1
|
Saini S, Gulati N, Awasthi R, Arora V, Singh SK, Kumar S, Gupta G, Dua K, Pahwa R, Dureja H. Monoclonal Antibodies and Antibody-drug Conjugates as Emerging Therapeutics for Breast Cancer Treatment. Curr Drug Deliv 2024; 21:993-1009. [PMID: 37519200 DOI: 10.2174/1567201820666230731094258] [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: 03/10/2023] [Revised: 06/01/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023]
Abstract
When breast cells divide and multiply out of control, it is called breast cancer. Symptoms include lump formation in the breast, a change in the texture or color of the breast, or a discharge from the nipple. Local or systemic therapy is frequently used to treat breast cancer. Surgical and radiation procedures limited to the affected area are examples of local management. There has been significant worldwide progress in the development of monoclonal antibodies (mAbs) since 1986, when the first therapeutic mAb, Orthoclone OKT3, became commercially available. mAbs can resist the expansion of cancer cells by inducing the destruction of cellular membranes, blocking immune system inhibitors, and preventing the formation of new blood vessels. mAbs can also target growth factor receptors. Understanding the molecular pathways involved in tumor growth and its microenvironment is crucial for developing effective targeted cancer therapeutics. Due to their unique properties, mAbs have a wide range of clinical applications. Antibody-drug conjugates (ADCs) are drugs that improve the therapeutic index by combining an antigen-specific antibody with a payload. This review focuses on the therapeutic applications, mechanistic insights, characteristics, safety aspects, and adverse events of mAbs like trastuzumab, bevacizumab, pertuzumab, ertumaxomab, and atezolizumab in breast cancer treatment. The creation of novel technologies utilizing modified antibodies, such as fragments, conjugates, and multi-specific antibodies, must be a central focus of future studies. This review will help scientists working on developing mAbs to treat cancers more effectively.
Collapse
Affiliation(s)
- Swati Saini
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Nisha Gulati
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Rajendra Awasthi
- Department of Pharmaceutical Sciences, School of Health Sciences & Technology, University of Petroleum and Energy Studies (UPES), Bidholi, Dehradun 248 007, Uttarakhand, India
| | - Vimal Arora
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology (MIET), Meerut, Uttar Pradesh, 250005, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Rakesh Pahwa
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| |
Collapse
|
2
|
Li M, Wang Y, Zhang L, Liu Q, Jiang F, Hou W, Wang Y, Fang H, Zhang Y. Cancer Cell Membrane-Enveloped Dexamethasone Normalizes the Tumor Microenvironment and Enhances Gynecologic Cancer Chemotherapy. ACS NANO 2023; 17:16703-16714. [PMID: 37603464 DOI: 10.1021/acsnano.3c03013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
The aberrant tumor microenvironment (TME), especially immature and leaky vessels, prevents the penetration and accumulation of chemotherapeutics and results in the failure of chemotherapy to treat gynecologic cancer. Herein, dexamethasone (Dex), a glucocorticoid steroid used to moderate tumor extracellular matrix and normalize vessels, was enclosed within a biocompatible material known as poly(lactic-co-glycolic acid) (PLGA), and the obtained Dex@PLGA was further coated with a mouse cervical cancer cell membrane (CM). The formulated Dex@PLGA-CM nanoparticles showed efficient extravascular diffusion within the tumor owing to the homologous targeting abilities inherited from the source cancer cells. The Dex@PLGA-CM nanoparticles greatly reshaped the TME, enhancing the penetration of Doxil and thus markedly improving the therapeutic effect of this drug against cervical cancers. Excitingly, the Dex@PLGA-CM nanoparticles coated with mouse ovarian cancer cell membranes also promoted Doxil-mediated chemotherapy effects in metastatic ovarian cancer when administered intraperitoneally. This work presents an effective nanomedicine for the efficient modification of the TME to enhance the effects of gynecologic cancer chemotherapy.
Collapse
Affiliation(s)
- Mingzhuang Li
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| | - Yingyao Wang
- Department of gynecology, Kunshan Maternity and Children's Health Care Hospital, Suzhou 215300, China
| | - Lin Zhang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qin Liu
- Department of gynecology, Kunshan Maternity and Children's Health Care Hospital, Suzhou 215300, China
| | - Feizhou Jiang
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| | - Wenjie Hou
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| | - Yudong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Huapan Fang
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yueming Zhang
- Department of Gynecology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215123, China
| |
Collapse
|
3
|
Eskandarion MR, Tizmaghz Z, Andalib B, Parsa N, Emami SAH, Shahsiah R, Oghabian MA, Shirkoohi R. A case report of the sustained and rapid response of bevacizumab in a TP53-positive breast cancer and liver metastatic patient through personalized medicine. Front Oncol 2022; 12:940678. [PMID: 36119510 PMCID: PMC9479335 DOI: 10.3389/fonc.2022.940678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
Abstract
HER2-positive metastatic breast cancer is much less frequent than other subgroups of breast cancer. Treatment options for this cancer are mostly limited to systemic chemotherapy, which leads to moderate improvements. Targeted therapy against malignant breast cancer requires the identification of reliable biomarkers for personalized medicine to obtain the maximum benefit of this therapy. Any mutations in the TP53 signaling pathway can be considered as a significant causative factor of breast cancer, for which the identification of target genes plays an important role in selecting the appropriate treatment. The use of personalized gene expression profiling could be valuable to find the direct target of the treatment in this case. The present study assessed the genetic profile of an HER2-positive metastatic breast cancer patient (with a liver metastasis) and figured out a complete and sustained response to bevacizumab. According to the results of next-generation sequencing (NGS) analysis, the patient’s genetic profile showed an increased expression of p4EBP1 and PTEN and the activation of the mTOR signaling pathway with a mutation in the TP53 gene. Based on the common treatment of similar profiling, we administrated bevacizumab/Taxol/Gemzar chemotherapy up to six courses. Accordingly, as the response to treatment was revealed by reducing the volume of the liver metastasis from 4 to 1.4 cm, metastasectomy was performed as a complementary treatment. Hence, personalized gene expression profiling not only is useful for targeted therapy but also could be recommended to avoid prescription of non-responsive drugs.
Collapse
Affiliation(s)
- Mohammad Reza Eskandarion
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Tizmaghz
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiation Oncology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Andalib
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Radiation Oncology Central Clinic of Karaj (ROCK), Karaj, Iran
- *Correspondence: Reza Shirkoohi, ; Bahram Andalib,
| | - Nasser Parsa
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Cancer Association, Tehran, Iran
| | - Seyed Amir Hossein Emami
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Hematology and Medical Oncology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shahsiah
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anatomical and Clinical Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oghabian
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Medical Physics Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shirkoohi
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Reza Shirkoohi, ; Bahram Andalib,
| |
Collapse
|
4
|
Jiang P, Zhang Y, Cui J, Wang X, Li Y. Inhibitory effects of icotinib combined with antiangiogenic drugs in human non-small cell lung cancer xenograft models are better than single target drugs. Thorac Cancer 2021; 13:257-264. [PMID: 34855286 PMCID: PMC8758432 DOI: 10.1111/1759-7714.14261] [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: 06/02/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 11/27/2022] Open
Abstract
Background This study aimed to evaluate the inhibitory effects and potential mechanisms of icotinib combined with antiangiogenic drugs on lung adenocarcinoma in vivo. Methods A total of 72 mouse xenograft models established with human lung adenocarcinoma cells (HCC827) were randomly divided into six groups, including control, icotinib (Ic), bevacizumab (Bev), recombinant human endostatin (En), Ic + Bev and Ic + En groups. Mouse weights and tumor volumes were measured regularly. Half of the nude mice in each group were sacrificed after 16 days of drug treatment. The remaining animals were observed for another 16 days without drug supply. Immunohistochemical staining was performed to detect microvessel density in tumor heart, liver, brain specimens from the nude mice and Ki67 expression. Differential expression of vascular endothelial growth factor (VEGFA) in tumor tissue specimens was determined by ELISA and Western blot. Results The results showed that the combined drugs inhibited tumor growth more substantially compared with single drugs, without increasing the toxic effects. The antiangiogenesis effect of the combination was better than that of single drug treatment. In addition, both types of targeted drugs and combination medication not only significantly reduced microvessel density in the tumor tissue itself, but also had a certain impact on decreasing microvessel density in the liver. The combination decreased VEGFA and Ki‐67 amounts significantly more than icotinib or endostatin as a monotherapy. Conclusions Icotinib combined with bevacizumab or rh‐endostatin has a stronger inhibitory effect on tumor growth than single‐target drug in vivo, with no additional side effects.
Collapse
Affiliation(s)
- Peng Jiang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pulmonary and Critical Care Medicine, Weihai Municipal Hospital, Weihai, China
| | - Yan Zhang
- The Fourth People's Hospital of Jinan, Jinan, China
| | - Jiadong Cui
- Department of Pulmonary Medicine, Dong'e County People's Hospital, Liaocheng, China
| | - Xiuxiu Wang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu Li
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
5
|
Chainitikun S, Espinosa Fernandez JR, Long JP, Iwase T, Kida K, Wang X, Saleem S, Lim B, Valero V, Ueno NT. Pathological complete response of adding targeted therapy to neoadjuvant chemotherapy for inflammatory breast cancer: A systematic review. PLoS One 2021; 16:e0250057. [PMID: 33861773 PMCID: PMC8051801 DOI: 10.1371/journal.pone.0250057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/31/2021] [Indexed: 01/06/2023] Open
Abstract
Background The current use of targeted therapy plus neoadjuvant chemotherapy for inflammatory breast cancer (IBC) is based on data extrapolated from studies in non-IBC. We conducted a systematic review to determine whether neoadjuvant chemotherapy plus targeted therapy results in a higher pathologic complete response (pCR) rate than neoadjuvant chemotherapy alone in patients with IBC. Method and findings This systematic review was registered in the PROSPERO register with registration number CRD42018089465. We searched MEDLINE & PubMed, EMBASE, and EBSCO from December 1998 through July 2020. All English-language clinical studies, both randomized and non-randomized, that evaluated neoadjuvant systemic treatment with or without targeted therapy before definitive surgery and reported the pCR results of IBC patients. First reviewer extracted data and assessed the risk of bias using the Risk of Bias In Non-randomized Studies of Interventions tool. Second reviewer confirmed the accuracy. Studies were divided into 3 groups according to systemic treatment: chemotherapy with targeted therapy, chemotherapy alone, and high-dose chemotherapy with hematopoietic stem cell support (HSCS). Of 995 screened studies, 23 with 1,269 IBC patients met the inclusion criteria. For each of the 3 groups of studies, we computed a weighted average of the pCR rates across all studies with confidence interval (CI). The weighted averages (95% CIs) were as follows: chemotherapy with targeted therapy, 31.6% (26.4%-37.3%), chemotherapy alone, 13.0% (10.3%-16.2%), and high-dose chemotherapy with HSCS, 23.0% (18.7%-27.7%). The high pCR by targeted therapy group came from anti-HER2 therapy, 54.4% (44.3%-64.0%). Key limitations of this study included no randomized clinical studies that included only IBC patients. Conclusion Neoadjuvant chemotherapy plus targeted therapy is more effective than neoadjuvant chemotherapy alone for IBC patients. These findings support current IBC standard practice in particular the use of anti-HER2 targeted therapy.
Collapse
Affiliation(s)
- Sudpreeda Chainitikun
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jose Rodrigo Espinosa Fernandez
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - James P. Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Toshiaki Iwase
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Kumiko Kida
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiaoping Wang
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sadia Saleem
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bora Lim
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Vicente Valero
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Naoto T. Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
6
|
Raihan J, Ahmad U, Yong YK, Eshak Z, Othman F, Ideris A. Regression of solid breast tumours in mice by Newcastle disease virus is associated with production of apoptosis related-cytokines. BMC Cancer 2019; 19:315. [PMID: 30947706 PMCID: PMC6449948 DOI: 10.1186/s12885-019-5516-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/25/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Different strains of Newcastle disease virus (NDV) worldwide proved to have tumouricidal activity in several types of cancer cells. However, the possible anti-cancer activity of Malaysian NDV AF2240 strain and its mechanism of action remains unknown. The ability of cytokine-related apoptosis-inducing NDV AF2240 to treat breast cancer was investigated in the current study. METHODS A total of 90 mice were used and divided into 15 groups, each group comprising of 6 mice. Tumour, body weight and mortality of the mice were determined throughout the experiment, to observe the effect of NDV and NDV + tamoxifen treatments on the mice. In addition, the toxic effect of the treatments was determined through liver function test. In order to elucidate the involvement of cytokine production induced by NDV, a total of six cytokines, i.e. IL-6, IFN-γ, MCP-1, IL-10, IL12p70 and TNF-α were measured using cytometric bead array assay (plasma) and enzyme-linked immunosorbent spot (isolated splenocytes). RESULTS The results demonstrated that 4 T1 breast cancer cells in allotransplanted mice treated with AF2240 showed a noticeable inhibition of tumour growth and induce apoptotic-related cytokines. CONCLUSIONS NDV AF2240 suppression of breast tumour growth is associated with induction of apoptotic-related cytokines. It would be important to further investigate the molecular mechanism underlaying cytokines production by Newcastle disease virus.
Collapse
Affiliation(s)
- Juraimi Raihan
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor Malaysia
- Ministry of Health Malaysia, 62590 Putrajaya, Malaysia
| | - Umar Ahmad
- Medical Genetics Laboratory, Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
- Genetics and Cytogenetics Unit, Department of Anatomy, Faculty of Medicine, Bauchi State University, Gadau, PMB 65, Itas/Gadau, Nigeria
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor Malaysia
| | - Zolkapli Eshak
- Faculty of Pharmacy, Universiti Teknologi Mara, 42300 Bandar Puncak Alam, Selangor Malaysia
| | - Fauziah Othman
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor Malaysia
| | - Aini Ideris
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| |
Collapse
|
7
|
Peng JQ, Fumoto S, Suga T, Miyamoto H, Kuroda N, Kawakami S, Nishida K. Targeted co-delivery of protein and drug to a tumor in vivo by sophisticated RGD-modified lipid-calcium carbonate nanoparticles. J Control Release 2019; 302:42-53. [PMID: 30926479 DOI: 10.1016/j.jconrel.2019.03.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/02/2019] [Accepted: 03/25/2019] [Indexed: 01/26/2023]
Abstract
Synchronized bio-distribution of combination therapies has several merits such as synergistic effects and reduced side-effects. Co-delivery of a protein and small molecule drug using a single nanocarrier is challenging because they possess totally different characteristics. Herein, we report the development of sophisticated nanoparticles composed of lipids, calcium carbonate and RGD peptide ligands for the co-delivery of a protein and small molecule drug combination via a simple preparation method. A 'one-step' ethanol injection method was employed to prepare the highly organized nanoparticles. The nanoparticles exhibited a spherical shape with ca. 130 nm diameter, and clearly had an integrated lipid layer covering the periphery. As a ligand, an RGD-modified lipid was post-inserted into the nanoparticles, which was important to overcome the 'PEG dilemma'. The pH-sensitivity of the targeted nanoparticles contributed to the efficient intracellular co-delivery of a protein and drug combination in Colon26 tumor cells, and noticeably improved their accumulation in the tumor region of xenograft mice. Synchronized bio-distribution of the protein and drug was achieved, which was the foundation for the synergistic effects of the combination. The targeting capability of the nanoparticles along with their pH-sensitive drug release and the synchronized bio-distribution of their cargos led to the significant antitumor activity of the SOD and paclitaxel combination in mice. This study provides novel information for the design and preparation of functionalized nanoparticles for the delivery of a protein/drug combination in vivo.
Collapse
Affiliation(s)
- Jian Qing Peng
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan.
| | - Tadaharu Suga
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Hirotaka Miyamoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Naotaka Kuroda
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Shigeru Kawakami
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| |
Collapse
|
8
|
Rawal S, Patel MM. Threatening cancer with nanoparticle aided combination oncotherapy. J Control Release 2019; 301:76-109. [PMID: 30890445 DOI: 10.1016/j.jconrel.2019.03.015] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 12/14/2022]
Abstract
Employing combination therapy has become obligatory in cancer cases exhibiting high tumor load, chemoresistant tumor population, and advanced disease stages. Realization of this fact has now led many of the combination oncotherapies to become an integral part of anticancer regimens. Combination oncotherapy may encompass a combination of anticancer agents belonging to a similar therapeutic category or that of different therapeutic categories (e.g. chemotherapy + gene therapy). Differences in the physicochemical properties, pharmacokinetics and biodistribution pattern of different payloads are the major constraints that are faced by combination chemotherapy. Concordant efforts in the field of nanotechnology and oncology have emerged with several approaches to solve the major issues encountered by combination therapy. Unique colloidal behaviors of various types of nanoparticles and differential targeting strategies have accorded an unprecedented ability to optimize combination oncotherapeutic delivery. Nanocarrier based delivery of the various types of payloads such as chemotherapeutic agents and other anticancer therapeutics such as small interfering ribonucleic acid (siRNA), chemosensitizers, radiosensitizers, and antiangiogenic agents have been addressed in the present review. Various nano-delivery systems like liposomes, polymeric nanoparticles, polymerosomes, dendrimers, micelles, lipid based nanoparticles, prodrug based nanocarriers, polymer-drug conjugates, polymer-lipid hybrid nanoparticles, carbon nanotubes, nanosponges, supramolecular nanocarriers and inorganic nanoparticles (gold nanoparticles, silver nanoparticles, magnetic nanoparticles and mesoporous silica based nanoparticles) that have been extensively explored for the formulation of multidrug delivery is an imperative part of discussion in the review. The present review features the outweighing benefits of combination therapy over mono-oncotherapy and discusses several existent nanoformulation strategies that facilitate a successful combination oncotherapy. Several obstacles that may impede in transforming nanotechnology-based combination oncotherapy from bench to bedside, and challenges associated therein have also been discussed in the present review.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India
| | - Mayur M Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India.
| |
Collapse
|
9
|
Comunanza V, Bussolino F. Therapy for Cancer: Strategy of Combining Anti-Angiogenic and Target Therapies. Front Cell Dev Biol 2017; 5:101. [PMID: 29270405 PMCID: PMC5725406 DOI: 10.3389/fcell.2017.00101] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022] Open
Abstract
The concept that blood supply is required and necessary for cancer growth and spreading is intuitive and was firstly formalized by Judah Folkman in 1971, when he demonstrated that cancer cells release molecules able to promote the proliferation of endothelial cells and the formation of new vessels. This seminal result has initiated one of the most fascinating story of the medicine, which is offering a window of opportunity for cancer treatment based on the use of molecules inhibiting tumor angiogenesis and in particular vascular-endothelial growth factor (VEGF), which is the master gene in vasculature formation and is the commonest target of anti-angiogenic regimens. However, the clinical results are far from the remarkable successes obtained in pre-clinical models. The reasons of this discrepancy have been partially understood and well addressed in many reviews (Bergers and Hanahan, 2008; Bottsford-Miller et al., 2012; El-Kenawi and El-Remessy, 2013; Wang et al., 2015; Jayson et al., 2016). At present anti-angiogenic regimens are not used as single treatments but associated with standard chemotherapies. Based on emerging knowledge of the biology of VEGF, here we sustain the hypothesis of the efficacy of a dual approach based on targeting pro-angiogenic pathways and other druggable targets such as mutated oncogenes or the immune system.
Collapse
Affiliation(s)
- Valentina Comunanza
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
10
|
High-circulating Tie2 Is Associated With Pathologic Complete Response to Chemotherapy and Antiangiogenic Therapy in Breast Cancer. Am J Clin Oncol 2017; 39:248-54. [PMID: 24577164 DOI: 10.1097/coc.0000000000000046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Vascular endothelial growth factor (VEGF) is a central mediator of angiogenesis in breast cancer. Research in antiangiogenic cancer treatment has been marked by the development of the monoclonal antibody bevacizumab, which targets VEGF in many solid tumors. As patients do not equally benefit from bevacizumab, it has become necessary to define the profile of patients who will benefit from the drug. MATERIALS AND METHODS We have conducted a prospective phase II study in 39 patients using bevacizumab in breast cancer in the neoadjuvant setting, and found improved pathologic complete response (pCR) when bevacizumab was added to chemotherapy in patients with hormone receptor negative and invasive ductal carcinoma. Blood samples were collected at baseline and serially while patients were on treatment. Circulating angiogenesis-related proteins angiopoietin (ANG)1, ANG2, basic fibroblast growth factor, IL-1a, matrix metalloproteinase 9, platelet derived growth factor - BB, platelet endothelial cell adhesion molecule -1, Tie2, VEGF, and vascular endothelial growth factor receptor 2 were measured at baseline and during treatment. This correlative study was conducted to identify specific serum angiogenic factor profiles that might be associated with pCR in the neoadjuvant setting in breast cancer patients receiving bevacizumab and chemotherapy. RESULTS Elevated baseline serum Tie2 and basic fibroblast growth factor were associated with pCR in response to this combination. Changes in serum levels of these proteins were seen during treatment but were not significantly different between the pCR and non-pCR groups. CONCLUSIONS Baseline-circulating Tie2 levels may help distinguish patients who will have pCR from those who will not and may form the basis for future development of antiangiogenic therapy in breast cancer. Larger studies are needed to validate these findings. ClinicalTrials.gov Identifier: NCT00203502.
Collapse
|
11
|
Jiang H, Rugo HS. Human epidermal growth factor receptor 2 positive (HER2+) metastatic breast cancer: how the latest results are improving therapeutic options. Ther Adv Med Oncol 2015; 7:321-39. [PMID: 26557900 PMCID: PMC4622301 DOI: 10.1177/1758834015599389] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human epidermal growth factor receptor 2 positive (HER2+) metastatic breast cancer (MBC) remains an incurable disease, and approximately 25% of patients with HER2+ early breast cancer still relapse after adjuvant trastuzumab-based treatment. HER2 is a validated therapeutic target that remains relevant throughout the disease process. Recently, a number of novel HER2 targeted agents have become available, including lapatinib (a small molecule tyrosine kinase inhibitor of both HER2 and the epidermal growth factor receptor), pertuzumab (a new anti-HER2 monoclonal antibody) and ado-trastuzumab emtansine (T-DM1, a novel antibody-drug conjugate), which provide additional treatment options for patients with HER2+ MBC. The latest clinical trials have demonstrated improved outcome with treatment including pertuzumab or T-DM1 compared with standard HER2 targeted therapy. Here we review the clinical development of approved and investigational targeted agents for the treatment of HER2+ MBC, summarize the latest results of important clinical trials supporting use of these agents in the treatment of HER2+ MBC, and discuss how these results impact therapeutic options in clinical practice.
Collapse
Affiliation(s)
- Hanfang Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hope S. Rugo
- University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, 1600 Divisadero St, Box 1710 San Francisco CA 94115, USA
| |
Collapse
|
12
|
Zhao S, Ma W, Xu L, Wu X, Kuang H, Wang L, Xu C. Ultrasensitive SERS detection of VEGF based on a self-assembled Ag ornamented–AU pyramid superstructure. Biosens Bioelectron 2015; 68:593-597. [DOI: 10.1016/j.bios.2015.01.056] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/13/2015] [Accepted: 01/23/2015] [Indexed: 10/24/2022]
|
13
|
Foy KC, Miller MJ, Moldovan N, Carson WE, Kaumaya PTP. Combined vaccination with HER-2 peptide followed by therapy with VEGF peptide mimics exerts effective anti-tumor and anti-angiogenic effects in vitro and in vivo. Oncoimmunology 2014; 1:1048-1060. [PMID: 23170253 PMCID: PMC3494619 DOI: 10.4161/onci.20708] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Overexpression of HER-2 and VEGF plays a key role in the development and metastasis of several human cancers. Many FDA-approved therapies targeting both HER-2 (Trastuzumab, Herceptin) and VEGF (Bevacizumab, Avastin) are expensive, have unacceptable toxicities and are often associated with the development of resistance. Here, we evaluate the dual antitumor effects of combining designed particular HER-2 peptide vaccine with VEGF peptide mimics. In vitro, HER-2 phosphorylation and antibody-dependent cellular toxicity were used to validate whether combining HER-2- and VEGF-targeting therapies would be effective. Moreover, a two-pronged approach was tested in vivo: (1) active immunotherapy with conformational HER-2 B-cell epitope vaccines and (2) anti-angiogenic therapy with a peptide structured to mimic VEGF. A transplantable BALB/c mouse model challenged with TUBO cells was used to test the effects of the HER-2 peptide vaccine combined with VEGF peptide mimics. Tumor sections after treatment were stained for blood vessel density and actively dividing cells. Our results show that immunization with an HER-2 peptide epitope elicits high affinity HER-2 native antibodies that are effective in inhibiting tumor growth in vivo, an effect that is enhanced by VEGF peptide mimics. We demonstrate that the combination of HER-2 and VEGF peptides induces potent anti-tumor and anti-angiogenic responses.
Collapse
Affiliation(s)
- Kevin C Foy
- Department of Microbiology; The Ohio State University; Columbus, OH USA ; Department of Obstetrics and Gynecology; The Ohio State University; Columbus, OH USA
| | - Megan J Miller
- Department of Microbiology; The Ohio State University; Columbus, OH USA ; Department of Obstetrics and Gynecology; The Ohio State University; Columbus, OH USA
| | - Nicanor Moldovan
- Department of Internal Medicine; Division of Cardivascular Medicine; The Ohio State University; Columbus, OH USA
| | - William E Carson
- James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center; The Ohio State University; Columbus, OH USA
| | - Pravin T P Kaumaya
- Department of Microbiology; The Ohio State University; Columbus, OH USA ; Department of Obstetrics and Gynecology; The Ohio State University; Columbus, OH USA ; James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center; The Ohio State University; Columbus, OH USA
| |
Collapse
|
14
|
Chen HC, Qiu JT, Yang FL, Liu YC, Chen MC, Tsai RY, Yang HW, Lin CY, Lin CC, Wu TS, Tu YM, Xiao MC, Ho CH, Huang CC, Lai CS, Hua MY. Magnetic-Composite-Modified Polycrystalline Silicon Nanowire Field-Effect Transistor for Vascular Endothelial Growth Factor Detection and Cancer Diagnosis. Anal Chem 2014; 86:9443-50. [DOI: 10.1021/ac5001898] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hsiao-Chien Chen
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Biochemistry, School of Medicine, Taipei Medical University, Taipei 11031, Taiwan, Republic of China
| | - Jian-Tai Qiu
- Department
of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan, Republic of China
| | - Fu-Liang Yang
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Yin-Chih Liu
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Min-Cheng Chen
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Rung-Ywan Tsai
- Electronics
and Optoelectronics Research Laboratories, Industrial Technology Research Institute, Hsinchu 31040, Taiwan, Republic of China
| | - Hung-Wei Yang
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Chia-Yi Lin
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Chu-Chi Lin
- Department
of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan, Republic of China
| | - Tzong-Shoon Wu
- Department
of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
- Department
of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan, Republic of China
| | - Yi-Ming Tu
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Min-Cong Xiao
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Chia-Hua Ho
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Chien-Chao Huang
- National Nano Device Laboratories, Hsinchu Science Park, Hsinchu 30078, Taiwan, Republic of China
| | - Chao-Sung Lai
- Department
of Electronic Engineering and Biosensor Group,
Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| | - Mu-Yi Hua
- Department
of Chemical and Materials Engineering and Biosensor Group, Biomedical Engineering Research Center, Chang Gung University, Taoyuan 33302, Taiwan, Republic of China
| |
Collapse
|
15
|
Vu T, Sliwkowski MX, Claret FX. Personalized drug combinations to overcome trastuzumab resistance in HER2-positive breast cancer. Biochim Biophys Acta Rev Cancer 2014; 1846:353-65. [PMID: 25065528 DOI: 10.1016/j.bbcan.2014.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/11/2014] [Accepted: 07/11/2014] [Indexed: 12/13/2022]
Abstract
HER2-positive (HER2+) breast cancer accounts for 18%-20% of all breast cancer cases and has the second poorest prognosis among breast cancer subtypes. Trastuzumab, the first Food and Drug Administration-approved targeted therapy for breast cancer, established the era of personalized treatment for HER2+ metastatic disease. It is well tolerated and improves overall survival and time-to-disease progression; with chemotherapy, it is part of the standard of care for patients with HER2+ metastatic disease. However, many patients do not benefit from it because of resistance. Substantial research has been performed to understand the mechanism of trastuzumab resistance and develop combination strategies to overcome the resistance. In this review, we provide insight into the current pipeline of drugs used in combination with trastuzumab and the degree to which these combinations have been evaluated, especially in patients who have experienced disease progression on trastuzumab. We conclude with a discussion of the current challenges and future therapeutic approaches to trastuzumab-based combination therapy.
Collapse
Affiliation(s)
- Thuy Vu
- Department of Systems Biology, Unit 950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; Experimental Therapeutics Academic Program, The University of Texas Graduate School of Biomedical Sciences at Houston, 6767 Bertner Ave., Houston, TX 77030, USA
| | | | - Francois X Claret
- Department of Systems Biology, Unit 950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; Experimental Therapeutics Academic Program, The University of Texas Graduate School of Biomedical Sciences at Houston, 6767 Bertner Ave., Houston, TX 77030, USA.
| |
Collapse
|
16
|
Rego SL, Helms RS, Dréau D. Breast tumor cell TACE-shed MCSF promotes pro-angiogenic macrophages through NF-κB signaling. Angiogenesis 2013; 17:573-85. [PMID: 24197832 DOI: 10.1007/s10456-013-9405-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 10/28/2013] [Indexed: 12/12/2022]
Abstract
Most deaths associated with breast cancer, the most common malignancy in women, are caused by metastasis. Tumor associated macrophages significantly contribute to breast cancer progression and development of metastasis through the promotion of angiogenesis which involves a central regulator of macrophage functions: nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Macrophages are activated by macrophage colony stimulating factor (MCSF) and chemokine (C-C motif) ligand 2 (CCL2) to secrete angiogenic factors including vascular endothelial growth factor (VEGF). The release of MCSF from tumor cells is mediated by ectodomain shedding through tumor necrosis factor alpha converting enzyme activation (TACE). Here we determined whether tumor cells TACE-shed MCSF promotes angiogenesis through activation of the NF-κB pathway in macrophages and the subsequent release of VEGF. These interactions were modeled in vitro using a panel of mammary cells mimicking the breast cancer progression from normal murine mammary gland cells to metastatic 4T1 cells along with J774 macrophages, all derived from BALB/c mice. TACE and MCSF expressions were higher in metastatic cells compared to epithelial cells (p < 0.05). Tumor conditioned medias activated the expression of VEGF by macrophages through stimulation of the NF-κB pathway and resulting macrophage secretions that promoted high levels of endothelial cell tubes. Furthermore, the combinations of CCL2, also highly expressed by tumor cells, and MCSF promoted pro-angiogenic macrophages. These results highlight the key role of tumor cell TACE-shed MCSF and secreted CCL2 in stimulating pro-angiogenic macrophages.
Collapse
Affiliation(s)
- Stephen L Rego
- Cell and Molecular Division, Department of Biology, University of North Carolina, Charlotte, 9201 University City Blvd., Charlotte, NC, 28223, USA
| | | | | |
Collapse
|
17
|
Hasan N, Mann A, Ferrari M, Tanaka T. Mesoporous silicon particles for sustained gene silencing. Methods Mol Biol 2013; 1049:481-93. [PMID: 23913239 DOI: 10.1007/978-1-62703-547-7_36] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
RNA interference (RNAi) is a powerful approach for silencing oncogenes; however, in vivo RNAi delivery has remained a major challenge due to lack of safe, efficient, and sustained delivery. Here, we describe a novel approach to overcome these limitations using mesoporous silicon particles loaded with nanoparticles (i.e., liposomes) containing small interfering RNA (siRNA) targeted against oncoprotein that contributes to cancer cell survival. This delivery method resulted in sustained gene silencing for at least 3 weeks with substantial reduction of tumor growth with no overt toxicities in two independent orthotopic mouse models of ovarian cancer following a single intravenous administration of mesoporous silicon particles loaded with liposomal EphA2-siRNA.
Collapse
Affiliation(s)
- Nafis Hasan
- Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
18
|
Sorace AG, Saini R, Mahoney M, Hoyt K. Molecular ultrasound imaging using a targeted contrast agent for assessing early tumor response to antiangiogenic therapy. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2012; 31:1543-50. [PMID: 23011617 PMCID: PMC3464103 DOI: 10.7863/jum.2012.31.10.1543] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
OBJECTIVES Contrast-enhanced ultrasound (US) and targeted microbubbles have been shown to be advantageous for angiogenesis evaluation and disease staging in cancer. This study explored molecular US imaging of a multitargeted microbubble for assessing the early tumor response to antiangiogenic therapy. METHODS Target receptor expression of 2LMP breast cancer cells was quantified by flow cytometric analysis and characterization established with antibodies against mouse α(V)β3- integrin, P-selectin, and vascular endothelial growth factor receptor 2. Tumor-bearing mice (n = 15 per group) underwent contrast-enhanced US imaging of multitargeted microbubbles. Microbubble accumulation was calculated by destruction-replenishment techniques and time-intensity curve analysis. On day 0, mice underwent baseline imaging. Next, therapy group mice were injected with a 0.2-mg dose of bevacizumab, and controls received matched saline injections. Imaging was repeated on days 1 and 3. After imaging was completed on day 3, the mice were euthanized and tumors excised. Histologic analysis of microvessel density and intratumoral necrosis was completed on tumor sections. RESULTS On day 3 after bevacizumab dosing, a 71.8% change in tumor vasculature was shown between the therapy and control groups (P = .01). The therapy group had a 15.4% decrease in tumor vascularity, whereas the control group had a 56.4% increase. CONCLUSIONS Molecular US imaging of angiogenic markers can detect the early tumor response to drug therapy.
Collapse
Affiliation(s)
- Anna G Sorace
- MBA, Department of Biomedical Engineering, University of Alabama at Birmingham, G082 Volker Hall, 1670 University Blvd, Birmingham, AL 35294-0019, USA
| | | | | | | |
Collapse
|
19
|
Suppression of tumor growth in xenograft model mice by small interfering RNA targeting osteopontin delivery using biocompatible poly(amino ester). Int J Pharm 2012; 431:197-203. [DOI: 10.1016/j.ijpharm.2012.04.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/30/2012] [Accepted: 04/09/2012] [Indexed: 01/16/2023]
|
20
|
Lee SW, Park YW. Expression of endoglin and podoplanin in early and advanced oral squamous cell carcinoma. J Korean Assoc Oral Maxillofac Surg 2012. [DOI: 10.5125/jkaoms.2012.38.3.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Sang-Woon Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Gangneung-Wonju National University, Gangneung, Korea
| | - Young-Wook Park
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Gangneung-Wonju National University, Gangneung, Korea
| |
Collapse
|
21
|
Falanga A, Russo L. Epidemiology, risk and outcomes of venous thromboembolism in cancer. Hamostaseologie 2011; 32:115-25. [PMID: 21971578 DOI: 10.5482/ha-1170] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 07/11/2011] [Indexed: 12/16/2022] Open
Abstract
Cancer is associated with a fourfold increased risk of venous thromboembolism (VTE). The risk of VTE varies according to the type of malignancy (i. e. pancreatic cancer, brain cancer, lymphoma) and its disease stage and individual factors (i. e. sex, race, age, previous VTE history, immobilization, obesity). Preventing cancer-associated VTE is important because it represents a significant cause of morbidity and mortality. In order to identify cancer patient at particularly high risk, who need thromboprophylaxis, risk prediction models have become available and are under validation. These models include clinical risk factors, but also begin to incorporate biological markers. The major American and European scientific societies have issued their recommendations to guide the management of VTE in patients with cancer. In this review the principal aspects of epidemiology, risk factors and outcome of cancer-associated VTE are summarized.
Collapse
Affiliation(s)
- A Falanga
- Division of Immunohematology and Transfusion Medicine, Department Oncology-Hematology, Ospedali Riuniti, Bergamo, Italy.
| | | |
Collapse
|
22
|
Rugo HS, Stopeck AT, Joy AA, Chan S, Verma S, Lluch A, Liau KF, Kim S, Bycott P, Rosbrook B, Bair AH, Soulieres D. Randomized, Placebo-Controlled, Double-Blind, Phase II Study of Axitinib Plus Docetaxel Versus Docetaxel Plus Placebo in Patients With Metastatic Breast Cancer. J Clin Oncol 2011; 29:2459-65. [DOI: 10.1200/jco.2010.31.2975] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose This multicenter, randomized, double-blind, phase II study assessed safety and efficacy of axitinib plus docetaxel in metastatic breast cancer (MBC). Patients and Methods Women with MBC were randomly assigned 2:1 to receive docetaxel 80 mg/m2 once every 3 weeks plus axitinib 5 mg twice per day (combination arm) or placebo (placebo arm), following a lead-in phase I trial. The primary end point was time to progression (TTP). Results In all, 168 patients were enrolled; 112 were randomly assigned to axitinib and 56 to placebo. Median TTP was numerically longer in the combination arm than in the placebo arm (8.1 v 7.1 months), but this difference was not statistically significant (hazard ratio, 1.24; 95% CI, 0.82 to 1.87; one-sided P = .156). The difference in median TTP was greatest among patients who had received prior adjuvant chemotherapy (9.2 v 7.0 months; P = .043, prespecified subgroup analysis). Objective response rate was higher in the combination arm (41.1% v 23.6%; P = .011). The most common grades 3 to 4 treatment-related adverse events (combination/placebo) included diarrhea (10.8%/0%), fatigue (10.8%/5.4%), stomatitis (12.6%/1.8%), mucositis (9.0%/0%), asthenia (7.2%/0%), and hypertension (4.5%/0%). Three patients in the combination arm experienced serious thromboembolic events (one death). Febrile neutropenia was more frequent in the combination arm (15.3% v 7.1%); rates of other hematologic toxicities were comparable. Increased toxicity with axitinib was generally managed by dose reduction and/or growth factor support. Conclusion The addition of axitinib to docetaxel did not improve TTP in first-line MBC treatment. Combination therapy may be more effective in patients previously exposed to adjuvant chemotherapy.
Collapse
Affiliation(s)
- Hope S. Rugo
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Alison T. Stopeck
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Anil A. Joy
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Stephen Chan
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Shailendra Verma
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Anna Lluch
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Katherine F. Liau
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Sinil Kim
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Paul Bycott
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Brad Rosbrook
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Angel H. Bair
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Denis Soulieres
- From the University of California, San Francisco Helen Diller Family Comprehensive Cancer Center; Pfizer Oncology, San Diego, CA; University of Arizona, Arizona Cancer Center, Tucson, AZ; Cross Cancer Institute, Edmonton, Alberta; Ottawa Regional Cancer Center, Ottawa, Ontario; Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Nottingham City Hospital, Nottingham, United Kingdom; and Hospital Clinico Universitario de Valencia, Valencia, Spain
| |
Collapse
|
23
|
Foy KC, Liu Z, Phillips G, Miller M, Kaumaya PTP. Combination treatment with HER-2 and VEGF peptide mimics induces potent anti-tumor and anti-angiogenic responses in vitro and in vivo. J Biol Chem 2011; 286:13626-37. [PMID: 21325276 PMCID: PMC3075707 DOI: 10.1074/jbc.m110.216820] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 02/07/2011] [Indexed: 12/27/2022] Open
Abstract
HER-2 is a member of the EGF receptor family and is overexpressed in 20-30% of breast cancers. HER-2 overexpression causes increased expression of VEGF at both the RNA and protein levels. HER-2 and VEGF are therefore considered good targets for cancer treatment, which has led to the development of two humanized monoclonal antibodies (mAb) pertuzumab and bevacizumab. Although passive immunotherapy with these Abs are approved for treatment of advanced breast cancer, a number of concerns exist. Treatment is expensive, has a limited duration of action, and is usually accompanied by serious side effects. We hypothesized that therapy with conformational peptide mimics aimed at blocking receptor-ligand interaction is potentially safer with little toxicity, cheaper with a longer half-life, and has greater penetrating abilities than mAbs. We designed and synthesized peptides based on the binding of HER-2 with pertuzumab and VEGF with VEGFR2. We show that treatment with the peptide mimics induces potent anti-tumor responses in vitro as determined by cell viability, proliferation, and HER2 phosphorylation assays. We also demonstrate in a transplantable BALB/c mouse tumor model that treatment with the peptide mimics resulted in a greater delay in tumor growth and development. Similarly, treatment with the peptide mimics inhibited angiogenesis in vivo as assessed by a Matrigel plug assay. To address the problem of degradability of L-amino acid peptides in vivo, we synthesized the retro-inverso D-peptide mimics that resulted in higher efficacy in treatment. Our study shows that combination treatment with HER-2 and VEGF peptide mimics provides greater efficacy than individual treatments.
Collapse
MESH Headings
- Angiogenesis Inhibitors/chemical synthesis
- Angiogenesis Inhibitors/chemistry
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antineoplastic Combined Chemotherapy Protocols/chemistry
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Biomimetic Materials/chemical synthesis
- Biomimetic Materials/chemistry
- Biomimetic Materials/pharmacokinetics
- Drug Screening Assays, Antitumor/methods
- Female
- Humans
- Mice
- Mice, Inbred BALB C
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Peptides/chemical synthesis
- Peptides/chemistry
- Peptides/pharmacology
- Receptor, ErbB-2
- Vascular Endothelial Growth Factor A
Collapse
Affiliation(s)
- Kevin C. Foy
- From the Department of Microbiology
- Department of Obstetrics and Gynecology, and
| | | | - Gary Phillips
- Arthur G. James Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210
| | - Megan Miller
- From the Department of Microbiology
- Department of Obstetrics and Gynecology, and
| | - Pravin T. P. Kaumaya
- From the Department of Microbiology
- Ohio State Biochemistry Program
- Department of Obstetrics and Gynecology, and
- Arthur G. James Comprehensive Cancer Center, Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
24
|
Mukherjee SD, Swystun LL, Mackman N, Wang JG, Pond G, Levine MN, Liaw PC. Impact of chemotherapy on thrombin generation and on the protein C pathway in breast cancer patients. PATHOPHYSIOLOGY OF HAEMOSTASIS AND THROMBOSIS 2011; 37:88-97. [PMID: 21430357 DOI: 10.1159/000324166] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 01/06/2011] [Indexed: 12/19/2022]
Abstract
Although thromboembolism is a problematic complication of chemotherapy, the pathogenic mechanisms by which chemotherapeutic agents exert prothrombotic effects in vivo are unclear.The objective of this study was to examine the effects of adjuvant chemotherapy on thrombin generation, the protein C anticoagulant pathway, and microparticle tissue factor (MP TF) activity in 26 breast cancer patients (stages I to III). The patients received cyclophosphamide, 5-fluorouracil, and methotrexate, epirubicin, or doxorubicin. Plasma samples were collected on day 1 (baseline), day 2, and day 8 for the first 2 cycles of chemotherapy. Levels of thrombin-antithrombin (TAT) complexes, MP TF activity, and components of the protein C anticoagulant pathway, including protein C, activated protein C (APC), soluble thrombomodulin (sTM), and soluble endothelial protein C receptor (sEPCR), were measured. Compared to prechemotherapy baseline levels, plasma TAT, protein C, and APC were significantly different following the administration of chemotherapy (p < 0.01 for each). Plasma TAT was higher in cycle 1, day 2, and cycle 2, day 8, compared to baseline. Plasma protein C levels were lower in cycle 2, day 8, whereas plasma APC levels were lower in cycle 2, day 1, and cycle 2, day 8. No significant changes were found in plasma sEPCR, sTM, or MP TF activity. This study suggests that adjuvant chemotherapy in women with breast cancer increases thrombin generation and impairs the endothelium-based protein C anticoagulant pathway.
Collapse
Affiliation(s)
- Som D Mukherjee
- Department of Oncology, McMaster University, Hamilton, Ont., Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Agnelli G, Verso M. Thromboprophylaxis during chemotherapy in patients with advanced cancer. Thromb Res 2010; 125 Suppl 2:S17-20. [PMID: 20433999 DOI: 10.1016/s0049-3848(10)70007-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Venous thromboembolism is a common and potentially fatal complication in patients with advanced cancer. In these patients chemotherapy has been identified as an independent risk factor for venous thromboembolism. The annual incidence of venous thromboembolism in cancer patients who receive chemotherapy is estimated to be about 10%. This risk increases up to 15-20% depending on type and combination of anticancer agents. The occurrence of venous thromboembolic events in cancer patients is associated with a poor prognosis. The benefit of antithrombotic prophylaxis for venous thromboembolism in cancer patients who receive chemotherapy has been recently evaluated in a randomized, placebo-controlled, double-blind trial (the PROTECHT study). The aim of this study was to evaluate the efficacy and safety of the low molecular weight heparin nadroparin in the prevention of venous and arterial thromboembolic events during chemotherapy. The results of the PROTECHT study demonstrated that nadroparin reduces by 50% the rate of thromboembolic events in ambulatory patients with metastatic or locally advanced cancer while they receiving chemotherapy. The antithrombotic effect was more evident in patients with lung and gastrointestinal cancer. In future studies the clinical benefit of antithrombotic prophylaxis should be evaluated in cancer patients at particularly high risk for thromboembolism.
Collapse
Affiliation(s)
- Giancarlo Agnelli
- Internal and Cardiovascular Medicine - Stroke Unit, University of Perugia, Perugia, Italy.
| | | |
Collapse
|
26
|
Singh G, Kumar P, Parshad R, Seith A, Thulkar S, Hosten N. Role of color Doppler indices in predicting disease-free survival of breast cancer patients during neoadjuvant chemotherapy. Eur J Radiol 2010; 75:e158-62. [DOI: 10.1016/j.ejrad.2009.12.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 12/18/2009] [Indexed: 10/20/2022]
|
27
|
Ogba N, Doughman YQ, Chaplin LJ, Hu Y, Gargesha M, Watanabe M, Montano MM. HEXIM1 modulates vascular endothelial growth factor expression and function in breast epithelial cells and mammary gland. Oncogene 2010; 29:3639-49. [PMID: 20453883 PMCID: PMC2892028 DOI: 10.1038/onc.2010.110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recently, we found that mutation of the C-terminus of transcription factor Hexamethylene bisacetamide inducible protein 1 (HEXIM1) in mice leads to abnormalities in cardiovascular development due to aberrant vascular endothelial growth factor (VEGF) expression. HEXIM1 regulation of some genes has also been shown to be positive transcription elongation factor b (P-TEFb)-dependent. However, it is not known whether HEXIM1 regulates VEGF in the mammary gland. We demonstrate that HEXIM1 regulates estrogen-induced VEGF transcription via inhibition of Estrogen Receptor alpha recruitment to the VEGF promoter in a P-TEFb-independent manner in MCF-7 cells. Under hypoxic conditions, HEXIM1 inhibits estrogen-induced Hypoxia-inducible factor-1 alpha (HIF-1α) protein expression and recruitment of HIF-1α to the hypoxia response element in the VEGF promoter. In the mouse mammary gland, increased HEXIM1 expression decreased estrogen-driven VEGF and HIF-1α expression. Conversely, a mutation in the C-terminus of HEXIM1 (HEXIM11-312) led to increased VEGF and HIF-1α expression and vascularization in mammary glands of heterozygous HEXIM11-312 mice when compared to their wild-type littermates. Additionally, HEXIM11-312 mice have a higher incidence of carcinogen-induced mammary tumors with increased vascularization, suggesting an inhibitory role for HEXIM1 during angiogenesis. Taken together, our data provide evidence to suggest a novel role for HEXIM1 in cancer progression.
Collapse
Affiliation(s)
- N Ogba
- Department of Pharmacology, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Tamaki K, Sasano H, Maruo Y, Takahashi Y, Miyashita M, Moriya T, Sato Y, Hirakawa H, Tamaki N, Watanabe M, Ishida T, Ohuchi N. Vasohibin-1 as a potential predictor of aggressive behavior of ductal carcinoma in situ of the breast. Cancer Sci 2010; 101:1051-8. [PMID: 20704578 PMCID: PMC11158447 DOI: 10.1111/j.1349-7006.2009.01483.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Vasohibin-1 is a recently identified negative feedback regulator of angiogenesis induced by VEGF-A and bFGF. In this study, we first evaluated mRNA expression of vasohibin-1 and CD31 in 39 Japanese female breast carcinoma specimens including 22 invasive ductal carcinoma (IDC) and 17 ductal carcinoma in situ (DCIS) using a real-time quantitative RT-PCR (QRT-PCR) with LightCycler system. In addition, we also immunolocalized vasohibin-1 and CD31 and compared their immunoreactivity to nuclear grades and histological grades of 100 carcinoma cases (50 IDC and 50 DCIS). There were no statistically significant differences of CD31 mRNA expression and the number of CD31 positive vessels between DCIS and IDC (P = 0.250 and P = 0.191, respectively), whereas there was a statistically significant difference in vasohibin-1 mRNA expression and the number of vasohibin-1 positive vessels in DCIS and IDC (P = 0.022 and P < or = 0.001, respectively). There was a significant positive correlation between vasohibin-1 mRNA level and Ki-67 labeling index in DCIS (r(2) = 0.293, P < or = 0.001). In addition, vasohibin-1 mRNA expression was correlated with high nuclear and histological grades in DCIS cases and a significant positive correlation was detected between the number of vasohibin-1 positive vessels and Ki-67 labeling index or nuclear grade or Van Nuys classification of carcinoma cells (P < or = 0.001, respectively). These results all indicate the possible correlation between aggressive biological features in DCIS including increased tumor cell proliferation and the status of neovascularization determined by vasohibin-1 immunoreactivity.
Collapse
Affiliation(s)
- Kentaro Tamaki
- Department of Surgical Oncology, Tohoku University Graduated School of Medicine, Miyagi, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bukowski RM. Metastatic clear cell carcinoma of the kidney: therapeutic role of bevacizumab. Cancer Manag Res 2010; 2:83-96. [PMID: 21188099 PMCID: PMC3004565 DOI: 10.2147/cmar.s7540] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Indexed: 01/28/2023] Open
Abstract
The biology and pathogenesis of clear cell carcinoma of the kidney has been extensively investgated, and the role of von Hipple-Landau gene inactivation and tumor associated angiogenesis is now recognized. Development of vascular endothelial growth factor inhibitors and phase 3 clinical trials utilizing this class of agents has produced a new treatment paradigm for patients with metastatic renal cell carcinoma (RCC). One of the active regimens identified is the combination of bevacizumab and interferon-α. Recently published reports provided evidence of the clinical and biologic activity of this therapy. The current manuscript reviews the background and rationale for the activity of bevacizumab in RCC, and results from recent clinical trials with this agent alone or in combination with targeted agents or cytokines. The role of this therapy in contrast to other targeted agents is reviewed, and the potential utility as well as questions raised by recent studies are discussed.
Collapse
Affiliation(s)
- Ronald M Bukowski
- Cleveland Clinic Taussig Cancer Center, CCF Lerner College of Medicine of CWRU Cleveland, OH, USA
| |
Collapse
|
30
|
Affiliation(s)
- Pradip De
- Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Brian Leyland-Jones
- Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
31
|
Tamaki K, Sasano H, Maruo Y, Takahashi Y, Miyashita M, Moriya T, Sato Y, Hirakawa H, Tamaki N, Watanabe M, Ishida T, Ohuchi N. Vasohibin-1 as a potential predictor of aggressive behavior of ductal carcinomain situof the breast. Cancer Sci 2010. [DOI: 10.1111/j.1349-7006.2010.01483.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
32
|
Lowengrub JS, Frieboes HB, Jin F, Chuang YL, Li X, Macklin P, Wise SM, Cristini V. Nonlinear modelling of cancer: bridging the gap between cells and tumours. NONLINEARITY 2010; 23:R1-R9. [PMID: 20808719 PMCID: PMC2929802 DOI: 10.1088/0951-7715/23/1/r01] [Citation(s) in RCA: 224] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Despite major scientific, medical and technological advances over the last few decades, a cure for cancer remains elusive. The disease initiation is complex, and including initiation and avascular growth, onset of hypoxia and acidosis due to accumulation of cells beyond normal physiological conditions, inducement of angiogenesis from the surrounding vasculature, tumour vascularization and further growth, and invasion of surrounding tissue and metastasis. Although the focus historically has been to study these events through experimental and clinical observations, mathematical modelling and simulation that enable analysis at multiple time and spatial scales have also complemented these efforts. Here, we provide an overview of this multiscale modelling focusing on the growth phase of tumours and bypassing the initial stage of tumourigenesis. While we briefly review discrete modelling, our focus is on the continuum approach. We limit the scope further by considering models of tumour progression that do not distinguish tumour cells by their age. We also do not consider immune system interactions nor do we describe models of therapy. We do discuss hybrid-modelling frameworks, where the tumour tissue is modelled using both discrete (cell-scale) and continuum (tumour-scale) elements, thus connecting the micrometre to the centimetre tumour scale. We review recent examples that incorporate experimental data into model parameters. We show that recent mathematical modelling predicts that transport limitations of cell nutrients, oxygen and growth factors may result in cell death that leads to morphological instability, providing a mechanism for invasion via tumour fingering and fragmentation. These conditions induce selection pressure for cell survivability, and may lead to additional genetic mutations. Mathematical modelling further shows that parameters that control the tumour mass shape also control its ability to invade. Thus, tumour morphology may serve as a predictor of invasiveness and treatment prognosis.
Collapse
Affiliation(s)
- J S Lowengrub
- Department of Biomedical Engineering, Center for Mathematical and Computational Biology, University of California at Irvine, Irvine, CA 92697, USA
| | - H B Frieboes
- School of Health Information Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
- Department of Mathematics, University of California at Irvine, Irvine, CA 92697, USA
| | - F Jin
- School of Health Information Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
- Department of Mathematics, University of California at Irvine, Irvine, CA 92697, USA
| | - Y-L Chuang
- School of Health Information Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| | - X Li
- Department of Mathematics, University of California at Irvine, Irvine, CA 92697, USA
| | - P Macklin
- School of Health Information Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| | - S M Wise
- Department of Mathematics, University of Tennessee, Knoxville, TN 37996, USA
| | - V Cristini
- School of Health Information Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
33
|
Combination therapy: opportunities and challenges for polymer-drug conjugates as anticancer nanomedicines. Adv Drug Deliv Rev 2009; 61:1203-13. [PMID: 19699247 DOI: 10.1016/j.addr.2009.05.006] [Citation(s) in RCA: 494] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 05/14/2009] [Indexed: 11/23/2022]
Abstract
The discovery of new molecular targets and the subsequent development of novel anticancer agents are opening new possibilities for drug combination therapy as anticancer treatment. Polymer-drug conjugates are well established for the delivery of a single therapeutic agent, but only in very recent years their use has been extended to the delivery of multi-agent therapy. These early studies revealed the therapeutic potential of this application but raised new challenges (namely, drug loading and drugs ratio, characterisation, and development of suitable carriers) that need to be addressed for a successful optimisation of the system towards clinical applications.
Collapse
|
34
|
Chang CC, Lerman OZ, Thanik VD, Scharf CL, Greives MR, Schneider RJ, Formenti SC, Saadeh PB, Warren SM, Levine JP. Dose-dependent effect of radiation on angiogenic and angiostatic CXC chemokine expression in human endothelial cells. Cytokine 2009; 48:295-302. [PMID: 19782578 DOI: 10.1016/j.cyto.2009.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 06/25/2009] [Accepted: 08/25/2009] [Indexed: 12/17/2022]
Abstract
Blood vessel growth is regulated by angiogenic and angiostatic CXC chemokines, and radiation is a vasculogenic stimulus. We investigated the effect of radiation on endothelial cell chemokine signaling, receptor expression, and migration and apoptosis. Human umbilical vein endothelial cells were exposed to a single fraction of 0, 5, or 20 Gy of ionizing radiation (IR). All vasculogenic chemokines (CXCL1-3/5-8) increased 3-13-fold after 5 or 20 Gy IR. 20 Gy induced a marked increase (1.6-4-fold) in angiostatic CXC chemokines. CXCR4 expression increased 3.5 and 7-fold at 48 h after 5 and 20 Gy, respectively. Bone marrow progenitor cell chemotaxis was augmented by conditioned media from cells treated with 5 Gy IR. Whereas 5 Gy markedly decreased intrinsic cell apoptosis (0 Gy=16%+/-3.6 vs. 5 Gy=4.5%+/-0.3), 20 Gy increased it (21.4%+/-1.2); a reflection of pro-survival angiogenic chemokine expression. Radiation induces a dose-dependent increase in pro-angiogenic CXC chemokines and CXCR4. In contrast, angiostatic chemokines and apoptosis were induced at higher (20 Gy) radiation doses. Cell migration improved significantly following 5 Gy, but not 20 Gy IR. Collectively, these data suggest that lower doses of IR induce an angiogenic cascade while higher doses produce an angiostatic profile.
Collapse
Affiliation(s)
- Christopher C Chang
- The Institute of Reconstructive Plastic Surgery Laboratories, New York University School of Medicine, 560 First Avenue, TH-169, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Saravia M, Zapata G, Ferraiolo P, Racca L, Berra A. Anti-VEGF monoclonal antibody-induced regression of corneal neovascularization and inflammation in a rabbit model of herpetic stromal keratitis. Graefes Arch Clin Exp Ophthalmol 2009; 247:1409-16. [PMID: 19655160 DOI: 10.1007/s00417-009-1101-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 04/06/2009] [Accepted: 04/18/2009] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND To determine the efficacy of bevacizumab (Avastin), an anti-VEGF monoclonal antibody, administrated via subconjunctival injection as a corneal anti-angiogenic treatment. METHODS Right corneas of rabbits were infected with herpes simplex virus type 1, KOS strain. On day 13 post-infection (p.i.), animals were treated subconjunctivally (sc) with a single 10-microl dose (25 microg/microl) of bevacizumab (group A) or with the same volume of an isotype monoclonal antibody, as negative control (group B). All animals were observed clinically on days 2, 5, 7, 14, 21, and 28 p.i., and two corneas each day were obtained for histological assessment and viral titration. RESULTS Viral replication was observed no longer than 5 days after infection. By day 7 a dense neutrophil invasion of the cornea was detected, which significantly increased while herpetic stromal keratitis progressed in severity. Positive outcomes observed following the treatment with bevacizumab, compared to control, included: (1) Total involution of neovascularization, (2) reduction in disease severity, (3) improved corneal translucency, (4) absence of scarring, (5) preservation of corneal thickness, (6) no neutrophil infiltration of the cornea. CONCLUSIONS Subconjunctival administration of bevacizumab induced involution of new vessels, abolished inflammatory response, and resulted in return of corneal function. Furthermore, bevacizumab is a novel approach for the treatment of herpetic stromal keratitis.
Collapse
Affiliation(s)
- Mario Saravia
- Laboratory of Investigation in Ophthalmology, Department of Pathology, School of Medicine, University of Buenos Aires, J.E. Uriburu 950, Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
36
|
Gutierrez ME, Kummar S, Giaccone G. Next generation oncology drug development: opportunities and challenges. Nat Rev Clin Oncol 2009; 6:259-65. [PMID: 19390552 DOI: 10.1038/nrclinonc.2009.38] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The optimal development of novel molecularly targeted agents for the treatment of cancer requires a re-evaluation of the current drug development paradigm. Selection of patients, optimal biologic dose versus maximum tolerated dose, definition of response and clinical benefit and trial designs that address these considerations are the focus of debate in the field of early cancer therapeutics. We present a review of the opportunities and challenges facing drug development in oncology through the phases of clinical development starting with first-in-human trials.
Collapse
|
37
|
Shin SJ, Jeung HC, Ahn JB, Rha SY, Roh JK, Park KS, Kim DH, Kim C, Chung HC. A phase I pharmacokinetic and pharmacodynamic study of CKD-732, an antiangiogenic agent, in patients with refractory solid cancer. Invest New Drugs 2009; 28:650-8. [PMID: 19585083 DOI: 10.1007/s10637-009-9287-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 06/18/2009] [Indexed: 11/29/2022]
Abstract
We conducted a phase I trial of the antiangiogenic agent 6-O-(4-dimethylaminoethoxy) cinnamoyl fumagillol hemioxalate (CKD-732). Our aims were to determine the maximum tolerated dose (MTD), pharmacokinetics (PK), and safety profiles as well as identify the biologically active dose (BAD) from ex vivo pharmacodynamics (PD) and biomarkers of CKD-732. Using a dose escalation schedule, 19 patients with refractory solid tumors were enrolled at dose levels of CKD-732 ranging from 1 to 15 mg/m(2) given twice weekly for 2 weeks followed by a 1-week rest. No treatment-related deaths occurred in this study. Confusion and insomnia were dose-limiting toxicities (DLTs), and MTD was 15 mg/m(2). The area under the concentration-time curve (AUC) and maximum concentration (Cmax) increased dose dependently with increasing doses. The BAD was 5 mg/m(2) according to ex vivo PD. A decrement in soluble vascular endothelial growth factor receptor-3 (sVEGF-3) level was correlated with a reduction in tumor size (r = 0.54, P = 0.045). The results from this study showed an MTD of 15 mg/m(2) and a BAD of 5 mg/m(2).
Collapse
Affiliation(s)
- Sang Joon Shin
- Department of Internal Medicine, Cancer Metastasis Research Center, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Falanga A. The Incidence and Risk of Venous Thromboembolism Associated With Cancer and Nonsurgical Cancer Treatment. Cancer Invest 2009; 27:105-15. [DOI: 10.1080/07357900802563028] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Mathew J, Asgeirsson K, Cheung K, Chan S, Dahda A, Robertson J. Neoadjuvant chemotherapy for locally advanced breast cancer: A review of the literature and future directions. Eur J Surg Oncol 2009; 35:113-22. [DOI: 10.1016/j.ejso.2008.03.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Accepted: 03/28/2008] [Indexed: 01/08/2023] Open
|
40
|
Tamaki K, Moriya T, Sato Y, Ishida T, Maruo Y, Yoshinaga K, Ohuchi N, Sasano H. Vasohibin-1 in human breast carcinoma: a potential negative feedback regulator of angiogenesis. Cancer Sci 2009; 100:88-94. [PMID: 19037993 PMCID: PMC11159213 DOI: 10.1111/j.1349-7006.2008.01015.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 09/21/2008] [Accepted: 09/22/2008] [Indexed: 11/28/2022] Open
Abstract
Vasohibin-1 is a recently identified negative feedback inhibitor or suppressor of angiogenesis induced by vascular endothelial growth factor (VEGF)-A. The status of vasohibin-1 in human breast carcinoma has not been examined. We examined 151 breast specimens including 98 cases of invasive ductal carcinoma (IDC), 12 of ductal carcinoma in situ (DCIS), 16 of fibroadenoma (FA), six of inflammatory lesion, nine of fibrocystic change and seven of non-pathological breast tissue. We immunolocalized vasohibin-1 and compared its immunoreactivity to that of VEGF-A, basic fibroblastic growth factor (bFGF), VEGF receptor 2 (Flk-1), CD31, CD34 and Ki-67/MIB-1. The correlation of vasohibin-1 immunoreactivity with overall survival (OS), and disease-free survival (DFS) of the patients with breast carcinoma was also evaluated. In addition, we evaluated Ki-67 and CD31, and Ki-67 and vasohibin-1 double-immunostaining for further characterization of neovascularization. Vasohibin-1 was detected in endothelial cells of human breast and its immunodensity was significantly higher in IDC and inflammatory lesions than the other types (P<0.001). In addition, a significant positive correlation was detected between vasohibin-1 and VEGF-A, bFGF or Flk-1 (P<0.001). There was also positive associations between vasohibin-1 and OS (P=0.004) and between vasohibin-1 and DFS (P
Collapse
Affiliation(s)
- Kentaro Tamaki
- Department of Surgical Oncology, Tohoku University School of Medicine, and Department of Pathology, Tohoku University Hospital, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
A survey of current literature on the topic of epistaxis revealed only a circumscript number of publications with a high methodologic value. The analysis of these publications showed that there is a controversy on the necessity of routine coagulation tests in epistaxis patients. These tests should only be performed in cases with clinical evidence of a coagulation disorder. Also, there is an ongoing controversy on the value of local cooling with ice or cold packs. Nasal creams and decongestive nose drops have been found to be effective in uncomplicated epistaxis. Rhinoscopically and endoscopically targeted coagulation of bleeding vessels and nasal packing are recommended treatment options. There is a debate on discontinuation of anticoagulant therapy, if INR is within normal limits in Cumadin patients. Intractable epistaxis requires a broad armamentarium of different diagnostic and therapeutic options. Recurrent epistaxis in hereditary syndromes remains to be a challenge, although some advances have been made in diagnosis and symptomatic treatment. Some new medical drugs, as Viagra or Cialis may have nosebleeds as side-effects.
Collapse
Affiliation(s)
- B J Folz
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Karl Hansen Klinik, Antoniusstrasse 19, 33175 Bad Lippspringe, Deutschland.
| | | | | |
Collapse
|
42
|
Descourt R, Jezequel P, Couturaud F, Leroyer C, Girard P. [Venous thromboembolism and cancer]. REVUE DE PNEUMOLOGIE CLINIQUE 2008; 64:282-289. [PMID: 19084207 DOI: 10.1016/j.pneumo.2008.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Accepted: 09/19/2008] [Indexed: 05/27/2023]
Abstract
Cancer and venous thromboembolism (VTE), VTE and cancer: there is a close bond between these two diseases. On the one hand, a cancer patient runs a high risk of developing VTE. Certain cancer-specific factors, such as its metastatic nature increase this risk. The means involved in the care of cancer (insertion of a venous catheter, chemotherapy, etc.) also increase the probability of a thromboembolism. On the other hand, VTE, especially if it is idiopathic, may be the harbinger of a neoplasm. The present paper involves the dual nature of this relationship, first dealing with several points specific to the occurrence of VTE in a cancer patient, before dealing with the specific care in a curative and prophylactic situation. VTE is then considered as a clinical manifestation prior to a cancer. Several characteristics evoking an underlying neoplasm are known. However, the benefits of the screening for cancer when confronted with an episode of VTE remains to be debated.
Collapse
Affiliation(s)
- R Descourt
- Groupe d'étude de la thrombose de Bretagne occidentale, EA 3878, CHU de la Cavale-Blanche, boulevard Tanguy-Prigent, 29609 Brest cedex, France
| | | | | | | | | |
Collapse
|
43
|
Pathak AP, Hochfeld WE, Goodman SL, Pepper MS. Circulating and imaging markers for angiogenesis. Angiogenesis 2008; 11:321-35. [PMID: 18925424 DOI: 10.1007/s10456-008-9119-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Accepted: 09/30/2008] [Indexed: 12/25/2022]
Abstract
Abundant preclinical and indirect clinical data have for several decades convincingly supported the notion that anti-angiogenesis is an effective strategy for the inhibition of tumor growth. The recent success achieved in patients with metastatic colon carcinoma using a neutralizing antibody directed against vascular endothelial growth factor (VEGF) has translated preclinical optimism into a clinical reality.With this transformation in the field of angiogenesis has come a need for reliable surrogate markers. A surrogate marker by definition serves as a substitute for the underlying process in question, and in the case of angiogenesis, microvessel density (usually in so-called "hot-spots") has until now been the most widely used parameter. However, this parameter is more akin to a static "snap-shot" and does not lend itself either to the dynamic in situ assessment of the status of the tumor microvasculature or to the molecular factors that regulate its growth and involution. This has led to an acute need for developing circulating and imaging markers of angiogenesis that can be monitored in vivo at repeated intervals in large number of patients with a variety of tumors in a non-invasive manner. Such markers of angiogenesis are the subject of this review.
Collapse
Affiliation(s)
- Arvind P Pathak
- JHU ICMIC Program, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
44
|
Schlotter CM, Vogt U, Allgayer H, Brandt B. Molecular targeted therapies for breast cancer treatment. Breast Cancer Res 2008; 10:211. [PMID: 18671839 PMCID: PMC2575526 DOI: 10.1186/bcr2112] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Targeting the oestrogen receptor, HER2 (human epidermal growth factor receptor 2) and vascular endothelial growth factor has markedly improved breast cancer therapy. New targeted therapeutic approaches to induction of apoptosis or inhibition of anti-apoptosis, cell cycle progression, signal transduction and angiogenesis are described. The molecular pathways and their inhibitory or repair mechanisms are discussed in the preclinical and clinical settings.
Collapse
Affiliation(s)
- Claus M Schlotter
- Department of Obstetrics and Gynecology, Breast Centre Klinikum Lüdenscheid, Academic Teaching Hospital of the University Bonn, Luedenscheid, Paulmannshoeher Str, 14 - 58515 Luedenscheid Germany
| | | | | | | |
Collapse
|
45
|
Linkins LA. Management of venous thromboembolism in patients with cancer: role of dalteparin. Vasc Health Risk Manag 2008; 4:279-87. [PMID: 18561503 PMCID: PMC2496976 DOI: 10.2147/vhrm.s2132] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cancer is a major risk factor for the development of venous thromboembolism (VTE). Conventional anticoagulant therapy with a vitamin K antagonist is more problematic in cancer patients due to an increased risk of recurrent VTE, and an increased risk of anticoagulant-related bleeding. In recent years, there has been a shift toward treating cancer patients with VTE with extended duration dalteparin. Dalteparin, a low-molecular-weight heparin, has been shown to be more effective, and as safe as conventional anticoagulant therapy, in cancer patients with VTE. This paper will (a) review the relationship between cancer and VTE, and (b) provide an overview of the role of dalteparin in the management of VTE in patients with cancer.
Collapse
Affiliation(s)
- Lori-Ann Linkins
- Department of Hematology and Thrombosis, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
46
|
Bartsch R, Steger GG. Adjuvant chemotherapy in breast cancer. MEMO - MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2008. [DOI: 10.1007/s12254-008-0019-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
47
|
[Thromboembolic risk associated with use of angiogenesis inhibitors used for the treatment of cancers]. ACTA ACUST UNITED AC 2008; 56:195-204. [PMID: 18450388 DOI: 10.1016/j.patbio.2008.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 02/28/2008] [Indexed: 01/16/2023]
Abstract
Among antiangiogenic agents, thalidomide is not the most potent nor the most specific even so when venous thromboembolic events have been reported with the prescription of thalidomide in multiple myeloma. This side effect has been related to the antiangiogenic effect of this immunomodulator. In keeping with this observation venous thromboembolic events have been reported in other indications of thalidomide and with thalidomide analogues (Lenalidomide and Actimid). The thrombotic side effects are mostly venous but arterial thrombotic events are also observed with the use of these molecules. With the other and more specific antiagiogenic agents an increase in thrombotic events are also observed. This increase was not immediately evident since the situation in which they are prescribed (metastatic cancers) are already characterized by a high rate of thrombotic events. The prothrombotic effect of antiangiogenic agents are probably linked to an effect on endothelium (decrease of antithrombotic activities and stimulation of a prothrombotuic state). The other sides effects of antiangiogenic agents (hemorrhages, hypertension, proteinuria, microangiotpahia, delay in scaring) are also probably related to endothelial effects. The prothrombotic effect of antoangiogenic agents appears as potentiating the prothrombotic conditions of the disease (myeloma, cancer) and the prothombotic effects of the associated treatments (chemotherapy, high dose corticosteroids, erythropoietin). The increased thrombotic risk linked to prescription of antiangiogenic agents and specially of thalidomide and analogues for multiple myeloma is such that it is recommended to associate a preventive antithrombotic treatment. Some efficacy has been reported with the use of aspirin, oral anticoagulant or low molecular weight heparin. No head to head comparative trial do not allow to prefer one strategy. From published data full dose oral anticoagulants appear to confer the highest hemorrhagic risk and perhaps low molecular weight heparin the best benefit-risk ratio.
Collapse
|
48
|
Fraile Gil S, Hidalgo Correas FJ, Lara Alvarez MA, Garrote Martínez FJ. [Bevacizumab-induced serious heart failure in a patient treated with anthracyclines]. FARMACIA HOSPITALARIA 2008; 31:256-7. [PMID: 18052624 DOI: 10.1016/s1130-6343(07)75385-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
49
|
Abstract
Venous thromboembolism is a common and life-threatening complication in patients with advanced cancer. Chemotherapy has been identified as a risk factor for venous thromboembolism in cancer patients. The annual incidence of venous thromboembolism in cancer patients receiving chemotherapy is estimated to be about 10%. This risk increases up to 15-20% depending on type and combination of anticancer drugs. Hormonal and supportive therapies are also associated with increased risk for thrombembolic complications. Emerging data support the hypothesis that the occurrence of venous thrombembolic events in cancer patients is associated with a poor prognosis. The benefit of antithrombotic prophylaxis for venous thrombembolism in cancer patients receiving chemotherapy remains to be established. A randomized, placebo controlled, double blind trial (the PROTECHT study) designed to evaluate the efficacy and safety of the low-molecular-weight heparin nadroparin in the prophylaxis of venous and arterial thrombembolic events during chemotherapy has been recently completed and the results are currently analysed.
Collapse
Affiliation(s)
- Giancarlo Agnelli
- Division of Internal and Cardiovascular Medicine, Stroke Unit, University of Perugia, Perugia, Italy.
| | | |
Collapse
|
50
|
|