51
|
Ostróżka-Cieślik A, Dolińska B. The Role of Hormones and Trophic Factors as Components of Preservation Solutions in Protection of Renal Function before Transplantation: A Review of the Literature. Molecules 2020; 25:E2185. [PMID: 32392782 PMCID: PMC7248710 DOI: 10.3390/molecules25092185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Transplantation is currently a routine method for treating end-stage organ failure. In recent years, there has been some progress in the development of an optimal composition of organ preservation solutions, improving the vital functions of the organ and allowing to extend its storage period until implantation into the recipient. Optimizations are mostly based on commercial solutions, routinely used to store grafts intended for transplantation. The paper reviews hormones with a potential nephroprotective effect, which were used to modify the composition of renal perfusion and preservation solutions. Their effectiveness as ingredients of preservation solutions was analysed based on a literature review. Hormones and trophic factors are innovative preservation solution supplements. They have a pleiotropic effect and affect normal renal function. The expression of receptors for melatonin, prolactin, thyrotropin, corticotropin, prostaglandin E1 and trophic factors was confirmed in the kidneys, which suggests that they are a promising therapeutic target for renal IR (ischemia-reperfusion) injury. They can have anti-inflammatory, antioxidant and anti-apoptotic effects, limiting IR injury.
Collapse
Affiliation(s)
- Aneta Ostróżka-Cieślik
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
| | - Barbara Dolińska
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
- “Biochefa” Pharmaceutical Research and Production Plant, Kasztanowa 3, 41-200 Sosnowiec, Poland
| |
Collapse
|
52
|
Quddus MR, Hansen K, Tantravahi U, Sung CJ. Human epidermal growth factor receptor 2 (Her2) testing for uterine serous carcinoma: Report of scenarios of unusual overexpression. Gynecol Oncol Rep 2020; 32:100561. [PMID: 32258332 PMCID: PMC7103767 DOI: 10.1016/j.gore.2020.100561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 01/06/2023] Open
Abstract
The human epidermal growth factor receptor 2 (Her2) is tested in many human cancers, including breast, bladder, pancreatic, ovarian and stomach. The American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) have issued Clinical Practice Guidelines for reporting Her2 results for breast carcinomas (Wolf et al., 2018). For the last 1-2 years Her2/neu is tested in endometrial serous carcinoma, especially in recurrent tumors or non-responsive tumors as an option for additional treatment. College of American Pathologists (CAP) offers a template for prognostic marker reporting results for specimens with endometrial carcinomas (Fitzgibbons et al., 2019). Her2/neu testing by immunohistochemistry (IHC) mandates rigorous fixation time control, e.g., fixation time should fall within 6-72 h (Recommendations for Her2 Testing in Breast Cancer, 2013). For that reason, in breast cancers, Her2/neu testing is done on initial core biopsy specimens. The test is however, repeated on excision specimen in high grade tumors where Her2/neu expression was initially negative on core biopsies. For endometrial serous carcinoma no guidelines have been set or proposed as of yet. The Gynecologic Oncologists request this test because of proven benefit of adding Trastuzumab (Fader et al., 2018) and that is why it is important to documenting the findings in this report in the literature so that an informed request can be made by the treating oncologist when multiple tissue samples from the same patient are available for testing. Similarly pathologists also can decide which would be the best sample to test when no instruction is received. We report here three separate scenarios of uterine serous carcinomas in which the Her2/neu expressions were unique enough to justify documentation and therefore have implications for determining which specimen is ideal for the Her2 overexpression testing and likely to have highest possibility in identifying the Her2/neu overexpressed clone in the tumor which would expand the therapeutic options for the patients.
Collapse
Affiliation(s)
- M Ruhul Quddus
- Department of Pathology, Women Infants Hospital/Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Katrine Hansen
- Department of Pathology, Women Infants Hospital/Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Umadevi Tantravahi
- Department of Pathology, Women Infants Hospital/Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - C James Sung
- Department of Pathology, Women Infants Hospital/Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
53
|
Erickson BK, Zeybek B, Santin AD, Fader AN. Targeting human epidermal growth factor receptor 2 (HER2) in gynecologic malignancies. Curr Opin Obstet Gynecol 2020; 32:57-64. [PMID: 31833974 PMCID: PMC7307693 DOI: 10.1097/gco.0000000000000599] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe the role of the human epidermal growth factor receptor 2 (HER2) as a biomarker and potential target in gynecologic malignancies and to describe contemporary updates in the use of anti-HER2 treatments for these cancers. RECENT FINDINGS Approximately 25-30% of all patients with uterine serous carcinoma overexpress tumoral HER2. The anti-HER2 antibody trastuzumab represents an effective, targeted therapy with significant efficacy in the treatment of HER2-positive breast and gastric cancer. Recently, trastuzumab efficacy has also been demonstrated in a randomized controlled trial of women with advanced or recurrent uterine serous carcinoma. Additionally, trastuzumab may be effective in women with HER2-positive uterine carcinosarcoma. The role of anti-HER2 therapy is unclear in women with other gynecologic malignancies but is being evaluated. SUMMARY HER2 amplification/overexpression is an effective therapeutic target in select gynecologic malignancies, and especially in the rare endometrial cancer subtype, uterine serous carcinoma. As anti-HER2-targeted therapies become increasingly available, more treatment options may become available for women with HER2-positive disease.
Collapse
Affiliation(s)
- Britt K. Erickson
- Division of Gynecologic Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Burak Zeybek
- Division of Gynecologic Oncology, Yale University, New Haven, Connecticut
| | | | - Amanda N. Fader
- Division of Gynecologic Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
54
|
Jhaveri KL, Wang XV, Makker V, Luoh SW, Mitchell EP, Zwiebel JA, Sharon E, Gray RJ, Li S, McShane LM, Rubinstein LV, Patton D, Williams PM, Hamilton SR, Conley BA, Arteaga CL, Harris LN, O'Dwyer PJ, Chen AP, Flaherty KT. Ado-trastuzumab emtansine (T-DM1) in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction (GEJ) adenocarcinomas: results from the NCI-MATCH trial (EAY131) subprotocol Q. Ann Oncol 2019; 30:1821-1830. [PMID: 31504139 PMCID: PMC6927318 DOI: 10.1093/annonc/mdz291] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) is a national precision medicine study incorporating centralized genomic testing to direct refractory cancer patients to molecularly targeted treatment subprotocols. This treatment subprotocol was designed to screen for potential signals of efficacy of ado-trastuzumab emtansine (T-DM1) in HER2-amplified histologies other than breast and gastroesophageal tumors. METHODS Eligible patients had HER2 amplification at a copy number (CN) >7 based on targeted next-generation sequencing (NGS) with a custom Oncomine AmpliSeq™ (ThermoFisher Scientific) panel. Patients with prior trastuzumab, pertuzumab or T-DM1 treatment were excluded. Patients received T-DM1 at 3.6 mg/kg i.v. every 3 weeks until toxicity or disease progression. Tumor assessments occurred every three cycles. The primary end point was centrally assessed objective response rate (ORR). Exploratory end points included correlating response with HER2 CN by NGS. The impact of co-occurring genomic alterations and PTEN loss by immunohistochemistry were also assessed. RESULTS Thirty-eight patients were enrolled and 36 included in efficacy analysis. Median prior therapies in the metastatic setting was 3 (range 0-9; unknown in one patient). Median HER2 CN was 17 (range 7-139). Partial responses were observed in two (5.6%) patients: one mucoepidermoid carcinoma of parotid gland and one parotid gland squamous cell cancer. Seventeen patients (47%) had stable disease including 8/10 (80%) with ovarian and uterine carcinomas, with median duration of 4.6 months. The 6-month progression-free survival rate was 23.6% [90% confidence interval 14.2% to 39.2%]. Common toxicities included fatigue, anemia, fever and thrombocytopenia with no new safety signals. There was a trend for tumor shrinkage with higher levels of gene CN as determined by the NGS assay. CONCLUSION T-DM1 was well tolerated. While this subprotocol did not meet the primary end point for ORR in this heavily pre-treated diverse patient population, clinical activity was seen in salivary gland tumors warranting further study in this tumor type in dedicated trials.
Collapse
Affiliation(s)
- K L Jhaveri
- Department of Medicine, Memorial Sloan-Kettering Center, New York.
| | - X V Wang
- Biostatistics, E-A Biostatistical Center, Boston
| | - V Makker
- Gynecologic Medical Oncology Service, Memorial Sloan-Kettering Cancer Center, New York
| | - S-W Luoh
- Knight Cancer Institute, Oregon Health Science University, Portland
| | - E P Mitchell
- Medical Oncology, Thomas Jefferson University, Philadelphia
| | - J A Zwiebel
- Investigational Drug Branch, Division of Cancer Treatment and Diagnosis
| | - E Sharon
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda
| | - R J Gray
- Department of Biostatistics, Dana Farber Cancer Institutes, Boston
| | - S Li
- Department of Biostatistics, Dana Farber Cancer Institutes, Boston
| | - L M McShane
- Biometric Research Branch, National Cancer Institute, Bethesda
| | - L V Rubinstein
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, Bethesda
| | - D Patton
- Center for Biomedical, Informatics & Information Technology, National Cancer Institute, Bethesda
| | - P M Williams
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick
| | - S R Hamilton
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - B A Conley
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda
| | - C L Arteaga
- Department of Internal Medicine, University of Texas Southwestern, Dallas
| | - L N Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda
| | | | - A P Chen
- CTEP, National Cancer Institute, Bethesda
| | - K T Flaherty
- Cancer Center, Massachusetts General Hospital, Boston, USA
| |
Collapse
|
55
|
Dumbrava EEI, Balaji K, Raghav K, Hess K, Javle M, Blum-Murphy M, Ajani J, Kopetz S, Broaddus R, Routbort M, Demirhan M, Zheng X, Pant S, Tsimberidou AM, Subbiah V, Hong DS, Rodon J, Shaw KM, Piha-Paul SA, Meric-Bernstam F. Targeting ERBB2 ( HER2) Amplification Identified by Next-Generation Sequencing in Patients With Advanced or Metastatic Solid Tumors Beyond Conventional Indications. JCO Precis Oncol 2019; 3:PO.18.00345. [PMID: 32923865 PMCID: PMC7446516 DOI: 10.1200/po.18.00345] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2019] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2) is an effective therapeutic target in breast and gastric and gastroesophageal junction cancers. However, less is known about the prevalence of ERBB2 (HER2) amplification and the efficacy of HER2-targeted treatment in other tumors. PATIENTS AND METHODS We assessed HER2 amplification status among 5,002 patients with advanced disease (excluding breast cancer) who underwent next-generation sequencing. We evaluated the clinical benefit of HER2-targeted therapy by measuring the time-dependent overall survival (OS) from the genomic testing results, progression-free survival (PFS), and PFS during HER2-targeted therapy (PFS2) compared with PFS during prior therapy (PFS1). RESULTS Overall, 122 patients (2.4%) had HER2 amplification, including patients with endometrial (5.3%), bladder (5.2%), biliary or gallbladder (4.9%), salivary (4.7%), and colorectal cancer (3.6%). Forty patients (38%) with nongastric, nongastroesophageal junction, or nonesophageal cancers received at least one line of HER2-targeted therapy. Patients receiving HER2-targeted therapy had a median OS of 18.6 months, compared with 10.9 months for patients who did not receive HER2-targeted therapy (P = .070). On multivariable analysis, HER2-targeted therapy was significantly associated with increased OS (hazard ratio, 0.5; 95% CI, 0.27 to 0.93; P = .029), regardless of sex, age, or number of prior lines of treatment. The PFS2-to-PFS1 ratio was 1.3 or greater in 21 (57%) of 37 patients who received HER2-targeted therapy not in the first line of systemic treatment, and the median PFS2 and PFS1 times were 24 and 13 weeks, respectively (P < .001). CONCLUSION HER2 amplifications using next-generation sequencing can be identified in a variety of tumor types. HER2-targeted therapy may confer clinical benefit in tumor types other than those for which HER2 inhibitors are approved.
Collapse
Affiliation(s)
| | - Kavitha Balaji
- The University of Texas MD Anderson Cancer Center, Houston, TX
- Lexicon Pharmaceuticals, Houston, TX
| | - Kanwal Raghav
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kenneth Hess
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Milind Javle
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Jaffer Ajani
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Mark Routbort
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mehmet Demirhan
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiaofeng Zheng
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shubham Pant
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David S. Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jordi Rodon
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kenna M. Shaw
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
56
|
HER2 somatic mutation analysis in breast cancer: correlation with clinicopathological features. Hum Pathol 2019; 92:32-38. [DOI: 10.1016/j.humpath.2019.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/13/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
|
57
|
Sonkar K, Ayyappan V, Tressler CM, Adelaja O, Cai R, Cheng M, Glunde K. Focus on the glycerophosphocholine pathway in choline phospholipid metabolism of cancer. NMR IN BIOMEDICINE 2019; 32:e4112. [PMID: 31184789 PMCID: PMC6803034 DOI: 10.1002/nbm.4112] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/16/2019] [Accepted: 04/20/2019] [Indexed: 05/02/2023]
Abstract
Activated choline metabolism is a hallmark of carcinogenesis and tumor progression, which leads to elevated levels of phosphocholine and glycerophosphocholine in all types of cancer tested so far. Magnetic resonance spectroscopy applications have played a key role in detecting these elevated choline phospholipid metabolites. To date, the majority of cancer-related studies have focused on phosphocholine and the Kennedy pathway, which constitutes the biosynthesis pathway for membrane phosphatidylcholine. Fewer and more recent studies have reported on the importance of glycerophosphocholine in cancer. In this review article, we summarize the recent literature on glycerophosphocholine metabolism with respect to its cancer biology and its detection by magnetic resonance spectroscopy applications.
Collapse
Affiliation(s)
- Kanchan Sonkar
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vinay Ayyappan
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Caitlin M. Tressler
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Oluwatobi Adelaja
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ruoqing Cai
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Menglin Cheng
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristine Glunde
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
58
|
Yu S, Zhang J, Yan Y, Yao X, Fang L, Xiong H, Liu Y, Chu Q, Zhou P, Wu K. A novel asymmetrical anti-HER2/CD3 bispecific antibody exhibits potent cytotoxicity for HER2-positive tumor cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:355. [PMID: 31412896 PMCID: PMC6694677 DOI: 10.1186/s13046-019-1354-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/01/2019] [Indexed: 01/05/2023]
Abstract
Background Human epidermal growth factor receptor 2 (HER2) is overexpressed in multiple cancers, which is associated with poor prognosis. Herceptin and other agents targeting HER2 have potent antitumor efficacy in patients with HER2-positive cancers. However, the development of drug resistance adversely impacts the efficacy of these treatments. It is therefore urgent to develop new HER2-targeted therapies. Bispecific antibodies (BsAbs) could guide immune cells toward tumor cells, and produced remarkable effects in some cancers. Methods A BsAb named M802 that targets HER2 and CD3 was produced by introducing a salt bridge and knobs-into-holes (KIHs) packing into the structure. Flow cytometry was performed to determine its binding activity and cytotoxicity. CCK-8, Annexin V/PI staining, western blotting, and ELISA were utilized to study its effect on cell proliferation, apoptosis, the signaling pathways of tumor cells, and the secretion of cytokines by immune cells. Subcutaneous tumor mouse models were used to analyze the in vivo antitumor effects of M802. Results We generated a new format of BsAb, M802, consisting of a monovalent unit against HER2 and a single chain unit against CD3. Our in vitro and in vivo experiments indicated that M802 recruited CD3-positive immune cells and was more cytotoxic than Herceptin in cells with high expression of HER2, low expression of HER2, and Herceptin resistance. Although M802 showed weaker effects than Herceptin on the PI3K/AKT and MAPK pathways, it was more cytotoxic due to its specific recognition of HER2 and its ability to recruit effector cells via its anti-CD3 moiety. Conclusions Our results indicated that M802 exhibited potent antitumor efficacy in vitro and in vivo. M802 retained the function of Herceptin in antitumor signaling pathways, and also recruited CD3-positive immune cells to eliminate HER2-positive tumor cells. Therefore, M802 might be a promising HER2 targeted agent. Electronic supplementary material The online version of this article (10.1186/s13046-019-1354-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Jing Zhang
- Wuhan YZY Biopharma Co., Ltd, Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, People's Republic of China
| | - Yongxiang Yan
- Wuhan YZY Biopharma Co., Ltd, Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, People's Republic of China
| | - Xudong Yao
- Department of Orthopedics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lijuan Fang
- Wuhan YZY Biopharma Co., Ltd, Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, People's Republic of China
| | - Hui Xiong
- Wuhan YZY Biopharma Co., Ltd, Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, People's Republic of China
| | - Yang Liu
- Wuhan YZY Biopharma Co., Ltd, Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Pengfei Zhou
- Wuhan YZY Biopharma Co., Ltd, Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, People's Republic of China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
59
|
Muhammad IF, Borné Y, Bao X, Melander O, Orho-Melander M, Nilsson PM, Nilsson J, Engström G. Circulating HER2/ErbB2 Levels Are Associated With Increased Incidence of Diabetes: A Population-Based Cohort Study. Diabetes Care 2019; 42:1582-1588. [PMID: 31201260 DOI: 10.2337/dc18-2556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 05/23/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE HER2/ErbB2 is a member of the epidermal growth factor receptor family. It is widely used as a tumor marker, but it also has recently been associated with insulin resistance. Both ErbB2 and diabetes have been associated with cancer; however, the relationship between ErbB2 and diabetes has not been well explored. The aim of this population-based cohort study was to assess the association between plasma ErbB2 and incidence of diabetes. RESEARCH DESIGN AND METHODS The study population included participants from the Malmö Diet and Cancer-Cardiovascular Cohort (age range 46-68 years). After excluding participants with a history of diabetes and those missing data for ErbB2 and other covariates, the final study population consisted of 4,220 individuals. Incidence of diabetes was followed through linkages to local and national registers. Cox proportional hazards regression was used to assess the incidence of diabetes in relation to quartiles of ErbB2, adjusted for potential confounders. RESULTS Plasma ErbB2 was significantly and positively associated with glucose, insulin, and HbA1c after being adjusted for potential confounding factors. During a mean ± SD follow-up period of 20.20 ± 5.90 years, 615 participants (14.6%) were diagnosed with new-onset diabetes. Individuals with high levels of ErbB2 had a significantly higher risk of diabetes than those with low levels of ErbB2. The multivariable-adjusted hazard ratio was 1.31 (95% CI 1.03-1.66; P < 0.05) for the highest versus the lowest quartile of ErbB2 and was 1.15 (95% CI 1.05-1.25; P < 0.05) per 1-SD increase in ErbB2. CONCLUSIONS Elevated levels of ErbB2 are associated with increased incidence of diabetes.
Collapse
Affiliation(s)
| | - Yan Borné
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Xue Bao
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
60
|
Shepherd-Littlejohn AL, Hanft WJ, Kennedy VA, Alvarez EA. Afatinib use in recurrent epithelial ovarian carcinoma. Gynecol Oncol Rep 2019; 29:70-72. [PMID: 31360743 PMCID: PMC6639588 DOI: 10.1016/j.gore.2019.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/03/2022] Open
Abstract
Genomic tumor testing is an important tool in guiding treatment for gynecologic malignancies. Targetable mutations may lead to new therapies in gynecologic cancer treatment. Her2/neu mutations in serous ovarian carcinomas can be targeted with ERBB2 inhibitors. Afatinib shows promising response rates in lung cancers carrying Her2/neu mutations. Afatinib may be effective in serous ovarian tumors exhibiting Her2/neu or ERBB2 mutations.
Collapse
Affiliation(s)
| | - Wyatt J Hanft
- School of Public Health, UC Berkeley, Berkeley, CA,USA.,School of Medicine, UC Davis, Sacramento, CA,USA
| | | | - Edwin A Alvarez
- Department of Gynecologic Oncology, UC San Francisco, San Francisco, CA,USA
| |
Collapse
|
61
|
Vengoji R, Macha MA, Nimmakayala RK, Rachagani S, Siddiqui JA, Mallya K, Gorantla S, Jain M, Ponnusamy MP, Batra SK, Shonka N. Afatinib and Temozolomide combination inhibits tumorigenesis by targeting EGFRvIII-cMet signaling in glioblastoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:266. [PMID: 31215502 PMCID: PMC6582495 DOI: 10.1186/s13046-019-1264-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022]
Abstract
Background Glioblastoma (GBM) is an aggressive brain tumor with universal recurrence and poor prognosis. The recurrence is largely driven by chemoradiation resistant cancer stem cells (CSCs). Epidermal growth factor receptor (EGFR) and its mutant EGFRvIII are amplified in ~ 60% and ~ 30% of GBM patients, respectively; however, therapies targeting EGFR have failed to improve disease outcome. EGFRvIII-mediated cross-activation of tyrosine kinase receptor, cMET, regulates GBM CSC maintenance and promote tumor recurrence. Here, we evaluated the efficacy of pan-EGFR inhibitor afatinib and Temozolomide (TMZ) combination on GBM in vitro and in vivo. Methods We analyzed the effect of afatinib and temozolomide (TMZ) combination on GBM cells U87MG and U251 engineered to express wild type (WT) EGFR, EGFRvIII or EGFRvIII dead kinase, CSCs isolated from U87 and U87EGFRvIII in vitro. The therapeutic utility of the drug combination was investigated on tumor growth and progression using intracranially injected U87EGFRvIII GBM xenografts. Results Afatinib and TMZ combination synergistically inhibited the proliferation, clonogenic survival, motility, invasion and induced senescence of GBM cells compared to monotherapy. Mechanistically, afatinib decreased U87EGFRvIII GBM cell proliferation and motility/invasion by inhibiting EGFRvIII/AKT, EGFRvIII/JAK2/STAT3, and focal adhesion kinase (FAK) signaling pathways respectively. Interestingly, afatinib specifically inhibited EGFRvIII-cMET crosstalk in CSCs, resulting in decreased expression of Nanog and Oct3/4, and in combination with TMZ significantly decreased their self-renewal property in vitro. More interestingly, afatinib and TMZ combination significantly decreased the xenograft growth and progression compared to single drug alone. Conclusion Our study demonstrated significant inhibition of GBM tumorigenicity, CSC maintenance in vitro, and delayed tumor growth and progression in vivo by combination of afatinib and TMZ. Our results warrant evaluation of this drug combination in EGFR and EGFRvIII amplified GBM patients. Electronic supplementary material The online version of this article (10.1186/s13046-019-1264-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
62
|
Nakada T, Sugihara K, Jikoh T, Abe Y, Agatsuma T. The Latest Research and Development into the Antibody-Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy. Chem Pharm Bull (Tokyo) 2019; 67:173-185. [PMID: 30827997 DOI: 10.1248/cpb.c18-00744] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A major limitation of traditional chemotherapy for cancer is dose-limiting toxicity, caused by the exposure of non-tumor cells to cytotoxic agents. Use of molecular targeted drugs, such as specific kinase inhibitors and monoclonal antibodies, is a possible solution to overcome this limitation and has achieved clinical success so far. Use of an antibody-drug conjugate (ADC) is a rational strategy for improving efficacy and reducing systemic adverse events. ADCs use antibodies selectively to deliver a potent cytotoxic agent to tumor cells, thus drastically improving the therapeutic index of chemotherapeutic agents. Lessons learned from clinical failure of early ADCs during the 1980s to 90s have recently led to improvements in ADC technology, and resulted in the approval of four novel ADCs. Nonetheless, further advances in ADC technology are still required to streamline their clinical efficacy and reduce toxicity. [fam-] Trastuzumab deruxtecan (DS-8201a) is a next-generation ADC that satisfies these requirements based on currently available evidence. DS-8201a has several innovative features; a highly potent novel payload with a high drug-to-antibody ratio, good homogeneity, a tumor-selective cleavable linker, stable linker-payload in circulation, and a short systemic half-life cytotoxic agent in vivo; the released cytotoxic payload could exert a bystander effect. With respect to its preclinical profiles, DS-8201a could provide a valuable therapy with a great potential against HER2-expressing cancers in clinical settings. In a phase I trial, DS-8201a showed acceptable safety profiles with potential therapeutic efficacy, with the wide therapeutic index.
Collapse
Affiliation(s)
- Takashi Nakada
- Research and Development Division, Daiichi Sankyo Co., Ltd
| | | | - Takahiro Jikoh
- Research and Development Division, Daiichi Sankyo Co., Ltd
| | - Yuki Abe
- Research and Development Division, Daiichi Sankyo Co., Ltd
| | | |
Collapse
|
63
|
Musselman K, Glynn S, Mosquera JM, Elemento O, Sboner A, Beltran H, Holcomb K. Identification of a therapeutic target using molecular sequencing for treatment of recurrent uterine serous adenocarcinoma. Gynecol Oncol Rep 2019; 28:54-57. [PMID: 30906838 PMCID: PMC6411491 DOI: 10.1016/j.gore.2019.01.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 12/02/2022] Open
Abstract
Uterine serous adenocarcinoma is a rare but highly malignant form of endometrial cancer, comprising over 50% of recurrences and deaths from endometrial cancer. We report a case of a 68-year old woman with recurrent uterine serous adenocarcinoma who underwent molecular testing and genetic sequencing of her tumor. She was found to have focal amplification of ERBB2 confirmed by amplification and overexpression of HER2/neu via fluorescence in situ hybridization and immunohistochemistry. Given the identification of this potential target and progression of disease, trastuzumab was added to the patient's chemotherapy regimen with ultimate complete response. 68-year old woman with recurrent uterine serous adenocarcinoma who underwent molecular testing of her tumor. Amplification of ERBB2 and overexpression of HER2/neu was confirmed. Trastuzumab was added to the patient's therapy with complete response. Targeted therapies may be advantageous in patients with limited therapeutic options.
Collapse
Affiliation(s)
- Kelsey Musselman
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, United States of America
| | - Shannon Glynn
- Department of Medicine, Weill Cornell Medicine, United States of America
| | - Juan Miguel Mosquera
- Englander Institute for Precision Medicine, New York Presbyterian-Weill Cornell Medicine, United States of America
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, United States of America
| | - Olivier Elemento
- Englander Institute for Precision Medicine, New York Presbyterian-Weill Cornell Medicine, United States of America
| | - Andrea Sboner
- Englander Institute for Precision Medicine, New York Presbyterian-Weill Cornell Medicine, United States of America
| | - Himisha Beltran
- Department of Medicine, Weill Cornell Medicine, United States of America
- Englander Institute for Precision Medicine, New York Presbyterian-Weill Cornell Medicine, United States of America
- Department of Medical Oncology, Dana Farber Cancer Institute, United States of America
| | - Kevin Holcomb
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, United States of America
- Corresponding author at: 525 East 68th Street, J-130, New York, NY 10065, United States of America.
| |
Collapse
|
64
|
Irie H, Ito K, Fujioka Y, Oguchi K, Fujioka A, Hashimoto A, Ohsawa H, Tanaka K, Funabashi K, Araki H, Kawai Y, Shimamura T, Wadhwa R, Ohkubo S, Matsuo K. TAS0728, A Covalent-binding, HER2-selective Kinase Inhibitor Shows Potent Antitumor Activity in Preclinical Models. Mol Cancer Ther 2019; 18:733-742. [PMID: 30787176 DOI: 10.1158/1535-7163.mct-18-1085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/09/2018] [Accepted: 01/22/2019] [Indexed: 11/16/2022]
Abstract
Activated HER2 is a promising therapeutic target for various cancers. Although several reports have described HER2 inhibitors in development, no covalent-binding inhibitor selective for HER2 has been reported. Here, we report a novel compound TAS0728 that covalently binds to HER2 at C805 and selectively inhibits its kinase activity. Once TAS0728 bound to HER2 kinase, the inhibitory activity was not affected by a high ATP concentration. A kinome-wide biochemical panel and cellular assays established that TAS0728 possesses high specificity for HER2 over wild-type EGFR. Cellular pharmacodynamics assays using MCF10A cells engineered to express various mutated HER2 genes revealed that TAS0728 potently inhibited the phosphorylation of mutated HER2 and wild-type HER2. Furthermore, TAS0728 exhibited robust and sustained inhibition of the phosphorylation of HER2, HER3, and downstream effectors, thereby inducing apoptosis of HER2-amplified breast cancer cells and in tumor tissues of a xenograft model. TAS0728 induced tumor regression in mouse xenograft models bearing HER2 signal-dependent tumors and exhibited a survival benefit without any evident toxicity in a peritoneal dissemination mouse model bearing HER2-driven cancer cells. Taken together, our results demonstrated that TAS0728 may offer a promising therapeutic option with improved efficacy as compared with current HER2 inhibitors for HER2-activated cancers. Assessment of TAS0728 in ongoing clinical trials is awaited (NCT03410927).
Collapse
Affiliation(s)
- Hiroki Irie
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan.
| | - Kimihiro Ito
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Yayoi Fujioka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kei Oguchi
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Akio Fujioka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Akihiro Hashimoto
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Hirokazu Ohsawa
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kenji Tanaka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kaoru Funabashi
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Hikari Araki
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Yuichi Kawai
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Tadashi Shimamura
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Renu Wadhwa
- DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), DAICENTER, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Shuichi Ohkubo
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kenichi Matsuo
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| |
Collapse
|
65
|
Xia L, Wang Y, Hu Y, Zhang C, Gu T, Wang L, Li J, Yu W, Tian Z. Human epithelial growth factor receptor 2 in human salivary carcinoma ex pleomorphic adenoma: a potential therapeutic target. Cancer Manag Res 2018; 10:6571-6579. [PMID: 30555262 PMCID: PMC6280888 DOI: 10.2147/cmar.s182652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background To inhibit human epithelial growth factor receptor 2 (HER2) in salivary carcinoma ex pleomorphic adenoma (CXPA) and investigate the effects on tumor cell proliferation, cell cycle, and apoptosis. To assess the possibility of blocking HER2 to improve the malignant biological behavior of CXPA. Materials and methods HER2 expression and amplification were examined using an immunofluorescence assay and fluorescence in situ hybridization in 2 CXPA cell lines (SM-AP1 and SM-AP4 cells). The effects on tumor cell proliferation, cell cycle, apoptosis, and HER2 downstream pathways were verified after the application of a HER2 inhibitor. Results HER2 was overexpressed and amplified in SM-AP1 and SM-AP4 cell lines. After blocking HER2, the tumor proliferation and cell cycle were significantly induced, and the apoptosis process was activated. Moreover, the downstream pathways PI3K/AKT and MAPK/ERK were significantly inhibited. Conclusion HER2 was overexpressed and amplified in CXPA cell lines and might thus play an important role in tumor development. Inhibiting HER2 may be a novel targeted therapy for poor biological behavior of CXPA.
Collapse
Affiliation(s)
- Liang Xia
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China, .,Department of Oral and Cranio-maxillofacial Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Yang Wang
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Yuhua Hu
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Chunye Zhang
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Ting Gu
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Lizhen Wang
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Jiang Li
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Wenwen Yu
- Department of Oral and Cranio-maxillofacial Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| | - Zhen Tian
- Department of Oral Pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai 200011, People's Republic of China,
| |
Collapse
|
66
|
Leal AD, Krishnamurthy A, Head L, Messersmith WA. Antibody drug conjugates under investigation in phase I and phase II clinical trials for gastrointestinal cancer. Expert Opin Investig Drugs 2018; 27:901-916. [PMID: 30359534 DOI: 10.1080/13543784.2018.1541085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Antibody drug conjugates (ADCs) represent a developing class of anticancer therapeutics which are designed to selectively deliver a cytotoxic payload to tumors, while limiting systemic toxicity to healthy tissues. There are several ADCs which are currently in various stages of clinical development for the treatment of gastrointestinal malignancies. AREAS COVERED We discuss the biologic rationale and review the clinical experience with ADCs in the treatment of gastrointestinal malignancies, summarizing the pre-clinical and phase I/II clinical trial data that have been completed or are ongoing. EXPERT OPINION While there have been significant advances in the development of ADCs since they were first introduced, several challenges remain. These challenges include (i) the selection of an ideal antigen target which is tumor specific and internalized upon binding, (ii) selection of an antibody which has high affinity for its antigen target and low immunogenicity, (iii) selection of a potent payload which is cytotoxic at sub-nanomolar concentrations, and (iv) optimal design of a linker to confer ADC stability with limited off-site toxicity. Efforts are ongoing to address these issues and innovate the ADC technology to improve the safety and efficacy of these agents.
Collapse
Affiliation(s)
- Alexis D Leal
- a Division of Medical Oncology , University of Colorado , Aurora , CO , USA
| | | | - Lia Head
- b Department of Internal Medicine , University of Colorado , Aurora , CO , USA
| | | |
Collapse
|
67
|
Fuentes I, García-Mendiola T, Sato S, Pita M, Nakamura H, Lorenzo E, Teixidor F, Marques F, Viñas C. Metallacarboranes on the Road to Anticancer Therapies: Cellular Uptake, DNA Interaction, and Biological Evaluation of Cobaltabisdicarbollide [COSAN] . Chemistry 2018; 24:17239-17254. [PMID: 30222214 DOI: 10.1002/chem.201803178] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Indexed: 01/24/2023]
Abstract
After uptake by U87 MG and A375 cancer cells, cobaltabisdicarbollide [COSAN]- distributes between membrane and nucleus and presents no relevant cytotoxicity against both cell lines even for long incubation times. The cytotoxicity of Na[COSAN] was also tested towards one normal cell line, the V79 fibroblasts, in order to ascertain the noncytotoxic profile of the compound. As the cell's nucleus contains DNA, the interaction between [COSAN]- and double-stranded calf thymus DNA (CT-dsDNA) has been investigated. There is a strong interaction between both molecules forming a nanohybrid CT-dsDNA-[COSAN] biomaterial, which was fully characterized. Moreover, Na[COSAN] shows characteristic redox peaks ascribed to the oxidation/reduction of Co3+/2+ at a formal potential of -1.444 V and it can be accumulated at a surface-immobilized DNA layer of glassy carbon electrodes. The equilibrium surface-binding constants (Kox /Kred ), which confirm that [COSAN]- interacts with DNA by an intercalative or electrostatic mode, depending on the ionic strength of the solution, were estimated. In addition, high binding affinity of Na[COSAN] to proteins was observed by 11 B{1 H} NMR and confirmed in vivo. Finally, biodistribution studies of [COSAN]- in normal mice were run. After administration, Na[COSAN] was distributed into many organs but mainly accumulated in the reticuloendothelial system (RES), including liver and spleen. After 1 h, the formation of aggregates by plasma protein interaction plays a role in the biodistribution profile; the aggregates accumulate mostly in the lungs. Na[COSAN], which displays low toxicity and high uptake by relevant cancer cells accumulating boron within the nucleus, could act as a suitable compound for further developments as boron neutron capture therapy (BNCT) agents.
Collapse
Affiliation(s)
- Isabel Fuentes
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Barcelona, Spain
| | - Tania García-Mendiola
- Instituto Madrileño de Estudios Avanzados (IMDEA) Nanociencia, 28049, Madrid, Spain.,Departamento Química AnalíticayAnálisis Instrumental, Universidad Autónoma de Madrid, Spain.,Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Shinichi Sato
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, 226-8503, Japan
| | - Marcos Pita
- Instituto de Catalisis (CSIC), Campus Universidad Autónoma, 28049, Madrid, Spain
| | - Hiroyuki Nakamura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, 226-8503, Japan
| | - Encarnación Lorenzo
- Instituto Madrileño de Estudios Avanzados (IMDEA) Nanociencia, 28049, Madrid, Spain.,Departamento Química AnalíticayAnálisis Instrumental, Universidad Autónoma de Madrid, Spain.,Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Francesc Teixidor
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Barcelona, Spain
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139.7), 2695-066, Bobadela, LRS, Portugal
| | - Clara Viñas
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
68
|
Kuo WY, Hsu HJ, Wu CY, Chen HS, Chou YC, Tsou YL, Peng HP, Jian JW, Yu CM, Chiu YK, Chen IC, Tung CP, Hsiao M, Lin CL, Wang YA, Wang AHJ, Yang AS. Antibody-drug conjugates with HER2-targeting antibodies from synthetic antibody libraries are highly potent against HER2-positive human gastric tumor in xenograft models. MAbs 2018; 11:153-165. [PMID: 30365359 PMCID: PMC6343809 DOI: 10.1080/19420862.2018.1541370] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
HER2-ECD (human epidermal growth factor receptor 2 – extracellular domain) is a prominent therapeutic target validated for treating HER2-positive breast and gastric cancer, but HER2-specific therapeutic options for treating advanced gastric cancer remain limited. We have developed antibody-drug conjugates (ADCs), comprising IgG1 linked via valine-citrulline to monomethyl auristatin E, with potential to treat HER2-positive gastric cancer in humans. The antibodies optimally selected from the ADC discovery platform, which was developed to discover antibody candidates suitable for immunoconjugates from synthetic antibody libraries designed using antibody-antigen interaction principles, were demonstrated to be superior immunoconjugate targeting modules in terms of efficacy and off-target toxicity. In comparison with the two control humanized antibodies (trastuzumab and H32) derived from murine antibody repertoires, the antibodies derived from the synthetic antibody libraries had enhanced receptor-mediated internalization rate, which could result in ADCs with optimal efficacies. Along with the ADCs, two other forms of immunoconjugates (scFv-PE38KDEL and IgG1-AL1-PE38KDEL) were used to test the antibodies for delivering cytotoxic payloads to xenograft tumor models in vivo and to cultured cells in vitro. The in vivo experiments with the three forms of immunoconjugates revealed minimal off-target toxicities of the selected antibodies from the synthetic antibody libraries; the off-target toxicities of the control antibodies could have resulted from the antibodies’ propensity to target the liver in the animal models. Our ADC discovery platform and the knowledge gained from our in vivo tests on xenograft models with the three forms of immunoconjugates could be useful to anyone developing optimal ADC cancer therapeutics.
Collapse
Affiliation(s)
- Wei-Ying Kuo
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Hung-Ju Hsu
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Chun-Yi Wu
- b Department of Biomedical Imaging and Radiological Science, College of Medicine , China Medical University , Taichung , Taiwan
| | - Hong-Sen Chen
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Yu-Chi Chou
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Yueh-Liang Tsou
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Hung-Pin Peng
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Jhih-Wei Jian
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan.,c Institute of Biomedical Informatics , National Yang-Ming University , Taipei , Taiwan.,d Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science , Academia Sinica , Taipei , Taiwan
| | - Chung-Ming Yu
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Yi-Kai Chiu
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Ing-Chien Chen
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Chao-Ping Tung
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Michael Hsiao
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Chia-Lung Lin
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | | | - Andrew H-J Wang
- f Institute of Biological Chemistry , Academia Sinica , Taipei , Taiwan
| | - An-Suei Yang
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| |
Collapse
|
69
|
Zhang Y, Xu J, Yu Y, Shang W, Ye A. Anti-Cancer Drug Sensitivity Assay with Quantitative Heterogeneity Testing Using Single-Cell Raman Spectroscopy. Molecules 2018; 23:molecules23112903. [PMID: 30405051 PMCID: PMC6278387 DOI: 10.3390/molecules23112903] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/01/2018] [Accepted: 11/05/2018] [Indexed: 01/06/2023] Open
Abstract
A novel anti-cancer drug sensitivity testing (DST) approach was developed based on in vitro single-cell Raman spectrum intensity (RSI). Generally, the intensity of Raman spectra (RS) for a single living cell treated with drugs positively relates to the sensitivity of the cells to the drugs. In this study, five cancer cell lines (BGC 823, SGC 7901, MGC 803, AGS, and NCI-N87) were exposed to three cytotoxic compounds or to combinations of these compounds, and then they were evaluated for their responses with RSI. The results of RSI were consistent with conventional DST methods. The parametric correlation coefficient for the RSI and Methylthiazolyl tetrazolium assay (MTT) was 0.8558 ± 0.0850, and the coefficient of determination was calculated as R² = 0.9529 ± 0.0355 for fitting the dose⁻response curve. Moreover, RSI data for NCI-N87 cells treated by trastuzumab, everolimus (cytostatic), and these drugs in combination demonstrated that the RSI method was suitable for testing the sensitivity of cytostatic drugs. Furthermore, a heterogeneity coefficient H was introduced for quantitative characterization of the heterogeneity of cancer cells treated by drugs. The largest possible variance between RSs of cancer cells were quantitatively obtained using eigenvalues of principal component analysis (PCA). The ratio of H between resistant cells and sensitive cells was greater than 1.5, which suggested the H-value was effective to describe the heterogeneity of cancer cells. Briefly, the RSI method might be a powerful tool for simple and rapid detection of the sensitivity of tumor cells to anti-cancer drugs and the heterogeneity of their responses to these drugs.
Collapse
Affiliation(s)
- Yong Zhang
- Key Laboratory for the Physics and Chemistry of Nanodevices, School of Electronics Engineering and Computer Science, Peking University, No.5 Yiheyuan Road, Beijing 100871, China.
- Beijing Institute of Biomedicine, No.15 Xinjiangongmen Road, Beijing 100091, China.
| | - Jingjing Xu
- Academy for Advanced Interdisciplinary Studies, Peking University, No.5 Yiheyuan Road, Beijing 100871, China.
| | - Yuezhou Yu
- Academy for Advanced Interdisciplinary Studies, Peking University, No.5 Yiheyuan Road, Beijing 100871, China.
| | - Wenhao Shang
- Academy for Advanced Interdisciplinary Studies, Peking University, No.5 Yiheyuan Road, Beijing 100871, China.
| | - Anpei Ye
- Key Laboratory for the Physics and Chemistry of Nanodevices, School of Electronics Engineering and Computer Science, Peking University, No.5 Yiheyuan Road, Beijing 100871, China.
- Academy for Advanced Interdisciplinary Studies, Peking University, No.5 Yiheyuan Road, Beijing 100871, China.
| |
Collapse
|
70
|
Harnessing the immune system in glioblastoma. Br J Cancer 2018; 119:1171-1181. [PMID: 30393372 PMCID: PMC6251037 DOI: 10.1038/s41416-018-0258-8] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common primary malignant brain tumour. Survival is poor and improved treatment options are urgently needed. Although immunotherapies have emerged as effective treatments for a number of cancers, translation of these through to brain tumours is a distinct challenge, particularly due to the blood-brain barrier and the unique immune tumour microenvironment afforded by CNS-specific cells. This review discusses the immune system within the CNS, mechanisms of immune escape employed by glioblastoma, and the immunological effects of conventional glioblastoma treatments. Novel therapies for glioblastoma that harness the immune system and their current clinical progress are outlined, including cancer vaccines, T-cell therapies and immune checkpoint modulators.
Collapse
|
71
|
Bonello M, Sims AH, Langdon SP. Human epidermal growth factor receptor targeted inhibitors for the treatment of ovarian cancer. Cancer Biol Med 2018; 15:375-388. [PMID: 30766749 PMCID: PMC6372909 DOI: 10.20892/j.issn.2095-3941.2018.0062] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/26/2018] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is the second most lethal gynecological cancer worldwide and while most patients respond to initial therapy, they often relapse with resistant disease. Human epidermal growth factor receptors (especially HER1/EGFR and HER2/ERBB2) are involved in disease progression; hence, strategies to inhibit their action could prove advantageous in ovarian cancer patients, especially in patients resistant to first line therapy. Monoclonal antibodies and tyrosine kinase inhibitors are two classes of drugs that act on these receptors. They have demonstrated valuable antitumor activity in multiple cancers and their possible use in ovarian cancer continues to be studied. In this review, we discuss the human epidermal growth factor receptor family; review emerging clinical studies on monoclonal antibodies and tyrosine kinase inhibitors targeting these receptors in ovarian cancer patients; and propose future research possibilities in this area.
Collapse
Affiliation(s)
- Maria Bonello
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Andrew Harvey Sims
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Simon Peter Langdon
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| |
Collapse
|
72
|
Sokolova EA, Vodeneev VA, Deyev SM, Balalaeva IV. 3D in vitro models of tumors expressing EGFR family receptors: a potent tool for studying receptor biology and targeted drug development. Drug Discov Today 2018; 24:99-111. [PMID: 30205170 DOI: 10.1016/j.drudis.2018.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/27/2018] [Accepted: 09/05/2018] [Indexed: 12/29/2022]
Abstract
Carcinomas overexpressing EGFR family receptors are of high clinical importance, because the receptors have prognostic value and are used as molecular targets for anticancer therapy. Insufficient drug efficacy necessitates further in-depth research of the receptor biology and improvement in preclinical stages of drug evaluation. Here, we review the currently used advanced 3D in vitro models of tumors, including tumor spheroids, models in natural and synthetic matrices, tumor organoids and microfluidic-based models, as a potent tool for studying EGFR biology and targeted drug development. We are especially focused on factors that affect the biology of tumor cells, causing modification in the expression and basic phosphorylation of the receptors, crosstalk with other signaling pathways and switch between downstream cascades, resulting ultimately in the resistance to antitumor agents.
Collapse
Affiliation(s)
- Evgeniya A Sokolova
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia; Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklay St., Moscow 117997, Russia
| | - Vladimir A Vodeneev
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia
| | - Sergey M Deyev
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia; Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklay St., Moscow 117997, Russia
| | - Irina V Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky University, 23 Gagarin ave., Nizhny Novgorod 603950, Russia; I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya str., Moscow 119991, Russia.
| |
Collapse
|
73
|
Román-Rosales AA, García-Villa E, Herrera LA, Gariglio P, Díaz-Chávez J. Mutant p53 gain of function induces HER2 over-expression in cancer cells. BMC Cancer 2018; 18:709. [PMID: 29970031 PMCID: PMC6029411 DOI: 10.1186/s12885-018-4613-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/20/2018] [Indexed: 11/10/2022] Open
Abstract
Background HER2 over-expression is related with a poor prognosis in patients with invasive breast cancer tumors. Clinical associations have reported that somatic mutations of p53 more frequently detected in cases of sporadic breast cancer of the HER2 subtypes, besides a high percentage of HER2-amplifying tumors carry germline mutations of p53. The mechanisms responsible for the acquisition of oncogenic functions of p53 mutant proteins (mtp53), known as Gain of Function (GOF), over HER2 expression have not been reported. The objective of this study was to evaluate a possible relationship between p53 mutants and HER2 regulation. Methods HER2 expression (transcription and protein), as well as HER2 protein stabilization have been evaluated after inducing or silencing of p53 mutants’ expression in cell lines. Finally, we evaluated the interaction of the p53 mutants over the HER2 receptor promoter. Results Higher HER2 expression in cell lines harboring endogenous mtp53 compared with wt or null expression of p53 cell lines. Transfection of p53 mutants (R248Q and R273C) in cell lines increased the expression of HER2. Silencing of p53 mutants, decrease HER2 expression. The p53 mutants R248Q and R273C significantly increase the luciferase activity on the HER2 promoter, and both mutants also promote acetylation of H3 and H4 histones binding in it. Conclusions These findings show for the first time that p53 mutants induce over-expression of HER2 at transcriptional level of the HER2 protein. Our results could have clinical implications in breast cancer and other types of cancer where HER2 is over-expressed and used as a therapy target. Electronic supplementary material The online version of this article (10.1186/s12885-018-4613-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- A A Román-Rosales
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Av. San Fernando No. 22, Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.,Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), 07360, Ciudad de México, Mexico
| | - E García-Villa
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), 07360, Ciudad de México, Mexico
| | - L A Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Av. San Fernando No. 22, Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico
| | - P Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), 07360, Ciudad de México, Mexico.
| | - J Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Av. San Fernando No. 22, Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| |
Collapse
|
74
|
A clinicopathological study of parotid carcinoma: 18-year review of 171 patients at a single institution. Int J Clin Oncol 2018; 23:615-624. [PMID: 29564569 PMCID: PMC6097105 DOI: 10.1007/s10147-018-1266-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/14/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND This study investigated the clinical outcomes of patients with parotid carcinoma at a single institution during an 18-year period, with the focus on diagnosis, treatment, and survival. METHODS The subjects were 171 patients with parotid carcinoma treated at our department during the 18-year period from September 1999 to August 2017. There were 19 patients in stage I, 65 patients in stage II, 22 patients in stage III, and 65 patients in stage IV. The symptoms, preoperative diagnosis, node metastasis, survival rate, prognostic factors, and immunohistological findings were investigated. RESULTS Preoperative diagnosis of the histological grade by fine-needle aspiration cytology was only possible in 34% of the patients, while the histological grade was correctly determined by frozen section biopsy in 72%. The overall frequency of lymph node metastasis was 29%, with 59% in patients with high-grade carcinoma and only 6% in those with low-/intermediate-grade tumors. The disease-specific 5-year survival rate was 100% for patients in stage I, 95.2% in stage II, 70.4% in stage III, and 45.1% in stage IV. Multivariate analysis showed that the pathological grade was the most important prognostic factor. Immunohistological investigation showed patients with HER-2 or androgen receptor-positive tumors had a significantly worse prognosis. CONCLUSIONS Although a high-grade tumor is the most important prognostic factor, preoperative diagnosis of the grade was not always accurate. Since advanced cancer has a poor prognosis with a limited response to surgery and radiation therapy, development of new treatment strategies, such as molecular-targeting therapies directed against HER-2 and AR, is required.
Collapse
|
75
|
Emanuele S, D'Anneo A, Calvaruso G, Cernigliaro C, Giuliano M, Lauricella M. The Double-Edged Sword Profile of Redox Signaling: Oxidative Events As Molecular Switches in the Balance between Cell Physiology and Cancer. Chem Res Toxicol 2018. [PMID: 29513521 DOI: 10.1021/acs.chemrestox.7b00311] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intracellular redox state in the cell depends on the balance between the level of reactive oxygen species (ROS) and the activity of defensive systems including antioxidant enzymes. This balance is a dynamic process that can change in relation to many factors and/or stimuli induced within the cell. ROS production is derived from physiological metabolic events. For instance, mitochondria represent the major ROS sources during oxidative phosphorylation, but other systems, such as NADPH oxidase or specific enzymes in certain metabolisms, may account for ROS production as well. Whereas high levels of ROS perturb the cell environment, causing oxidative damage to biological macromolecules, low levels of ROS can exert a functional role in the cell, influencing the activity of specific enzymes or modulating some intracellular signaling cascades. Of particular interest appears to be the role of ROS in tumor systems not only because ROS are known to be tumorigenic but also because tumor cells are able to modify their redox state, regulating ROS production to sustain tumor growth and proliferation. Overall, the scope of this review was to critically discuss the most recent findings pertaining to ROS physiological roles as well as to highlight the controversial involvement of ROS in tumor systems.
Collapse
|
76
|
Verma S, Goyal S, Kumari A, Singh A, Jamal S, Grover A. Structural investigations on mechanism of lapatinib resistance caused by HER-2 mutants. PLoS One 2018; 13:e0190942. [PMID: 29389942 PMCID: PMC5794075 DOI: 10.1371/journal.pone.0190942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/23/2017] [Indexed: 01/12/2023] Open
Abstract
HER-2 belongs to the human epidermal growth factor receptor (HER) family. Via different signal transduction pathways, HER-2 regulates normal cell proliferation, survival, and differentiation. Recently, it was reported that MCF10A, BT474, and MDA-MB-231 cells bearing the HER2 K753E mutation were resistant to lapatinib. Present study revealed that HER-2 mutant K753E showed some contrasting behaviour as compared to wild, L768S and V773L HER-2 in complex with lapatinib while similar to previously known lapatinib resistant L755S HER-2 mutant. Lapatinib showed stable but reverse orientation in binding site of K753E and the highest binding energy among studied HER2-lapatinib complexes but slightly lesser than L755S mutant. Results indicate that K753E has similar profile as L755S mutant for lapatinib. The interacting residues were also found different from other three studied forms as revealed by free energy decomposition and ligplot analysis.
Collapse
Affiliation(s)
- Sharad Verma
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sukriti Goyal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan, India
| | - Anchala Kumari
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Department of Biotechnology, TERI School of Advanced Studies, Vasant Kunj, New Delhi, India
| | - Aditi Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Department of Biotechnology, TERI School of Advanced Studies, Vasant Kunj, New Delhi, India
| | - Salma Jamal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan, India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- * E-mail:
| |
Collapse
|
77
|
Abdel-Fatah TMA, Rees RC, Pockley AG, Moseley P, Ball GR, Chan SYT, Ellis IO, Miles AK. The localization of pre mRNA splicing factor PRPF38B is a novel prognostic biomarker that may predict survival benefit of trastuzumab in patients with breast cancer overexpressing HER2. Oncotarget 2017; 8:112245-112257. [PMID: 29348822 PMCID: PMC5762507 DOI: 10.18632/oncotarget.22496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/28/2017] [Indexed: 11/25/2022] Open
Abstract
Cancer biomarkers that can define disease status and provide a prognostic insight are essential for the effective management of patients with breast cancer (BC). The prevalence, clinicopathological and prognostic significance of PRPF38B expression in a consecutive series of 1650 patients with primary invasive breast carcinoma were examined using immunohistochemistry. Furthermore, the relationship(s) between clinical outcome and PRPF38B expression was explored in 627 patients with ER-negative (oestrogen receptor) disease, and 322 patients with HER2-overexpressing disease. Membranous expression of PRPF38B was observed in 148/1388 (10.7%) cases and was significantly associated with aggressive clinicopathological features, including high grade, high mitotic index, pleomorphism, invasive ductal carcinoma of no specific type (IDC-NST), ER-negative, HER2-overexpression and p53 mutational status (all p < 0.01). In patients with ER-negative disease receiving chemotherapy, nuclear expression of PRPF38B was significantly associated with a reduced risk of relapse (p = 0.0004), whereas membranous PRPF38B expression was significantly associated with increased risk of relapse (p = 0.004; respectively) at a 5 year follow-up. When patients were stratified according to ER-negative/HER2-positive status, membranous PRPF38B expression was associated with a higher risk of relapse in those patients that did not receive trastuzumab therapy (p = 0.02), whereas in those patients with ER-negative/HER2-positive disease that received trastuzumab adjuvant therapy, membranous PRPF38B expression associated with a lower risk of relapse (p = 0.00018). Nuclear expression of PRPF38B is a good prognostic indicator in both ER-negative patients and ER-negative/HER2-positive BC (breast cancer) patients, whereas membranous localisation of PRPF38B is a poor prognostic biomarker that predicts survival benefit from trastuzumab therapy in patients with ER-negative/HER2-overexpressing BC.
Collapse
Affiliation(s)
- Tarek M A Abdel-Fatah
- Department of Clinical Oncology, Nottingham City Hospital, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Robert C Rees
- The John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - A Graham Pockley
- The John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Paul Moseley
- Department of Clinical Oncology, Nottingham City Hospital, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Graham R Ball
- The John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Stephen Y T Chan
- Department of Clinical Oncology, Nottingham City Hospital, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Ian O Ellis
- Department of Histopathology, Nottingham City Hospital, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Amanda K Miles
- The John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| |
Collapse
|
78
|
Han JH, Park J, Seo YW, Kim TH. Designing a cancer therapeutic peptide by combining the mitochondrial targeting domain of Noxa and ErbB2-targeting moieties. FEBS Lett 2017; 592:103-111. [PMID: 29193033 DOI: 10.1002/1873-3468.12922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/15/2017] [Accepted: 11/21/2017] [Indexed: 11/06/2022]
Abstract
Many anticancer drugs target epidermal growth factor receptors to inhibit receptor tyrosine kinases and tumor growth. Here, we show that an ErbB2-targeting pronecrotic peptide (KWSY:MTD) selectively kills tumor cells expressing ErbB2 in vitro. An antibody against ErbB2 inhibits KWSY:MTD-induced cell death. KWSY:MTD causes membrane permeability which allows propidium iodide entry into the cytosol and the release of HMGB1 into the media, indicative of necrosis. Mitochondrial swelling occurs in response to KWSY:MTD. Moreover, in vivo analysis using a mouse model shows that KWSY:MTD partially suppressed growth in tumor tissue bearing ErbB2-expressing cells, but did not have obvious toxicity in mouse liver or kidney tissue. Taken together, KWSY:MTD has potential as an ErbB2-targeting anticancer drug.
Collapse
Affiliation(s)
- Ji-Hye Han
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwang-Ju, Korea
| | - Junghee Park
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwang-Ju, Korea
| | - Young-Woo Seo
- Korea Basic Science Institute Gwang-Ju Center, Chonnam National University, Gwang-Ju, Korea
| | - Tae-Hyoung Kim
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwang-Ju, Korea
| |
Collapse
|
79
|
Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, Wu K. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Exp Hematol Oncol 2017; 6:31. [PMID: 29209558 PMCID: PMC5704598 DOI: 10.1186/s40164-017-0091-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/20/2017] [Indexed: 12/15/2022] Open
Abstract
HER2-targeted immunotherapy consists of monoclonal antibodies (e.g. trastuzumab, pertuzumab), bispecific antibodies (e.g. MM-111, ertumaxomab) and activated T cells armed with anti-HER2 bispecific antibody (HER2Bi-aATC). Trastuzumab is a classic drug for the treatment of HER2 positive metastatic breast cancer. The combined application of pertuzumab, trastuzumab and paclitaxel has been suggested as a standard therapy for HER2 positive advanced breast cancer. The resistance to anti-HER2 antibody has resulted in disease progression. HER2-directed bispecific antibody may be a promising therapeutic approach for these patients. Ertumaxomab enhanced the interaction of immune effector cells and tumor cells. MM-111 simultaneously binds to HER2 and HER3 and blocks downstream signaling. Besides, HER2Bi-aATC is also an alternative therapeutic approach for HER2 positive cancers. In this review, we summarized the recent advancement of HER2-targeted monoclonal antibodies (trastuzumab, pertuzumab and T-DM1) and bispecific antibodies (MM-111, ertumaxomab and HER2Bi-aATC), especially focus on clinical trial results.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Xinwei Han
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Anping Li
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
80
|
Yamaoka T, Ohba M, Ohmori T. Molecular-Targeted Therapies for Epidermal Growth Factor Receptor and Its Resistance Mechanisms. Int J Mol Sci 2017; 18:ijms18112420. [PMID: 29140271 PMCID: PMC5713388 DOI: 10.3390/ijms18112420] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/11/2017] [Accepted: 11/12/2017] [Indexed: 12/20/2022] Open
Abstract
Cancer therapies targeting epidermal growth factor receptor (EGFR), such as small-molecule kinase inhibitors and monoclonal antibodies, have been developed as standard therapies for several cancers, such as non-small cell lung cancer, colorectal cancer, pancreatic cancer, breast cancer, and squamous cell carcinoma of the head and neck. Although these therapies can significantly prolong progression-free survival, curative effects are not often achieved because of intrinsic and/or acquired resistance. The resistance mechanisms to EGFR-targeted therapies can be categorized as resistant gene mutations, activation of alternative pathways, phenotypic transformation, and resistance to apoptotic cell death. Analysis of the processes that modulate EGFR signal transduction by EGFR-targeted inhibitors, such as tyrosine kinase inhibitors and monoclonal antibodies, has revealed new therapeutic opportunities and has elucidated novel mechanisms contributing to the discovery of more effective anticancer treatments. In this review, we discuss the roles of EGFR in cancer development, therapeutic strategies for targeting EGFR, and resistance mechanisms to EGFR-targeted therapies, with a focus on cancer therapies for individual patients.
Collapse
Affiliation(s)
- Toshimitsu Yamaoka
- Institute of Molecular Oncology, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Motoi Ohba
- Institute of Molecular Oncology, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Tohru Ohmori
- Institute of Molecular Oncology, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| |
Collapse
|
81
|
Gluck WL, Martin JC, Edenfield WJ, Chung KY, Arguello D. Prolonged response of widely metastatic HER2-positive colon cancer to trastuzumab therapy. COLORECTAL CANCER 2017. [DOI: 10.2217/crc-2017-0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Emerging evidence suggests a small subset of late-stage colon cancer driven by HER2, a biomarker routinely evaluated in select breast and gastric cancers, may respond to HER2-targeted therapy. Herein, we describe a 49-year-old male with widely metastatic colon cancer originating in the sigmoid colon. After failing standard therapy, a biopsy specimen of the tumor was evaluated for novel biomarkers using molecular profiling. After identification of ERBB2 (HER2) amplification using in situ hybridization, the patient subsequently received a trial of trastuzumab monotherapy and experienced a dramatic and durable response. This report builds on our understanding of using precision oncology to improve survival in metastatic colon cancer.
Collapse
Affiliation(s)
- W Larry Gluck
- Greenville Health System, Institute for Translational Oncology Research, Greenville, SC, USA
| | - Julie C Martin
- Greenville Health System, Institute for Translational Oncology Research, Greenville, SC, USA
| | - W Jeffrey Edenfield
- Greenville Health System, Institute for Translational Oncology Research, Greenville, SC, USA
| | - Ki Y Chung
- Greenville Health System, Institute for Translational Oncology Research, Greenville, SC, USA
| | | |
Collapse
|
82
|
Hodgson A, Amemiya Y, Seth A, Cesari M, Djordjevic B, Parra-Herran C. Genomic abnormalities in invasive endocervical adenocarcinoma correlate with pattern of invasion: biologic and clinical implications. Mod Pathol 2017; 30:1633-1641. [PMID: 28731050 DOI: 10.1038/modpathol.2017.80] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 05/06/2017] [Accepted: 05/07/2017] [Indexed: 02/07/2023]
Abstract
The pattern-based classification system for HPV-related endocervical adenocarcinoma, which classifies tumors based on the destructiveness of stromal invasion, is predictive of the risk of nodal metastases and adverse outcome. Previous studies have demonstrated clinically important molecular alterations in endocervical adenocarcinoma, including KRAS and PIK3CA mutations; however, correlation between the molecular landscape and pathological variables including pattern of invasion has not been thoroughly explored. In this study, 20 endocervical adenocarcinomas were classified using the pattern-based classification system and were subjected to targeted sequencing using the Ion AmpliSeq Cancer Hotspot Panel v2 (ThermoFisher Scientific, Waltham, MA, USA) that surveys hotspot regions of 50 oncogenes and tumor suppressor genes. Single-nucleotide polymorphisms were correlated with clinical and pathologic variables including pattern of invasion. Five (25%), six (30%), and nine (45%) cases were classified as patterns A, B, and C respectively. Lymph node metastases, advanced stage at presentation and mortality from disease were exclusively seen in destructively invasive tumors (patterns B or C). Prevalent mutations in the cohort involved PIK3CA (30%), KRAS (30%), MET (15%), and RB1 (10%). Most (94%) relevant genomic alterations were present in destructively invasive tumors with PIK3CA, KRAS, and RB1 mutations seen exclusively in pattern B or C subgroups. KRAS mutations correlated with advanced stage at presentation (FIGO stage II or higher). Our findings indicate that the pattern of stromal invasion correlates with genomic abnormalities detected by next-generation sequencing, suggesting that tumors without destructive growth (pattern A) are biologically distinct from those with destructive invasion (patterns B and C), and that pattern B endocervical adenocarcinoma is more closely related to its pattern C counterpart. The pattern-based classification may be used as a triage tool when considering molecular testing for prognostic or therapeutic purposes.
Collapse
Affiliation(s)
- Anjelica Hodgson
- Department of Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Yutaka Amemiya
- Department of Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Genomics Core Facility, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Arun Seth
- Department of Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Genomics Core Facility, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Matthew Cesari
- Department of Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Bojana Djordjevic
- Department of Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Carlos Parra-Herran
- Department of Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
83
|
Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: a phase 1 dose-escalation study. Lancet Oncol 2017; 18:1512-1522. [PMID: 29037983 DOI: 10.1016/s1470-2045(17)30604-6] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Antibody-drug conjugates have emerged as a powerful strategy in cancer therapy and combine the ability of monoclonal antibodies to specifically target tumour cells with the highly potent killing activity of drugs with payloads too toxic for systemic administration. Trastuzumab deruxtecan (also known as DS-8201) is an antibody-drug conjugate comprised of a humanised antibody against HER2, a novel enzyme-cleavable linker, and a topoisomerase I inhibitor payload. We assessed its safety and tolerability in patients with advanced breast and gastric or gastro-oesophageal tumours. METHODS This was an open-label, dose-escalation phase 1 trial done at two study sites in Japan. Eligible patients were at least 20 years old with breast or gastric or gastro-oesophageal carcinomas refractory to standard therapy regardless of HER2 status. Participants received initial intravenous doses of trastuzumab deruxtecan from 0·8 to 8·0 mg/kg and dose-limiting toxicities were assessed over a 21-day cycle; thereafter, dose reductions were implemented as needed and patients were treated once every 3 weeks until they had unacceptable toxic effects or their disease progressed. Primary endpoints included identification of safety and the maximum tolerated dose or recommended phase 2 dosing and were analysed in all participants who received at least one dose of study drug. The dose-escalation study is the first part of a two-part study with the second dose-expansion part ongoing and enrolling patients as of July 8, 2017, in Japan and the USA. This trial is registered at ClinicalTrials.gov, number NCT02564900. FINDINGS Between Aug 28, 2015, and Aug 26, 2016, 24 patients were enrolled and received trastuzumab deruxtecan (n=3 for each of 0·8, 1·6, 3·2, and 8·0 mg/kg doses; n=6 for each of 5·4 and 6·4 mg/kg). Up to the study cutoff date of Feb 1, 2017, no dose-limiting toxic effects, substantial cardiovascular toxic effects, or deaths occurred. One patient was removed from the activity analysis because they had insufficient target lesions for analysis. The most common grade 3 adverse events were decreased lymphocyte (n=3) and decreased neutrophil count (n=2); and grade 4 anaemia was reported by one patient. Three serious adverse events-febrile neutropenia, intestinal perforation, and cholangitis-were reported by one patient each. Overall, in 23 evaluable patients, including six patients with low HER2-expressing tumours, ten patients achieved an objective response (43%, 95% CI 23·2-65·5). Disease control was achieved in 21 (91%; 95% CI 72·0-98·9) of 23 patients. Median follow-up time was 6·7 months (IQR 4·4-10·2), with nine (90%) of ten responses seen at doses of 5·4 mg/kg or greater. INTERPRETATION The maximum tolerated dose of trastuzumab deruxtecan was not reached. In this small, heavily pretreated study population, trastuzumab deruxtecan showed antitumour activity, even in low HER2-expressing tumours. Based on safety and activity, the most likely recommended phase 2 dosing is 5·4 or 6·4 mg/kg. FUNDING Daiichi Sankyo Co, Ltd.
Collapse
|
84
|
Halle MK, Ojesina AI, Engerud H, Woie K, Tangen IL, Holst F, Høivik E, Kusonmano K, Haldorsen IS, Vintermyr OK, Trovik J, Bertelsen BI, Salvesen HB, Krakstad C. Clinicopathologic and molecular markers in cervical carcinoma: a prospective cohort study. Am J Obstet Gynecol 2017; 217:432.e1-432.e17. [PMID: 28599900 DOI: 10.1016/j.ajog.2017.05.068] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/15/2017] [Accepted: 05/31/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cervical cancer is a major health problem worldwide. Identification of effective clinicopathologic and molecular markers is vital to improve treatment stratification. OBJECTIVES The purpose of this study was to validate a set of well-defined clinicopathologic features in a large population-based, prospectively collected cervical cancer cohort to support their use in the clinic. Further, we explored p53 and human epidermal growth factor receptor 2 as potential prognostic markers in cervical cancer. STUDY DESIGN Tissue was collected from 401 patients with cervical cancer. Clinical data that included follow-up evaluations were collected from patient journals. Histopathologic data were evaluated and revised by an expert pathologist. The prognostic impact of selected clinicopathologic variables was analyzed in the whole cohort. Tissue microarrays were prepared from 292 carcinomas, and p53 and human epidermal growth factor receptor 2 protein levels were evaluated by immunohistochemistry. Fresh frozen samples from overlapping cervical carcinomas previously were subjected to human papilloma virus typing (n=94), whole exome (n=100) and RNA (n=79) sequencing; the results were available for our analyses. RESULTS Among the clinicopathologic variables, vascular space invasion, histologic type, and tumor size were verified as strong independent prognostic markers. High p53 protein levels were associated significantly with markers for aggressive phenotype and survival, also in multivariate survival analysis, but did not reflect TP53 mutational status. High human epidermal growth factor receptor 2 protein levels were identified in 21% of all tumors. ERBB2 amplification was associated with poor outcome (P=.003); human epidermal growth factor receptor 2 protein level was not. CONCLUSIONS Our findings support that the Féderation Internationale de Gynécologie et d'Obstétrique s guidelines should include vascular space invasion and tumor size 2-4 cm and that careful selection of histologic type is essential for stratification of patient risk groups. High p53 levels independently predict poor survival yet do not reflect mutational status in cervical cancer. Amplified ERBB2 significantly links to poor survival, while HercepTest does not. With optimal stratification, human epidermal growth factor receptor 2-based therapy may improve cervical cancer treatment.
Collapse
|
85
|
Mayekar MK, Bivona TG. Current Landscape of Targeted Therapy in Lung Cancer. Clin Pharmacol Ther 2017; 102:757-764. [PMID: 28786099 DOI: 10.1002/cpt.810] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Comprehensive genomic profiling of lung cancers revealed their genetic heterogeneity and complexity and identified numerous targetable oncogenic driver alterations. These molecular profiling efforts have made it possible to exploit the potential of molecularly targeted therapies. Selection of patients for targeted therapies is becoming biomarker-driven, where the oncogenic drivers in patient tumors are first identified, and subsequently patients bearing drug-sensitizing genetic aberrations are matched to the appropriate targeted therapy. Success of this design of clinical trials and practice was first demonstrated in EGFR inhibitor trials in lung cancer and has since been incorporated into subsequent targeted therapy trials including ALK-, ROS1-, and BRAF V600E-targeted therapies. In this review we discuss the current landscape of clinically approved and other promising molecularly targeted approaches for the treatment of lung cancers, the challenges with these approaches, and the strategies that could be deployed to overcome these challenges.
Collapse
Affiliation(s)
- Manasi K Mayekar
- Department of Medicine, University of California, San Francisco, San Francisco, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
86
|
Wang Y, Fan S, Zhong W, Zhou X, Li S. Development and Properties of Valine-Alanine based Antibody-Drug Conjugates with Monomethyl Auristatin E as the Potent Payload. Int J Mol Sci 2017; 18:ijms18091860. [PMID: 28841157 PMCID: PMC5618509 DOI: 10.3390/ijms18091860] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/07/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022] Open
Abstract
Antibody-drug conjugates (ADCs), designed to selectively deliver cytotoxic agents to antigen-bearing cells, are poised to become an important class of cancer therapeutics. Human epithelial growth factor receptor (HER2) is considered an effective target for cancer treatment, and a HER2-targeting ADC has shown promising results. Most ADCs undergoing clinical evaluation contain linkers that have a lysosomal protease-cleavable dipeptide, of which the most common is valine-citrulline (VC). However, valine-alanine (VA), another dipeptide comprising two human essential amino acids, has been used in next generation ADCs loading new toxins, but the druggable properties of ADCs loaded the most popular monomethyl auristatin E (MMAE) remain to be further explored. In this study, we generated VA-based ADCs that connected MMAE to an anti-HER2 antibody. We studied the differences in the preparation process, in vitro stability, cathepsin B activity and in vitro cytotoxicity of VA-based ADC compared to the ADC of VC. VA had comparable performance to VC, which preliminarily displays its practicability. Additional efficacy and safety studies in a xenograft model indicate this novel ADC exerted potent anti-tumor activity and negligible toxicity. The results of this study show the application potential of VA-based ADC with MMAE as the payload.
Collapse
Affiliation(s)
- Yanming Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China.
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Shiyong Fan
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Wu Zhong
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Xinbo Zhou
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Song Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China.
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
87
|
HER2 and TOP2A Gene Amplification and Protein Expression in Upper Tract Urothelial Carcinomas. Pathol Oncol Res 2017; 24:575-581. [DOI: 10.1007/s12253-017-0260-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 06/21/2017] [Indexed: 11/30/2022]
|
88
|
Heestand GM, Schwaederle M, Gatalica Z, Arguello D, Kurzrock R. Topoisomerase expression and amplification in solid tumours: Analysis of 24,262 patients. Eur J Cancer 2017; 83:80-87. [PMID: 28728050 PMCID: PMC5613945 DOI: 10.1016/j.ejca.2017.06.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/16/2017] [Indexed: 01/07/2023]
Abstract
Background Topoisomerase I (TOPO1) and topoisomerase IIα (TOP2A) are specific targets of multiple chemotherapy drugs. Increased expression of TOPO1 protein and amplification of the TOP2A gene have been associated with treatment response in colorectal and breast cancers, respectively. TOPO1 and TOP2A may be potential therapeutic targets in other malignancies as well. Summary of methods We analysed TOPO1 protein expression and TOP2A gene amplification in patients (n = 24,262 specimens) with diverse cancers. Since HER2 and TOP2A co-amplification have been investigated for predictive value regarding anthracycline benefit, we analysed specimens for HER2 amplification as well. Results Overexpressed TOPO1 protein was present in 51% of the tumours. Four percent of the tumours had TOP2A amplification, with gallbladder tumours and gastroesophageal/oesophageal tumours having rates over 10%. Overall, 4903 specimens were assessed for both TOP2A and HER2 amplification; 129 (2.6%) had co-amplification. High rates (>40%) of HER2 amplification were seen in patients with TOP2A amplification in breast, ovarian, gastroesophageal/oesophageal and pancreatic cancer. Conclusion Our data indicate that increased TOPO1 expression and TOP2A amplification, as well as HER2 co-alterations, are present in multiple malignancies. The implications of these observations regarding sensitivity to chemotherapy not traditionally administered to these tumour types merits investigation.
Collapse
Affiliation(s)
- Gregory M Heestand
- Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093, USA.
| | - Maria Schwaederle
- Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093, USA.
| | - Zoran Gatalica
- Caris Life Sciences, 4750 South 44th Place, Phoenix, AZ 85040, USA.
| | - David Arguello
- Caris Life Sciences, 4750 South 44th Place, Phoenix, AZ 85040, USA.
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
89
|
Takegawa N, Nonagase Y, Yonesaka K, Sakai K, Maenishi O, Ogitani Y, Tamura T, Nishio K, Nakagawa K, Tsurutani J. DS-8201a, a new HER2-targeting antibody-drug conjugate incorporating a novel DNA topoisomerase I inhibitor, overcomes HER2-positive gastric cancer T-DM1 resistance. Int J Cancer 2017; 141:1682-1689. [PMID: 28677116 DOI: 10.1002/ijc.30870] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/27/2017] [Accepted: 06/26/2017] [Indexed: 12/27/2022]
Abstract
Anti-HER2 therapies are beneficial for patients with HER2-positive breast or gastric cancer. T-DM1 is a HER2-targeting antibody-drug conjugate (ADC) comprising the antibody trastuzumab, a linker, and the tubulin inhibitor DM1. Although effective in treating advanced breast cancer, all patients eventually develop T-DM1 resistance. DS-8201a is a new ADC incorporating an anti-HER2 antibody, a newly developed, enzymatically cleavable peptide linker, and a novel, potent, exatecan-derivative topoisomerase I inhibitor (DXd). DS-8201a has a drug-to-antibody-ratio (DAR) of 8, which is higher than that of T-DM1 (3.5). Owing to these unique characteristics and unlike T-DM1, DS-8201a is effective against cancers with low-HER2 expression. In the present work, T-DM1-resistant cells (N87-TDMR), established using the HER2-positive gastric cancer line NCI-N87 and continuous T-DM1 exposure, were shown to be susceptible to DS-8201a. The ATP-binding cassette (ABC) transporters ABCC2 and ABCG2 were upregulated in N87-TDMR cells, but HER2 overexpression was retained. Furthermore, inhibition of ABCC2 and ABCG2 by MK571 restored T-DM1 sensitivity. Therefore, resistance to T-DM1 is caused by efflux of its payload DM1, due to aberrant expression of ABC transporters. In contrast to DM1, DXd payload of DS-8201a inhibited the growth of N87-TDMR cells in vitro. This suggests that either DXd may be a poor substrate of ABCC2 and ABCG2 in comparison to DM1, or the high DAR of DS-8201a relative to T-DM1 compensates for increased efflux. Notably, N87-TDMR xenograft tumor growth was prevented by DS-8201a. In conclusion, the efficacy of DS-8201a as a treatment for patients with T-DM1-resistant breast or gastric cancer merits investigation.
Collapse
Affiliation(s)
- Naoki Takegawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Yoshikane Nonagase
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Kimio Yonesaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Osamu Maenishi
- Department of Pathology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Yusuke Ogitani
- Biologics & Immuno-Oncology Laboratories, Daiichi Sankyo Co., Ltd, Shinagawa-ku, Tokyo, Japan
| | - Takao Tamura
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| | - Junji Tsurutani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayamashi, Osaka, Japan
| |
Collapse
|
90
|
Birkeland AC, Yanik M, Tillman BN, Scott MV, Foltin SK, Mann JE, Michmerhuizen NL, Ludwig ML, Sandelski MM, Komarck CM, Carey TE, Prince MEP, Bradford CR, McHugh JB, Spector ME, Brenner JC. Identification of Targetable ERBB2 Aberrations in Head and Neck Squamous Cell Carcinoma. JAMA Otolaryngol Head Neck Surg 2017; 142:559-67. [PMID: 27077364 DOI: 10.1001/jamaoto.2016.0335] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE ERBB2 (formerly HER2) is an important drug target in breast cancer, where anti-ERBB2 therapy has been shown to lead to improvements in disease recurrence and overall survival. ERBB2 status in head and neck squamous cell carcinoma (HNSCC) has not been well studied. Identification of ERBB2-positive tumors and characterization of response to ERBB2 therapy could lead to targeted treatment options in HNSCC. OBJECTIVE To identify ERBB2 aberrations in HNSCCs and investigate the potential for ERBB2-targeted therapy in HNSCCs. DESIGN, SETTING, AND PARTICIPANTS A retrospective case series of patients with laryngeal (42 tumor specimens) and oral cavity (94 tumor specimens) SCC enrolled in the University of Michigan Head and Neck Specialized Program of Research Excellence was conducted. Publicly available sequencing data (The Cancer Genome Atlas), as well as data from other studies, were reviewed to identify additional mutations and overexpression in ERBB2 in HNSCC. Established HNSCC cell lines were used for follow-up in vitro analysis. The study was conducted from October 1, 2014, to August 30, 2015. INTERVENTIONS With the use of targeted, amplicon-based sequencing with the Oncomine Cancer Panel, the copy number and mutation status of commonly altered genes in HNSCCs were assessed. Immunohistochemical staining was performed on tissue microarrays of HNSCCs to assess the expression of ERBB2. Western blotting for HNSCC cell line ERBB2 expression and cell survival assays after treatment with ERBB2 inhibitors were performed. MAIN OUTCOMES AND MEASURES The prevalence of ERBB2 genetic aberrations and ERBB2 overexpression in laryngeal and oral cavity SCCs, prevalence of ERBB2 aberrations in HNSCC in The Cancer Genome Atlas, ERBB2 protein expression in HNSCC cell lines, and response of HNSCC cell lines to targeted ERBB2 inhibitors. RESULTS Of the 42 laryngeal SCC samples screened by targeted sequencing, 4 (10%) were positive for ERBB2 amplification. Two of these samples showed ERBB2 overexpression on immunohistochemistry. Two of the 94 oral cavity SCC samples (2%) were positive for ERBB2 on immunohistochemistry. Analysis of 288 patients from publicly available HNSCC sequencing data revealed 9 amplifications (3%) in ERBB2. Protein expression was variable across HNSCC cell lines, and a subset of these cell lines showed responsiveness to anti-ERBB2 therapy. CONCLUSIONS AND RELEVANCE ERBB2 aberrations were identified in a subset of HNSCCs. These tumors may be responsive to targeted therapy against ERBB2. Screening for ERBB2 aberrations and applying targeted therapy in ERBB2-positive patients may be useful in personalized therapy trials, particularly in patients who are refractory to current treatment paradigms.
Collapse
Affiliation(s)
- Andrew C Birkeland
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Megan Yanik
- medical student, University of Michigan Medical School, Ann Arbor
| | - Brittny N Tillman
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Megan V Scott
- medical student, University of Michigan Medical School, Ann Arbor
| | - Susan K Foltin
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Jacqueline E Mann
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Nicole L Michmerhuizen
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Megan L Ludwig
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | | | - Christine M Komarck
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor
| | - Thomas E Carey
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - Mark E P Prince
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - Carol R Bradford
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - Jonathan B McHugh
- Department of Pathology, University of Michigan Medical School, Ann Arbor
| | - Matthew E Spector
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| | - J Chad Brenner
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor3Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor
| |
Collapse
|
91
|
Couto M, Mastandrea I, Cabrera M, Cabral P, Teixidor F, Cerecetto H, Viñas C. Small-Molecule Kinase-Inhibitors-Loaded Boron Cluster as Hybrid Agents for Glioma-Cell-Targeting Therapy. Chemistry 2017; 23:9233-9238. [PMID: 28605114 DOI: 10.1002/chem.201701965] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Indexed: 12/30/2022]
Abstract
The reported new anilinoquinazoline-icosahedral borane hybrids have been evaluated as glioma targeting for potential use in cancer therapy. Their anti-glioma activity depends on hybrids' lipophilicity; the most powerful compound against glioma cells, a 1,7-closo-derivative, displayed at least 3.3 times higher activity than the parent drug erlotinib. According to the cytotoxic effects on normal glia cells, the hybrids were selective for epidermal growth factor receptor (EGFR)-overexpressed tumor cells. These boron carriers could be used to enrich glioma cancer cells with boron for cancer therapy.
Collapse
Affiliation(s)
- Marcos Couto
- Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400, Montevideo, Uruguay.,Institut de Ciències dels Materials de Barcelona, ICMAB-CSIC Campus UAB, 08193, Bellaterra, Spain.,Área de Radiofarmacia, Centro de Investigaciones Nucleares Facultad de Ciencias, Universidad de la República, Mataojo 2055, 11400, Montevideo, Uruguay
| | - Ignacio Mastandrea
- Centro Universitario Paysandú. CenUR Litoral Norte, Universidad de la República., 60000, Paysandú, Uruguay.,Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Mauricio Cabrera
- Centro Universitario Paysandú. CenUR Litoral Norte, Universidad de la República., 60000, Paysandú, Uruguay
| | - Pablo Cabral
- Área de Radiofarmacia, Centro de Investigaciones Nucleares Facultad de Ciencias, Universidad de la República, Mataojo 2055, 11400, Montevideo, Uruguay
| | - Francesc Teixidor
- Institut de Ciències dels Materials de Barcelona, ICMAB-CSIC Campus UAB, 08193, Bellaterra, Spain
| | - Hugo Cerecetto
- Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400, Montevideo, Uruguay.,Área de Radiofarmacia, Centro de Investigaciones Nucleares Facultad de Ciencias, Universidad de la República, Mataojo 2055, 11400, Montevideo, Uruguay
| | - Clara Viñas
- Institut de Ciències dels Materials de Barcelona, ICMAB-CSIC Campus UAB, 08193, Bellaterra, Spain
| |
Collapse
|
92
|
Vitale SG, Laganà AS, Capriglione S, Angioli R, La Rosa VL, Lopez S, Valenti G, Sapia F, Sarpietro G, Butticè S, Tuscano C, Fanale D, Tropea A, Rossetti D. Target Therapies for Uterine Carcinosarcomas: Current Evidence and Future Perspectives. Int J Mol Sci 2017; 18:ijms18051100. [PMID: 28531111 PMCID: PMC5455008 DOI: 10.3390/ijms18051100] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/13/2017] [Accepted: 05/16/2017] [Indexed: 01/04/2023] Open
Abstract
Carcinosarcomas (CS) in gynecology are very infrequent and represent only 2-5% of uterine cancers. Despite surgical cytoreduction and subsequent chemotherapy being the primary treatment for uterine CS, the overall five-year survival rate is 30 ± 9% and recurrence is extremely common (50-80%). Due to the poor prognosis of CS, new strategies have been developed in the last few decades, targeting known dysfunctional molecular pathways for immunotherapy. In this paper, we aimed to gather the available evidence on the latest therapies for the treatment of CS. We performed a systematic review using the terms "uterine carcinosarcoma", "uterine Malignant Mixed Müllerian Tumors", "target therapies", "angiogenesis therapy", "cancer stem cell therapy", "prognostic biomarker", and "novel antibody-drug". Based on our results, the differential expression and accessibility of epithelial cell adhesion molecule-1 on metastatic/chemotherapy-resistant CS cells in comparison to normal tissues and Human Epidermal Growth Factor Receptor 2 (HER2) open up new possibilities in the field of target therapy. Nevertheless, future investigations are needed to clarify the impact of these new therapies on survival rate and medium-/long-term outcomes.
Collapse
Affiliation(s)
- Salvatore Giovanni Vitale
- Unit of Gynecology and Obstetrics, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Antonio Simone Laganà
- Unit of Gynecology and Obstetrics, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Stella Capriglione
- Department of Obstetrics and Gynecology, Campus Bio Medico University of Rome, 00128 Rome, Italy.
| | - Roberto Angioli
- Department of Obstetrics and Gynecology, Campus Bio Medico University of Rome, 00128 Rome, Italy.
| | - Valentina Lucia La Rosa
- Unit of Psychodiagnostics and Clinical Psychology, University of Catania, 95124 Catania, Italy.
| | - Salvatore Lopez
- Department of Obstetrics and Gynecology, Campus Bio Medico University of Rome, 00128 Rome, Italy.
| | - Gaetano Valenti
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Fabrizio Sapia
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Giuseppe Sarpietro
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Salvatore Butticè
- Department of Human Pathology, Unit of Urology, University of Messina, 98124 Messina, Italy.
| | - Carmelo Tuscano
- Radiation Oncology Department, AO "Bianchi-Melacrino-Morelli", 89124 Reggio Calabria, Italy.
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy.
| | - Alessandro Tropea
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Diego Rossetti
- Unit of Gynecology and Obstetrics, Desenzano del Garda Hospital, Section of Gavardo, 25085 Gavardo, Brescia, Italy.
| |
Collapse
|
93
|
Advances in systemic therapy for metastatic breast cancer: future perspectives. Med Oncol 2017; 34:119. [PMID: 28526922 DOI: 10.1007/s12032-017-0975-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 04/29/2017] [Indexed: 12/16/2022]
Abstract
Breast cancer (BC) is the most common cancer in women worldwide. One in eight women will develop the disease in her lifetime. Notwithstanding the incredible progress made in this field, BC still represents the second most common cause of cancer-related death in women. Targeted drugs have revolutionised breast cancer treatment and improved the prognosis as well as the life expectancy of millions of women. However, the phenomenon of primary and secondary pharmacological resistance is becoming increasingly evident, limiting the efficacy of these agents and calling for a better in-depth knowledge and understanding of the biology as well as the biochemical crosstalk underlying the disease. The advent of laboratory technologies in the clinical setting such as the routine use of next generation sequencing has allowed identification of new genetic alterations as well as providing a precise picture of the molecular landscapes of each tumour. Consequently, new specific therapeutic approaches are becoming available to minimise or delay the occurrence of resistance. In this review, we analyse the latest research and news from the clinical development side for each BC subtype.
Collapse
|
94
|
ErbB Family Signalling: A Paradigm for Oncogene Addiction and Personalized Oncology. Cancers (Basel) 2017; 9:cancers9040033. [PMID: 28417948 PMCID: PMC5406708 DOI: 10.3390/cancers9040033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
ErbB family members represent important biomarkers and drug targets for modern precision therapy. They have gained considerable importance as paradigms for oncoprotein addiction and personalized medicine. This review summarizes the current understanding of ErbB proteins in cell signalling and cancer and describes the molecular rationale of prominent cases of ErbB oncoprotein addiction in different cancer types. In addition, we have highlighted experimental technologies for the development of innovative cancer cell models that accurately predicted clinical ErbB drug efficacies. In the future, such cancer models might facilitate the identification and validation of physiologically relevant novel forms of oncoprotein and non-oncoprotein addiction or synthetic lethality. The identification of genotype-drug response relationships will further advance personalized oncology and improve drug efficacy in the clinic. Finally, we review the most important drugs targeting ErbB family members that are under investigation in clinical trials or that made their way already into clinical routine. Taken together, the functional characterization of ErbB oncoproteins have significantly increased our knowledge on predictive biomarkers, oncoprotein addiction and patient stratification and treatment.
Collapse
|
95
|
Özdemir BC, Dotto GP. Racial Differences in Cancer Susceptibility and Survival: More Than the Color of the Skin? Trends Cancer 2017; 3:181-197. [PMID: 28718431 DOI: 10.1016/j.trecan.2017.02.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
Abstract
Epidemiological studies point to race as a determining factor in cancer susceptibility. In US registries recording cancer incidence and survival by race (distinguishing 'black versus white'), individuals of African ancestry have a globally increased risk of malignancies compared with Caucasians and Asian Americans. Differences in socioeconomic status and health-care access play a key role. However, the lesser disease susceptibility of Hispanic populations with comparable lifestyles and socioeconomic status as African Americans (Hispanic paradox) points to the concomitant importance of genetic determinants. Here, we overview the molecular basis of racial disparity in cancer susceptibility ranging from genetic polymorphisms and cancer-driver gene mutations to obesity, chronic inflammation, and immune responses. We discuss implications for race-adapted cancer screening programs and clinical trials to reduce disparities in cancer burden.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Gian-Paolo Dotto
- Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Épalinges, Switzerland; Harvard Dermatology Department and Cutaneous Biology Research Center, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02129, USA.
| |
Collapse
|
96
|
Corona SP, Ravelli A, Cretella D, Cappelletti MR, Zanotti L, Dester M, Gobbi A, Petronini PG, Generali D. CDK4/6 inhibitors in HER2-positive breast cancer. Crit Rev Oncol Hematol 2017; 112:208-214. [PMID: 28325261 DOI: 10.1016/j.critrevonc.2017.02.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 10/20/2022] Open
Abstract
Notwithstanding the continuous progress made in cancer treatment in the last 20 years, and the availability of new targeted therapies, metastatic Breast Cancer (BC) is still incurable. Targeting the cell cycle machinery has emerged as an attractive strategy to tackle cancer progression, showing very promising results in the preclinical and clinical settings. The first selective inhibitors of CDK4/6 received breakthrough status and FDA approval in combination with letrozole (February 2015) and fulvestrant (February 2016) as first-line therapy in ER-positive advanced and metastatic BC. Considering the success of this family of compounds in hormone-positive BC, new possible applications are being investigated in other molecular subtypes. This review summarizes the latest findings on the use of CDK4/6 inhibitors in HER2 positive BC.
Collapse
Affiliation(s)
- Silvia Paola Corona
- Peter MacCallum Cancer Centre, Radiation Oncology Department, Moorabbin Campus, East Bentleigh Victoria 3165, Australia.
| | - Andrea Ravelli
- Universita degli Studi di Parma, Department of Clinical and Experimental Medicine, Experimental Oncology Unit, Via Gramsci, 14, Parma, Italy
| | - Daniele Cretella
- Universita degli Studi di Parma, Department of Clinical and Experimental Medicine, Experimental Oncology Unit, Via Gramsci, 14, Parma, Italy
| | - Maria Rosa Cappelletti
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Laura Zanotti
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Martina Dester
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Angela Gobbi
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Pier Giorgio Petronini
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Daniele Generali
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy; Universita degli Studi di Trieste, Department of Medical, Surgery and Health Sciences, Trieste, Italy
| |
Collapse
|
97
|
Shibamoto M, Hirata H, Eguchi H, Sawada G, Sakai N, Kajiyama Y, Mimori K. The loss of CASP4 expression is associated with poor prognosis in esophageal squamous cell carcinoma. Oncol Lett 2017; 13:1761-1766. [PMID: 28454321 DOI: 10.3892/ol.2017.5646] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/12/2016] [Indexed: 12/19/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) has high biological malignant potential among the various digestive tract cancers and is associated with a poor prognosis. To identify novel genes involved in tumor progression, the present study analyzed the genetic and transcriptional alterations in two clinical cohorts, totaling 157 cases of ESCC (78 cases from the discovery set and 79 cases from the validation set). From the discovery set, gene expression and copy number profiles were analyzed using expression arrays and array-comparative genomic hybridization, respectively. Notably, a copy number loss of caspase-4 (CASP4) was observed in 82% of ESCC cases and CASP4 expression levels were significantly associated with copy number levels. Gene set enrichment analysis demonstrated that the upregulation of CASP4 expression levels was associated with the signaling pathways involved in apoptosis, inflammatory responses and immune responses. The present study demonstrated that CASP4 expression levels were significantly associated with the expression levels of the endoplasmic reticulum (ER) stress marker glucose-regulated protein 78, indicating that CASP4 has a role in cell death induced by ER stress in ESCC. In the survival analysis the CASP4 low expression group exhibited a poor prognosis, compared with the CASP4 high expression group in the discovery set (P=0.003); this observation was reproduced in the validation set (P=0.037). Therefore, the results of the current study suggest that CASP4 may function as a tumor-suppressor gene and may have applications as a biomarker for the prediction of the prognosis in ESCC.
Collapse
Affiliation(s)
- Misako Shibamoto
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Hidenari Hirata
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Oita 874-0838, Japan.,Department of Radiology, Kyushu University Beppu Hospital, Beppu, Oita 874-0838, Japan
| | - Hidetoshi Eguchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Oita 874-0838, Japan
| | - Genta Sawada
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Oita 874-0838, Japan
| | - Noritaka Sakai
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Yoshiaki Kajiyama
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo 113-8431, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Oita 874-0838, Japan
| |
Collapse
|
98
|
Sicklick JK, Fanta PT, Shimabukuro K, Kurzrock R. Genomics of gallbladder cancer: the case for biomarker-driven clinical trial design. Cancer Metastasis Rev 2017; 35:263-75. [PMID: 26857926 DOI: 10.1007/s10555-016-9602-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Gallbladder carcinoma is a rare, aggressive malignancy of the biliary tract associated with a poor prognosis. Despite the deployment of targeted therapies that have demonstrated marked survival benefits in many tumor types, traditional cytotoxic chemotherapy has remained the mainstay of treatment for unresectable and metastatic gallbladder cancer. METHODS Systematic review of ongoing and prior clinical studies shows a paucity of biomarker-driven therapeutic trials using targeted agents in gallbladder cancer. In fact, over the past 6 years, of the 38 therapeutic biliary tract protocols listed on clinicaltrials.gov, only 6 (21 %) utilized targeted therapies based upon tumor biomarkers or genomics. Now that we have entered the era of next-generation sequencing and precision medicine, we are beginning to identify common and specific genetic alterations in gallbladder carcinomas. RESULTS A review of the literature reveals alterations in ARID1A, BRAF, CDKN2A/B, EGFR, ERBB2-4, HKN-RAS, PIK3CA, PBRM1, and TP53. Given the widespread use of tumor genomic profiling and the fact that most of the aforementioned alterations are pharmacologically tractable, these observations suggest the potential for new therapeutic strategies in this aggressive malignancy. CONCLUSIONS Taken together, further understanding of the genomic landscape of gallbladder cancer coupled with biomarker-driven clinical trials that match therapies to targets are urgently needed.
Collapse
Affiliation(s)
- Jason K Sicklick
- Center for Personalized Cancer Therapy, San Diego, CA, USA. .,Division of Surgical Oncology, Department of Surgery, University of California, San Diego Moores Cancer Center, 3855 Health Sciences Drive, MC 0987, La Jolla, CA, 92093-0987, USA.
| | - Paul T Fanta
- Center for Personalized Cancer Therapy, San Diego, CA, USA.,Division of Hematology and Oncology, University of California, San Diego Moores Cancer Center, 3855 Health Sciences Drive, MC 0987, La Jolla, 92093-0987, CA, USA
| | - Kelly Shimabukuro
- Center for Personalized Cancer Therapy, San Diego, CA, USA.,Division of Hematology and Oncology, University of California, San Diego Moores Cancer Center, 3855 Health Sciences Drive, MC 0987, La Jolla, 92093-0987, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, San Diego, CA, USA.,Division of Hematology and Oncology, University of California, San Diego Moores Cancer Center, 3855 Health Sciences Drive, MC 0987, La Jolla, 92093-0987, CA, USA
| |
Collapse
|
99
|
Martinho O, Silva-Oliveira R, Cury FP, Barbosa AM, Granja S, Evangelista AF, Marques F, Miranda-Gonçalves V, Cardoso-Carneiro D, de Paula FE, Zanon M, Scapulatempo-Neto C, Moreira MA, Baltazar F, Longatto-Filho A, Reis RM. HER Family Receptors are Important Theranostic Biomarkers for Cervical Cancer: Blocking Glucose Metabolism Enhances the Therapeutic Effect of HER Inhibitors. Theranostics 2017; 7:717-732. [PMID: 28255362 PMCID: PMC5327645 DOI: 10.7150/thno.17154] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/21/2016] [Indexed: 12/17/2022] Open
Abstract
Persistent HPV infection alone is not sufficient for cervical cancer development, which requires additional molecular alterations for tumor progression and metastasis ultimately leading to a lethal disease. In this study, we performed a comprehensive analysis of HER family receptor alterations in cervical adenocarcinoma. We detected overexpression of HER protein, mainly HER2, which was an independent prognostic marker for these patients. By using in vitro and in vivo approaches, we provided evidence that HER inhibitors, allitinib and lapatinib, were effective in reducing cervical cancer aggressiveness. Furthermore, combination of these drugs with glucose uptake blockers could overcome the putative HIF1-α-mediated resistance to HER-targeted therapies. Thus, we propose that the use of HER inhibitors in association with glycolysis blockers can be a potentially effective treatment option for HER-positive cervical cancer patients.
Collapse
Affiliation(s)
- Olga Martinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Renato Silva-Oliveira
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Fernanda P. Cury
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Ana Martins Barbosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Fábio Marques
- Department of Pathology of the School of Medicine of the Federal University of Goiás, Brazil
| | - Vera Miranda-Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Diana Cardoso-Carneiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Flávia E. de Paula
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Maicon Zanon
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | | | - Marise A.R. Moreira
- Department of Pathology of the School of Medicine of the Federal University of Goiás, Brazil
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adhemar Longatto-Filho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos, São Paulo, Brazil
- Laboratory of Medical Investigation (LIM) 14, Faculty of Medicine, São Paulo State University, Brazil
| | - Rui Manuel Reis
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| |
Collapse
|
100
|
Dahlhoff M, Muzumdar S, Schäfer M, Schneider MR. ERBB2 Is Essential for the Growth of Chemically Induced Skin Tumors in Mice. J Invest Dermatol 2016; 137:921-930. [PMID: 27931797 DOI: 10.1016/j.jid.2016.11.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 10/31/2016] [Accepted: 11/08/2016] [Indexed: 12/23/2022]
Abstract
Although the epidermal growth factor receptor has established roles in skin carcinogenesis, inflammation, and wound healing, the functions of the structurally related receptor ERBB2 in this tissue remain poorly explored. To assess the functions of ERBB2 in skin homeostasis, tumorigenesis, and wound healing, we employed keratin 5-directed, cre recombinase-mediated targeting of Erbb2 alleles in mice. Erbb2del mice, lacking ERBB2 specifically in keratinocytes, showed no noticeable spontaneous skin abnormalities. During early wound healing, the thickness and the number and proliferation rate of keratinocytes in the wound epithelium of Erbb2del mice were significantly reduced. Compared with control littermates, Erbb2del mice remained free of papillomas for a longer time and had significantly reduced tumor burden after application of the 7,12-dimethylbenz[a]anthracene/12-O-tetradecanoylphorbol-13-acetate multistage chemical carcinogenesis protocol. Furthermore, tumor cell proliferation was substantially reduced in Erbb2del mice, and loss of ERBB2 also decreased keratinocyte proliferation after 12-O-tetradecanoylphorbol-13-acetate application. Thus, ERBB2 is dispensable for the development and homeostasis of the epidermis and its appendages. However, reflecting its pro-proliferative role, ERBB2 is required for the normal healing of skin wounds and for the progression of tumors during skin chemical carcinogenesis in mice. Thus, ERBB2 may be a promising target for inhibiting human nonmelanoma skin cancer progression.
Collapse
Affiliation(s)
- Maik Dahlhoff
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Sukalp Muzumdar
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Matthias Schäfer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Marlon R Schneider
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany.
| |
Collapse
|