 32547942 PMCID: PMC7272683 DOI: 10.3389/fonc.2020.00736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Worldwide, breast cancer is the most important type of cancer in women with regard to incidence and prevalence. Several risk factors interact to increase the probability of breast cancer development. Biological environmental contaminants such as infectious agents play a significant role in tumor development, and helminths have been recognized as cancer enhancers or inducers due to their ability to regulate the host immune response. Toxocara canis is a zoonotic and cosmopolite nematode with immuno-regulatory abilities. T. canis infection has been related to T helper type-2 cell (Th2 or type 2) and regulatory responses. Type 2 and regulatory immune responses may favor the development of comorbidities that are usually controlled or eliminated through a type 1 response such as cancer. The aim of this study was to determine whether T. canis infection alters mammary tumor growth through modulation of the immune response. Infected mice developed larger tumors. Tumor immune cell milieu analysis revealed that infection reduced the proportions of CD8+ lymphocytes and increased the proportions of F4/80+ macrophages and CD19+ B cells. These changes were accompanied by a type 2 local response represented by increased amounts of IL-4 and VEGF and a regulatory microenvironment associated with higher IL-10 levels. Thus, this study demonstrates that T. canis infection enhances tumor development and suggests that this is through modulation of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Rocío Alejandra Ruiz-Manzano
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Margarita Isabel Palacios-Arreola
- Departamento de Genotoxicología y Mutagénesis Ambiental, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Rosalía Hernández-Cervantes
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Víctor Hugo Del Río-Araiza
- Laboratorio de Inmunología y Biología Molecular de Parásitos, Facultad de Medicina Veterinaria y Zootecnia, Departamento de Parasitología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Karen Elizabeth Nava-Castro
- Departamento de Genotoxicología y Mutagénesis Ambiental, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Pedro Ostoa-Saloma
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Samira Muñoz-Cruz
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
23
|
Danni L, Lingyun Z, Jian W, Hongfei Y, Lu X, Peng Y, Xiujuan Q, Yunpeng L, Yuee T. Significant response to apatinib monotherapy in heavily pretreated advanced HER2-positive breast cancer: a case report and literature review. Cancer Biol Ther 2020; 21:590-596. [PMID: 32233990 DOI: 10.1080/15384047.2020.1743159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although HER2-targeted therapy has been shown to prolong the survival of patients with HER2-positive breast cancer, most patients eventually progress due to drug resistance. Novel treatment options are urgently needed to overcome resistance to HER2-targeted therapy. The VEGF/VEGFR (Vascular endothelial growth factor and its receptors) pathway is essential in tumor angiogenesis, which may be a promising target in HER2-positive breast cancer providing a rationale for the use of tyrosine kinase inhibitors (TKIs) targeting VEGFR. Here, we present a case of a heavily pretreated advanced breast cancer patient who did not respond to HER2-targeted therapy and developed resistance to multiple lines of HER2-targeted treatment. The patient was treated with apatinib at a dose of 500 mg daily, and obtained partial remission (PR) with a progression-free-stage (PFS) of 6 months. Our case indicates that apatinib might have anti-tumor activity in patients with HER2-positive breast cancer with HER2-targeted resistance. This case is of value which may provide new insights into strategies for HER2-targeted therapy resistance options in the clinic.
Collapse
Affiliation(s)
- Li Danni
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Zhang Lingyun
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Wang Jian
- Department of Pathology, The First Hospital of China Medical University , Shenyang, China
| | - Yan Hongfei
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Xu Lu
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Yang Peng
- Nanjing Geneseeq Technology Inc , Nanjing, China
| | - Qu Xiujuan
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Liu Yunpeng
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Teng Yuee
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| |
Collapse
|
24
|
Li X, Li Z, Zhu Y, Li Z, Yao L, Zhang L, Yuan L, Shang Y, Liu J, Li C. miR-524-5p inhibits angiogenesis through targeting WNK1 in colon cancer cells. Am J Physiol Gastrointest Liver Physiol 2020; 318:G827-G839. [PMID: 32174132 DOI: 10.1152/ajpgi.00369.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
There is increasing evidence that microRNA (miRNA) abnormity is involved in the occurrence and the development of various malignancies, including colon cancer. MiRNA-524-5p has been reported to possess anticancer activity in various tumors, which function is seldom investigated in colon cancer cells. The aim of this study was to explore the effect of the miRNA-524-5p/with-no-lysine kinase 1 (WNK1) system on angiogenesis in a colon cancer cell line (HT-29 and COLO205 cells) and further investigate the potential mechanisms. We found miRNA-524-5p expression was relatively high in COLO205 cells and relatively low in HT-29 cells. Elevating miRNA-524-5p expression inhibited proliferation, induced cycle arrest, diminished vascular endothelial growth factor production, and thereby suppressed angiogenesis in HT-29 cells. WNK1 silencing exerted the ability of antiangiogenesis in HT-29 cells. Besides, miRNA-524-5p deficiency-induced angiogenesis was impeded by WNK1 silence in COLO205 cells. In a murine tumor model, miRNA-524-5p agomir treatment significantly suppressed colon cancer tumorigenicity with the downregulation of WNK1 expression. In summary, our results indicated that miRNA-524-5p inhibited angiogenesis in colon cancer cells via targeting WNK1.NEW & NOTEWORTHY MiRNA-524-5p inhibited angiogenesis in colon cancer cells via targeting with-no-lysine kinase 1.
Collapse
Affiliation(s)
- Xiang Li
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Zitao Li
- Department of Orthopedic Surgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Ye Zhu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, People's Republic of China
| | - Zhu Li
- Image Equipment and Technology Laboratory, College of Medical Imaging, Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Lihong Yao
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Lamei Zhang
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Lin Yuan
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Yang Shang
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Jianting Liu
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Caijuan Li
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China.,Key Laboratory of Tumor Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| |
Collapse
|
25
|
Low-Dose Perfusion Computed Tomography for Breast Cancer to Quantify Tumor Vascularity: Correlation With Prognostic Biomarkers. Invest Radiol 2019; 54:273-281. [PMID: 30570503 DOI: 10.1097/rli.0000000000000538] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the feasibility of using low-dose perfusion computed tomography (CT) in breast cancers for quantification of tumor vascularity and to correlate perfusion indexes with prognostic biomarkers. MATERIALS AND METHODS This preliminary study was approved by our institutional review board. Signed informed consent was obtained from all 70 enrolled patients with invasive breast cancers. Low-dose perfusion CT was performed with the patient in the prone position using a spectral CT device set at 80 kVp and 30 mAs (1.30-1.40 mSv). Images were analyzed using commercial software applying the maximum slope algorithm. On CT perfusion maps, perfusion (mL/min per 100 mL), blood volume (mL/100 g), time-to-peak enhancement (second), and peak enhancement intensity (HU) were measured in the tumor, normal breast glandular tissues, and fat. Tumor grade, estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2), and Ki67 level were evaluated using histopathology. Statistically, CT perfusion indexes of the tumor and normal glandular tissues or fat were compared using the Wilcoxon signed-rank test, and CT indexes were correlated with histological characteristics using the Mann-Whitney U or Kruskal-Wallis tests. We also correlated CT indexes with magnetic resonance imaging enhancement characteristics. RESULTS In breast cancers, perfusion, blood volume, and peak enhancement intensity values were significantly higher, and time to peak was shorter than in normal glandular tissues and fat (P < 0.001). Perfusion increased significantly in high-grade, ER-, or HER2+ cancers (P < 0.05). Time to peak decreased in ER-, HER2+, and high-grade cancers or in those with high Ki67 levels (P < 0.05). Peak enhancement intensity significantly increased in high-grade cancers (P < 0.05). HER2 overexpressing cancers showed significantly higher perfusion and shorter time to peak than luminal-type cancers (P < 0.05). Perfusion increased and time to peak decreased significantly in cancers with washout enhancement patterns on magnetic resonance imaging. CONCLUSIONS Low-dose perfusion CT in the prone position is feasible to quantify tumor vascularity in breast cancers, and CT perfusion indexes are significantly correlated with prognostic biomarkers and molecular subtypes of breast cancer.
Collapse
|
26
|
Guarischi-Sousa R, Monteiro JS, Alecrim LC, Michaloski JS, Cardeal LB, Ferreira EN, Carraro DM, Nunes DN, Dias-Neto E, Reimand J, Boutros PC, Setubal JC, Giordano RJ. A transcriptome-based signature of pathological angiogenesis predicts breast cancer patient survival. PLoS Genet 2019; 15:e1008482. [PMID: 31846472 PMCID: PMC6917213 DOI: 10.1371/journal.pgen.1008482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022] Open
Abstract
The specific genes and molecules that drive physiological angiogenesis differ from those involved in pathological angiogenesis, suggesting distinct mechanisms for these seemingly related processes. Unveiling genes and pathways preferentially associated with pathologic angiogenesis is key to understanding its mechanisms, thereby facilitating development of novel approaches to managing angiogenesis-dependent diseases. To better understand these different processes, we elucidated the transcriptome of the mouse retina in the well-accepted oxygen-induced retinopathy (OIR) model of pathological angiogenesis. We identified 153 genes changed between normal and OIR retinas, which represent a molecular signature relevant to other angiogenesis-dependent processes such as cancer. These genes robustly predict the survival of breast cancer patients, which was validated in an independent 1,000-patient test cohort (40% difference in 15-year survival; p = 2.56 x 10-21). These results suggest that the OIR model reveals key genes involved in pathological angiogenesis, and these may find important applications in stratifying tumors for treatment intensification or for angiogenesis-targeted therapies.
Collapse
Affiliation(s)
- Rodrigo Guarischi-Sousa
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jhonatas S. Monteiro
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Lilian C. Alecrim
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Jussara S. Michaloski
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Laura B. Cardeal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Elisa N. Ferreira
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Dirce M. Carraro
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Diana N. Nunes
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Emmanuel Dias-Neto
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
- Laboratory of Neurosciences (LIM27), Institute & Department of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- * E-mail: (PCB); (JCS); (RJG)
| | - João C. Setubal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| | - Ricardo J. Giordano
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| |
Collapse
|
27
|
Bonelli P, Borrelli A, Tuccillo FM, Silvestro L, Palaia R, Buonaguro FM. Precision medicine in gastric cancer. World J Gastrointest Oncol 2019; 11:804-829. [PMID: 31662821 PMCID: PMC6815928 DOI: 10.4251/wjgo.v11.i10.804] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/11/2019] [Accepted: 09/05/2019] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a complex disease linked to a series of environmental factors and unhealthy lifestyle habits, and especially to genetic alterations. GC represents the second leading cause of cancer-related deaths worldwide. Its onset is subtle, and the majority of patients are diagnosed once the cancer is already advanced. In recent years, there have been innovations in the management of advanced GC including the introduction of new classifications based on its molecular characteristics. Thanks to new technologies such as next-generation sequencing and microarray, the Cancer Genome Atlas and Asian Cancer Research Group classifications have also paved the way for precision medicine in GC, making it possible to integrate diagnostic and therapeutic methods. Among the objectives of the subdivision of GC into subtypes is to select patients in whom molecular targeted drugs can achieve the best results; many lines of research have been initiated to this end. After phase III clinical trials, trastuzumab, anti-Erb-B2 receptor tyrosine kinase 2 (commonly known as ERBB2) and ramucirumab, anti-vascular endothelial growth factor receptor 2 (commonly known as VEGFR2) monoclonal antibodies, were approved and introduced into first- and second-line therapies for patients with advanced/metastatic GC. However, the heterogeneity of this neoplasia makes the practical application of such approaches difficult. Unfortunately, scientific progress has not been matched by progress in clinical practice in terms of significant improvements in prognosis. Survival continues to be low in contrast to the reduction in deaths from many common cancers such as colorectal, lung, breast, and prostate cancers. Although several target molecules have been identified on which targeted drugs can act and novel products have been introduced into experimental therapeutic protocols, the overall approach to treating advanced stage GC has not substantially changed. Currently, surgical resection with adjuvant or neoadjuvant radiotherapy and chemotherapy are the most effective treatments for this disease. Future research should not underestimate the heterogeneity of GC when developing diagnostic and therapeutic strategies aimed toward improving patient survival.
Collapse
Affiliation(s)
- Patrizia Bonelli
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Antonella Borrelli
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Franca Maria Tuccillo
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Lucrezia Silvestro
- Abdominal Medical Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Raffaele Palaia
- Gastro-pancreatic Surgery Division, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| |
Collapse
|
28
|
De Vita F, Borg C, Farina G, Geva R, Carton I, Cuku H, Wei R, Muro K. Ramucirumab and paclitaxel in patients with gastric cancer and prior trastuzumab: subgroup analysis from RAINBOW study. Future Oncol 2019; 15:2723-2731. [PMID: 31234645 DOI: 10.2217/fon-2019-0243] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: This subgroup analysis of the RAINBOW study evaluated the efficacy and safety of ramucirumab in patients with gastric cancer/gastroesophageal junction adenocarcinoma who received prior trastuzumab therapy. Patients & methods: Of adult patients enrolled in the RAINBOW study, 39 had received prior trastuzumab therapy. Of these, 20 patients were treated with ramucirumab plus paclitaxel and 19 patients with placebo plus paclitaxel within the RAINBOW trial. Results: Overall survival was longer with ramucirumab plus paclitaxel (11.4 months; 95% CI: 7.0-17.9) versus placebo plus paclitaxel (7.0 months; 95% CI: 3.4-14.6), hazard ratio: 0.68 (0.33-1.41); p = 0.30. Longer progression-free survival, higher objective response were observed in ramucirumab combination group. Conclusion: Ramucirumab plus paclitaxel demonstrated efficacy benefits with manageable safety profile in a subgroup of patients pretreated with trastuzumab. Clinical trial registration number: NCT01170663.
Collapse
Affiliation(s)
- Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, 80138, Italy
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon, 25000, France
| | - Gabriella Farina
- Department of Medical Oncology, Fatebenefratelli Sacco Hospital, Milan, 20121, Italy
| | - Ravit Geva
- Oncology Division, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Iris Carton
- Eli Lilly & Company, Brussels, 1000, Belgium
| | - Hera Cuku
- Eli Lilly & Company, Florence, 50019, Italy
| | - Ran Wei
- Eli Lilly & Company, Indianapolis, IN 46285, USA
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, 464-8681, Japan
| |
Collapse
|
29
|
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
|
30
|
Méndez-García LA, Nava-Castro KE, Ochoa-Mercado TDL, Palacios-Arreola MI, Ruiz-Manzano RA, Segovia-Mendoza M, Solleiro-Villavicencio H, Cázarez-Martínez C, Morales-Montor J. Breast Cancer Metastasis: Are Cytokines Important Players During Its Development and Progression? J Interferon Cytokine Res 2018; 39:39-55. [PMID: 30321090 DOI: 10.1089/jir.2018.0024] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In breast cancer, an uncontrolled cell proliferation leads to tumor formation and development of a multifactorial disease. Metastasis is a complex process that involves tumor spread to distant parts of the body from its original site. Metastatic dissemination represents the main physiopathology of cancer. Inter- and intracellular communication in all systems in vertebrates is mediated by cytokines, which are highly inducible, secretory proteins, produced not only by immune system cells, but also by endocrine and nervous system cells. It has become clear in recent years that cytokines, as well as their receptors are produced in the organisms under physiological and pathological conditions; recently, they have been closely related to breast cancer metastasis. The exact initiation process of breast cancer metastasis is unknown, although several hypotheses have emerged. In this study, we thoroughly reviewed the role of several cytokines in breast cancer metastasis. Data reviewed suggest that cytokines and growth factors are key players in the breast cancer metastasis induction. This knowledge must be considered with the aim to development of new therapeutic approaches to counter breast cancer metastasis.
Collapse
Affiliation(s)
| | - Karen Elizabeth Nava-Castro
- 2 Laboratorio de Genotoxicología y Medicina Ambientales, Departamento de Ciencias Ambientales, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, México DF, México
| | - Tania de Lourdes Ochoa-Mercado
- 3 Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Margarita Isabel Palacios-Arreola
- 2 Laboratorio de Genotoxicología y Medicina Ambientales, Departamento de Ciencias Ambientales, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, México DF, México
| | - Rocío Alejandra Ruiz-Manzano
- 3 Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mariana Segovia-Mendoza
- 3 Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Helena Solleiro-Villavicencio
- 4 Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, México DF, Mexico
| | - Cinthia Cázarez-Martínez
- 2 Laboratorio de Genotoxicología y Medicina Ambientales, Departamento de Ciencias Ambientales, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, México DF, México
| | - Jorge Morales-Montor
- 3 Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
31
|
Ciesielski M, Szajewski M, Pęksa R, Lewandowska MA, Zieliński J, Walczak J, Szefel J, Kruszewski WJ. The relationship between HER2 overexpression and angiogenesis in gastric cancer. Medicine (Baltimore) 2018; 97:e12854. [PMID: 30334990 PMCID: PMC6211927 DOI: 10.1097/md.0000000000012854] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In gastric cancer, HER2 protein overexpression is considered to be conducive to the higher proliferation activity of the tumor cells. Tumor formation is associated with angiogenesis in order to secure an abundant supply of oxygen and glucose to cancer cells. The aim of the study was to assess if HER2 overexpression is related to higher microvessel density (MVD) in the tumor stroma.The archival samples of primary tumor from 144 consecutive patients that underwent gastric resection for cancer between August 1, 2006 and December 31, 2013 in the Department of Oncological Surgery of Medical University of Gdańsk were analyzed. CD34 was used as a marker of MVD in the tumor stroma. Both CD34 and HER2 protein expressions were tested by immunohistochemistry.The assays were unsuccessful to estimate HER2 in 10 cases and CD34 in 14 cases due to technical reasons. The results were obtained for 128 patients. HER2 0 and HER2 1+ were considered negative, while HER2+ and HER2 3+ were recognized as positive. Mean MVD (mean number of vessels in the visual field) was 32.4 (median 29.5). Microvessel density was insignificantly higher in HER2 positive tumors. The slight difference was also seen between IHC 2+ and 3+ groups. The differences did not reach the level of statistical significance.Statistical analysis performed in our study did not reveal the significant relationship between HER2 overexpression on the tumor cells and MVD in the tumor stroma.
Collapse
Affiliation(s)
- Maciej Ciesielski
- Department of Oncological Surgery, Gdynia Oncology Centre, Gdynia
- Division of Propedeutics of Oncology, Medical University Of Gdańsk
| | - Mariusz Szajewski
- Department of Oncological Surgery, Gdynia Oncology Centre, Gdynia
- Division of Propedeutics of Oncology, Medical University Of Gdańsk
| | - Rafał Pęksa
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk
| | | | - Jacek Zieliński
- Department of Oncological Surgery, Medical University of Gdańsk, Gdańsk, Poland
| | - Jakub Walczak
- Department of Oncological Surgery, Gdynia Oncology Centre, Gdynia
| | - Jarosław Szefel
- Department of Oncological Surgery, Gdynia Oncology Centre, Gdynia
- Division of Propedeutics of Oncology, Medical University Of Gdańsk
| | - Wiesław Janusz Kruszewski
- Department of Oncological Surgery, Gdynia Oncology Centre, Gdynia
- Division of Propedeutics of Oncology, Medical University Of Gdańsk
| |
Collapse
|
32
|
Angiogenesis and cancer stem cells: New perspectives on therapy of ovarian cancer. Eur J Med Chem 2017; 142:87-94. [PMID: 28651817 DOI: 10.1016/j.ejmech.2017.06.030] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/18/2022]
Abstract
Failure in ovarian cancer therapy, following cytoreduction and chemotherapy, is related to the presence of cancer stem cells - a small subpopulation of cells resistant to chemotherapy and irradiation - in the tumour which may cause cancer relapse and manifestation of metastases. Therapies targeted at Cancer Stem Cells (CSCs), such as those employing metformin (a drug used in the treatment of diabetes type II) and salinomycin, an antibiotic isolated from Streptococcus albus bacteria, seem promising. Anti-angiogenic therapy with bevacizumab was found to be effective in all phases of ovarian cancer treatment. The presence of CSCs has been associated with angiogenesis. Several CSC biomarkers correlate with the markers of angiogenesis and some signalling pathways, e.g. Notch, and are used by both CSCs and by pro-angiogenic factors.
Collapse
|
33
|
Nasir A, Holzer TR, Chen M, Man MZ, Schade AE. Differential expression of VEGFR2 protein in HER2 positive primary human breast cancer: potential relevance to anti-angiogenic therapies. Cancer Cell Int 2017; 17:56. [PMID: 28533703 PMCID: PMC5438568 DOI: 10.1186/s12935-017-0427-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/12/2017] [Indexed: 12/28/2022] Open
Abstract
Background Clinically relevant predictive biomarkers to tailor anti-angiogenic therapies to breast cancer (BRC) patient subpopulations are an unmet need. Methods We analyzed tumor vascular density and VEGFR2 protein expression in various subsets of primary human BRCs (186 females; Mean age: 59 years; range 33–88 years), using a tissue microarray. Discrete VEGFR2+ and CD34+ tumor vessels were manually scored in invasive ductal, lobular, mixed ductal-lobular and colloid (N = 139, 22, 18, 7) BRC cores. Results The observed CD34+ and VEGFR2+ tumor vascular counts in individual cases were heterogeneous. Mean CD34+ and VEGFR2+ tumor vessel counts were 11 and 3.4 per tumor TMA core respectively. Eighty-nine of 186 (48%) cases had >10 CD34+ tumor vessels, while 97/186 (52%) had fewer CD34+ vessels in each TMA core. Of 169 analyzable cores in the VEGFR2 stained TMA, 90 (53%) showed 1–5 VEGFR2+ tumor vessels/TMA core, while 42/169 (25%) cores had no detectable VEGFR2+ tumor vessels. Thirteen of 169 (8%) cases also showed tumor cell (cytoplasmic/membrane) expression of VEGFR2. Triple-negative breast cancers (TNBCs) appeared to be less vascular (Mean VD = 9.8, range 0–34) than other breast cancer subtypes. Overall, VEGFR2+ tumor vessel counts were significantly higher in HER2+ as compared to HR+ (p = 0.04) and TNBC (p = 0.02) tissues. Compared to HER2− cases, HER2+ breast cancers had higher VEGFR2+ tumor vessel counts (p = 0.007). Conclusion Characterization of pathologic angiogenesis in HER2+ breast cancer provides scientific rationale for future investigation of clinical activity of agents targeting the VEGF/VEGFR2 axis in this clinically aggressive breast cancer subtype.
Collapse
Affiliation(s)
- Aejaz Nasir
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA.,Eli Lilly and Company, Lilly Corporate Center, DC0424, Indianapolis, IN 46285 USA
| | - Timothy R Holzer
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA
| | - Mia Chen
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA
| | - Michael Z Man
- Oncology Statistics, Eli Lilly and Company, Indianapolis, IN USA
| | - Andrew E Schade
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA
| |
Collapse
|
34
|
Dewangan J, Srivastava S, Rath SK. Salinomycin: A new paradigm in cancer therapy. Tumour Biol 2017; 39:1010428317695035. [PMID: 28349817 DOI: 10.1177/1010428317695035] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The primary hurdle in the treatment of cancer is acquisition of resistance by the tumor cells toward multiple drugs and selectively targeting the cancer stem cells. This problem was overcome by the chemotherapeutic property of recently discovered drug salinomycin. Exact mechanism of action of salinomycin is not yet known, but there are multiple pathways by which salinomycin inhibits tumor growth. Salinomycin decreases the expression of adenosine triphosphate-binding cassette transporter in multidrug resistance cells and interferes with Akt signaling pathway, Wnt/β-catenin, Hedgehog, and Notch pathways of cancer progression. Salinomycin selectively targets cancer stem cells. The potential of salinomycin to eliminate both cancer stem cells and therapy-resistant cancer cells may characterize the compound as a novel and an efficient chemotherapeutic drug.
Collapse
Affiliation(s)
- Jayant Dewangan
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sonal Srivastava
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Srikanta Kumar Rath
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
35
|
Drooger JC, van Tinteren H, de Groot SM, Ten Tije AJ, de Graaf H, Portielje JEA, Jager A, Honkoop A, Linn SC, Kroep JR, Erdkamp FLG, Hamberg P, Imholz ALT, van Rossum-Schornagel QC, Heijns JB, van Leeuwen-Stok AE, Sleijfer S. A randomized phase 2 study exploring the role of bevacizumab and a chemotherapy-free approach in HER2-positive metastatic breast cancer: The HAT study (BOOG 2008-2003), a Dutch Breast Cancer Research Group trial. Cancer 2016; 122:2961-70. [PMID: 27315546 DOI: 10.1002/cncr.30141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/11/2016] [Accepted: 04/25/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND To explore the role of bevacizumab and a chemotherapy-free approach, the authors evaluated the combination of bevacizumab, trastuzumab, and paclitaxel (HAT) and the regimen of trastuzumab and bevacizumab (HA) with the addition of paclitaxel after progression (HA-HAT) as first-line treatment for patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer. METHODS In a noncomparative phase 2 trial, patients were randomized between HAT and HA-HAT. The primary endpoint was the progression-free rate at 1 year (1-year PFR). In the HA-HAT group, progression-free survival (PFS) was separately established for HA (PFS1) and HAT (PFS2). RESULTS Eighty-four patients received HAT (n = 39) or HA-HAT (n = 45). The 1-year PFR was 74.4% (95% confidence interval [CI], 61.8%-89.4%) and 62.2% (95% CI, 49.6%-89.4%) in the HAT and HA-HAT arms, respectively. The median PFS was 19.8 months (95% CI, 14.9-25.6 months) in the HAT arm and 19.6 months (95% CI, 12.0-32.0 months) in the HA-HAT arm. In the HA-HAT arm, the median PFS1 was 10.4 months (95% CI, 6.2-15.0 months), and the median PFS2 was 8.2 months (95% CI, 7.0-12.6 months). The number and severity of adverse events were comparable between the arms. CONCLUSIONS Both HAT and HA-HAT have promising activity in patients with HER2-positive metastatic breast cancer. In particular, starting with only targeted agents and delaying chemotherapy is worth further exploration. Cancer 2016;122:2961-2970. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Jan C Drooger
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute and Cancer Genomics Netherlands, Rotterdam, the Netherlands. .,Department of Medical Oncology, Ikazia Hospital, Rotterdam, the Netherlands.
| | - Harm van Tinteren
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Steffen M de Groot
- Comprehensive Cancer Center of the Netherlands, Amsterdam, the Netherlands
| | - Albert J Ten Tije
- Department of Medical Oncology, Amphia Hospital, Breda, the Netherlands
| | - Hiltje de Graaf
- Department of Medical Oncology, Leeuwarden Medical Center, Leeuwarden, the Netherlands
| | | | - Agnes Jager
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute and Cancer Genomics Netherlands, Rotterdam, the Netherlands
| | - Aafke Honkoop
- Department of Medical Oncology, Isala Clinic, Zwolle, the Netherlands
| | - Sabine C Linn
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frans L G Erdkamp
- Department of Medical Oncology, Orbis Medical Center, Sittard, the Netherlands
| | - Paul Hamberg
- Department of Medical Oncology, Sint Franciscus Gasthuis, Rotterdam, the Netherlands
| | - Alex L T Imholz
- Department of Medical Oncology, Deventer Hospital, Deventer, the Netherlands
| | | | - Joan B Heijns
- Department of Medical Oncology, Amphia Hospital, Breda, the Netherlands
| | | | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute and Cancer Genomics Netherlands, Rotterdam, the Netherlands
| |
Collapse
|
36
|
Rao A, Manyam G, Rao G, Jain R. Integrative Analysis of mRNA, microRNA, and Protein Correlates of Relative Cerebral Blood Volume Values in GBM Reveals the Role for Modulators of Angiogenesis and Tumor Proliferation. Cancer Inform 2016; 15:29-33. [PMID: 27053917 PMCID: PMC4814129 DOI: 10.4137/cin.s33014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/29/2016] [Accepted: 12/07/2015] [Indexed: 12/12/2022] Open
Abstract
Dynamic susceptibility contrast-enhanced magnetic resonance imaging is routinely used to provide hemodynamic assessment of brain tumors as a diagnostic as well as a prognostic tool. Recently, it was shown that the relative cerebral blood volume (rCBV), obtained from the contrast-enhancing as well as -nonenhancing portion of glioblastoma (GBM), is strongly associated with overall survival. In this study, we aim to characterize the genomic correlates (microRNA, messenger RNA, and protein) of this vascular parameter. This study aims to provide a comprehensive radiogenomic and radioproteomic characterization of the hemodynamic phenotype of GBM using publicly available imaging and genomic data from the Cancer Genome Atlas GBM cohort. Based on this analysis, we identified pathways associated with angiogenesis and tumor proliferation underlying this hemodynamic parameter in GBM.
Collapse
Affiliation(s)
- Arvind Rao
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju Manyam
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganesh Rao
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajan Jain
- Department of Radiology, NY University School of Medicine, New York, NY, USA
| |
Collapse
|
37
|
Nalwoga H, Ahmed L, Arnes JB, Wabinga H, Akslen LA. Strong Expression of Hypoxia-Inducible Factor-1α (HIF-1α) Is Associated with Axl Expression and Features of Aggressive Tumors in African Breast Cancer. PLoS One 2016; 11:e0146823. [PMID: 26760782 PMCID: PMC4711940 DOI: 10.1371/journal.pone.0146823] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/22/2015] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Inhibition of hypoxia-inducible factor (HIF) and Axl receptor tyrosine kinase is being evaluated for targeted therapy in solid tumors. Both HIF-1α and Axl influence tumor growth and metastatic potential, and they have been linked to treatment failure in many cancers. However, there is a lack of reports on HIF-1α expression in African breast cancer, which has a poor prognosis, and novel treatment targets must therefore be established. Here, we aimed to evaluate HIF-1α in relation to Axl expression, angiogenesis markers, and other tumor characteristics in a series of African breast cancer. METHODS Using immunohistochemistry, we examined 261 invasive breast cancers on tissue microarrays for HIF-1α and Axl as well as several other markers, and a subset of 185 cases had information on VEGF (vascular endothelial growth factor) expression, microvessel density (MVD), proliferating microvessel density (pMVD) and vascular proliferation index (VPI) for important comparisons. RESULTS Strong HIF-1α expression was associated with increased Axl (p = 0.007), VEGF (p<0.0005), and p53 (p = 0.032) expression, as well as high tumor cell proliferation by Ki-67 (p = 0.006), and high tumor grade (p = 0.003). Tumors with strong HIF-1α expression had significantly higher MVD (p = 0.019) and higher pMVD (p = 0.027) than tumors with weak expression. CONCLUSIONS High HIF-1α expression is significantly associated with Axl and VEGF expression, and with markers of poor prognosis in this series of breast cancer, suggesting HIF-1α and Axl as potential therapeutic targets in African breast cancer.
Collapse
Affiliation(s)
- Hawa Nalwoga
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Haukeland University Hospital, Bergen, Norway
- Department of Pathology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Lavina Ahmed
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Haukeland University Hospital, Bergen, Norway
- BerGenBio AS, Bergen, Norway
| | - Jarle B. Arnes
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Henry Wabinga
- Department of Pathology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Haukeland University Hospital, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| |
Collapse
|
38
|
Shi JH, Cui NP, Wang S, Zhao MZ, Wang B, Wang YN, Chen BP. Overexpression of YB1 C-terminal domain inhibits proliferation, angiogenesis and tumorigenicity in a SK-BR-3 breast cancer xenograft mouse model. FEBS Open Bio 2016; 6:33-42. [PMID: 27047740 PMCID: PMC4794790 DOI: 10.1002/2211-5463.12004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/27/2015] [Accepted: 11/29/2015] [Indexed: 12/12/2022] Open
Abstract
Y-box-binding protein 1 (YB1) is a multifunctional transcription factor with vital roles in proliferation, differentiation and apoptosis. In this study, we have examined the role of its C-terminal domain (YB1 CTD) in proliferation, angiogenesis and tumorigenicity in breast cancer. Breast cancer cell line SK-BR-3 was infected with GFP-tagged YB1 CTD adenovirus expression vector. An 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) proliferation assay showed that YB1 CTD decreased SK-BR-3 cell proliferation, and down-regulated cyclin B1 and up-regulated p21 levels in SK-BR-3 cells. YB1 CTD overexpression changed the cytoskeletal organization and slightly inhibited the migration of SK-BR-3 cells. YB1 CTD also inhibited secreted VEGF expression in SK-BR-3 cells, which decreased SK-BR-3-induced EA.hy926 endothelial cell angiogenesis in vitro. YB1 CTD overexpression attenuated the ability of SK-BR-3 cells to form tumours in nude mice, and decreased in vivo VEGF levels and angiogenesis in the xenografts in SK-BR-3 tumour-bearing mice. Taken together, our findings demonstrate the vital role of YB1 CTD overexpression in inhibiting proliferation, angiogenesis and tumorigenicity of breast cancer cell line SK-BR-3.
Collapse
Affiliation(s)
- Jian-Hong Shi
- Central Laboratory Hebei Laboratory of Mechanism and Procedure of Cancer Radiotherapy and Chemotherapy Affiliated Hospital of Hebei University Baoding China
| | - Nai-Peng Cui
- Department of Oncology Affiliated Hospital of Hebei University Baoding China
| | - Shuo Wang
- Central Laboratory Hebei Laboratory of Mechanism and Procedure of Cancer Radiotherapy and Chemotherapy Affiliated Hospital of Hebei University Baoding China
| | - Ming-Zhi Zhao
- Department of Oncology Affiliated Hospital of Hebei University Baoding China
| | - Bing Wang
- Department of Oncology Affiliated Hospital of Hebei University Baoding China
| | - Ya-Nan Wang
- Department of Pathology Affiliated Hospital of Hebei University Baoding China
| | - Bao-Ping Chen
- Department of Oncology Affiliated Hospital of Hebei University Baoding China
| |
Collapse
|
39
|
Analysis of gene expression of secreted factors associated with breast cancer metastases in breast cancer subtypes. Sci Rep 2015; 5:12133. [PMID: 26173622 PMCID: PMC4648401 DOI: 10.1038/srep12133] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 06/02/2015] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is a heterogeneous disease, having multiple subtypes with different malignant phenotypes. The triple-negative breast cancer, or basal breast cancer, is highly aggressive, metastatic, and difficult to treat. Previously, we identified that key molecules (IL6, CSF2, CCL5, VEGFA, and VEGFC) secreted by tumor cells and stromal cells in basal breast cancer can promote metastasis. It remains to assess whether these molecules function similarly in other subtypes of breast cancer. Here, we characterize the relative gene expression of the five secreted molecules and their associated receptors (GP130, GMRA, GMRB, CCR5, VEGFR2, NRP1, VEGFR3, NRP2) in the basal, HER2 (human epidermal growth factor receptor 2) positive, luminal A, and luminal B subtypes using high throughput data from tumor samples in The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC). IL6 and CCL5 gene expression are basal breast cancer specific, whereas high gene expression of GP130 was observed in luminal A/B. VEGFA/C and CSF2 mRNA are overexpressed in HER2 positive breast cancer, with VEGFA and CSF2 also overexpressed in basal breast cancer. Further study of the specific protein function of these factors within their associated cancer subtypes may yield personalized biomarkers and treatment modalities.
Collapse
|
40
|
Zhang L, Li J, Xiao Y, Cui H, Du G, Wang Y, Li Z, Wu T, Li X, Tian J. Identifying ultrasound and clinical features of breast cancer molecular subtypes by ensemble decision. Sci Rep 2015; 5:11085. [PMID: 26046791 PMCID: PMC4457139 DOI: 10.1038/srep11085] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/14/2015] [Indexed: 11/30/2022] Open
Abstract
Breast cancer is molecularly heterogeneous and categorized into four molecular subtypes: Luminal-A, Luminal-B, HER2-amplified and Triple-negative. In this study, we aimed to apply an ensemble decision approach to identify the ultrasound and clinical features related to the molecular subtypes. We collected ultrasound and clinical features from 1,000 breast cancer patients and performed immunohistochemistry on these samples. We used the ensemble decision approach to select unique features and to construct decision models. The decision model for Luminal-A subtype was constructed based on the presence of an echogenic halo and post-acoustic shadowing or indifference. The decision model for Luminal-B subtype was constructed based on the absence of an echogenic halo and vascularity. The decision model for HER2-amplified subtype was constructed based on the presence of post-acoustic enhancement, calcification, vascularity and advanced age. The model for Triple-negative subtype followed two rules. One was based on irregular shape, lobulate margin contour, the absence of calcification and hypovascularity, whereas the other was based on oval shape, hypovascularity and micro-lobulate margin contour. The accuracies of the models were 83.8%, 77.4%, 87.9% and 92.7%, respectively. We identified specific features of each molecular subtype and expanded the scope of ultrasound for making diagnoses using these decision models.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jing Li
- Department of Ultrasonic medicine, The 1st Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hao Cui
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guoqing Du
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying Wang
- Department of general surgery, The Second Hospital of Hebei Medical Universtiy, Shijiazhuang, Hebei, China
| | - Ziyao Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tong Wu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
41
|
Zheng L, Li X, Gu Y, Ma Y, Xi T. Pseudogene CYP4Z2P 3'UTR promotes angiogenesis in breast cancer. Biochem Biophys Res Commun 2014; 453:545-551. [PMID: 25281903 DOI: 10.1016/j.bbrc.2014.09.112] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 09/26/2014] [Indexed: 01/22/2023]
Abstract
Pseudogenes have long been marked as "false" genes, which are similar with real genes but have no apparent function. The 3'UTR is well-known to regulate gene expression post-transcriptionally. Our recent evidence, however, indicates novel functional roles of pseudogene CYP4Z2P 3'UTR (Z2P-UTR). We found that ectopic expression of Z2P-UTR in breast cancer cells significantly increased the expression of VEGF-A without affecting cell proliferation in vitro. Meanwhile, conditioned medium (CM) from Z2P-UTR overexpression cells enhanced proliferation, migration and tube formation of HUVEC, and promoted angiogenesis in ex vivo models. Also, CM increased the expression of VEGFR2 in HUVEC. Our data suggest that Z2P-UTR can promote breast cancer angiogenesis partly via paracrine pathway of VEGF-A/VEGFR2.
Collapse
Affiliation(s)
- Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiaoman Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yi Gu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yihua Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
42
|
From sprouting angiogenesis to erythrocytes generation by cancer stem cells: evolving concepts in tumor microcirculation. BIOMED RESEARCH INTERNATIONAL 2014; 2014:986768. [PMID: 25162040 PMCID: PMC4138761 DOI: 10.1155/2014/986768] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 01/26/2023]
Abstract
Angiogenesis is essential for tumor growth and metastasis. Over the last decades, a substantial progress has been achieved in defining different patterns of tumor microcirculation. Sprouting angiogenesis, the oldest model of microcirculation, is the de novo vessel formation from preexisting blood vessels. Vessel splitting and hijacking, also known, respectively, as intussusception and cooption, are alternative models that account for tumor resistance to antiangiogenic therapy. In addition to remodeling the microenvironment, the tumor cell can undergo intrinsic changes and survive hypoxic conditions by acquiring stem cell properties. In line with the concept of pluripotency, tumor cells can form vascular mimicry structures creating their own microcirculation despite a latent vessel growth. The recent identification of the polyploid giant cancer cells and tumor-derived erythrocytes is the most innovative survival mechanism in hypoxia and provides a potential target for more effective therapies.
Collapse
|
43
|
Kümler I, Christiansen OG, Nielsen DL. A systematic review of bevacizumab efficacy in breast cancer. Cancer Treat Rev 2014; 40:960-73. [PMID: 24909311 DOI: 10.1016/j.ctrv.2014.05.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 12/21/2022]
Abstract
UNLABELLED Angiogenesis is a key component of cancer growth, invasion and metastasis. Therefore, inhibition of angiogenesis is an attractive strategy for the treatment of cancer. We systematically describe phase II and III clinical trials of bevacizumab for the treatment of breast cancer. METHODS A computer-based literature search was carried out using PUBMED and conference databases. Original phase II and III studies reporting ≥15 patients who received bevacizumab were included. RESULTS 41 phase II trials were identified in the metastatic setting. Most trials found bevacizumab treatment feasible. Response rates (RR) varied from 0% to 76.5%, time to progression (TTP)/progression free survival (PFS) from 2.4 to 25.3 months and overall survival from 11.5 to more than 38 months. 14 phase III trials including more than 4400 patients with MBC unanimously showed increased RR and PFS, however, no trials demonstrated an OS benefit. In the neoadjuvant setting 23 phase II and III trials were identified. All studies found increased pCR/tpCR but no benefit in terms of OS could be demonstrated. The only study conducted in the adjuvant setting failed to show any survival benefit of bevacizumab. CONCLUSION Despite increased response rates in both the metastatic and neoadjuvant setting, bevacizumab has failed to show any OS benefit. Future trials should include identification of robust predictive biomarkers in order to improve our understanding of molecular biomarkers and mechanisms.
Collapse
Affiliation(s)
- Iben Kümler
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | - Ole Grummedal Christiansen
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | - Dorte Lisbet Nielsen
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| |
Collapse
|
44
|
Le Saux O, You B, Freyer G. Antiangiogenic therapy in patients with HER2-positive metastatic breast cancer: a case series. Clin Breast Cancer 2014; 14:e89-94. [PMID: 24424314 DOI: 10.1016/j.clbc.2013.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 11/11/2013] [Accepted: 11/17/2013] [Indexed: 11/17/2022]
Affiliation(s)
- Olivia Le Saux
- Department of Medical Oncology, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France.
| | - Benoît You
- Department of Medical Oncology, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Gilles Freyer
- Department of Medical Oncology, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| |
Collapse
|
45
|
Cao Z, Shang B, Zhang G, Miele L, Sarkar FH, Wang Z, Zhou Q. Tumor cell-mediated neovascularization and lymphangiogenesis contrive tumor progression and cancer metastasis. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1836:273-286. [PMID: 23933263 DOI: 10.1016/j.bbcan.2013.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/22/2013] [Accepted: 08/01/2013] [Indexed: 12/12/2022]
Abstract
Robust neovascularization and lymphangiogenesis have been found in a variety of aggressive and metastatic tumors. Endothelial sprouting angiogenesis is generally considered to be the major mechanism by which new vasculature forms in tumors. However, increasing evidence shows that tumor vasculature is not solely composed of endothelial cells (ECs). Some tumor cells acquire processes similar to embryonic vasculogenesis and produce new vasculature through vasculogenic mimicry, trans-differentiation of tumor cells into tumor ECs, and tumor cell-EC vascular co-option. In addition, tumor cells secrete various vasculogenic factors that induce sprouting angiogenesis and lymphangiogenesis. Vasculogenic tumor cells actively participate in the formation of vascular cancer stem cell niche and a premetastatic niche. Therefore, tumor cell-mediated neovascularization and lymphangiogenesis are closely associated with tumor progression, cancer metastasis, and poor prognosis. Vasculogenic tumor cells have emerged as key players in tumor neovascularization and lymphangiogenesis and play pivotal roles in tumor progression and cancer metastasis. However, the mechanisms underlying tumor cell-mediated vascularity as they relate to tumor progression and cancer metastasis remain unclear. Increasing data have shown that various intrinsic and extrinsic factors activate oncogenes and vasculogenic genes, enhance vasculogenic signaling pathways, and trigger tumor neovascularization and lymphangiogenesis. Collectively, tumor cells are the instigators of neovascularization. Therefore, targeting vasculogenic tumor cells, genes, and signaling pathways will open new avenues for anti-tumor vasculogenic and metastatic drug discovery. Dual targeting of endothelial sprouting angiogenesis and tumor cell-mediated neovascularization and lymphangiogenesis may overcome current clinical problems with anti-angiogenic therapy, resulting in significantly improved anti-angiogenesis and anti-cancer therapies.
Collapse
Affiliation(s)
- Zhifei Cao
- Cyrus Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu 215006, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Vitamin k2, a naturally occurring menaquinone, exerts therapeutic effects on both hormone-dependent and hormone-independent prostate cancer cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:287358. [PMID: 24062781 PMCID: PMC3767046 DOI: 10.1155/2013/287358] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 05/30/2013] [Indexed: 12/31/2022]
Abstract
In recent years, several studies have shown that vitamin k2 (VK2) has anticancer activity in a variety of cancer cells. The antitumor effects of VK2 in prostate cancer are currently not known. In the present study, we sought to characterize the anticancer potential of VK2 in both androgen-dependent and -independent prostate cancer cells. Our investigations show that VK2 is able to suppress viability of androgen-dependent and androgen-independent prostate cancer cells via caspase-3 and -8 dependent apoptosis. We also show that VK2 treatment reduces androgen receptor expression and PSA secretion in androgen-dependent prostate cancer cells. Our results also implicate VK2 as a potential anti-inflammatory agent, as several inflammatory genes are downregulated in prostate cancer cells following treatment with VK2. Additionally, AKT and NF-kB levels in prostate cancer cells are reduced significantly when treated with VK2. These findings correlated with the results of the Boyden chamber and angiogenesis assay, as VK2 treatment reduced cell migration and angiogenesis potential of prostate cancer cells. Finally, in a nude mice model, VK2 administration resulted in significant inhibition of both androgen-dependent and androgen-independent tumor growth. Overall, our results suggest that VK2 may be a potential therapeutic agent in the treatment of prostate cancer.
Collapse
|