1
|
Li Y, Zhan W, Jia Y, Xiong H, Lin B, Li Q, Liu H, Qiu L, Zhang Y, Ding J, Fu C, Chen W. Application of tangent-arc technology for deep inspiration breath-hold radiotherapy in left-sided breast cancer. Front Oncol 2023; 13:1145332. [PMID: 37795446 PMCID: PMC10547143 DOI: 10.3389/fonc.2023.1145332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/31/2023] [Indexed: 10/06/2023] Open
Abstract
Objective To explore the advantages of dosimetry and the treatment efficiency of tangent-arc technology in deep inspiration breath-hold radiotherapy for breast cancer. Methods Forty patients with left-sided breast cancer who were treated in our hospital from May 2020 to June 2021 were randomly selected and divided into two groups. The first group's plan was a continuous semi-arc that started at 145° ( ± 5°) and stopped at 325° ( ± 5°). The other group's plan, defined as the tangent-arc plan, had two arcs: the first arc started at 145° ( ± 5°) and stopped at 85° ( ± 5°), and the second arc started at 25° ( ± 5°) and stopped at 325° ( ± 5°). We compared the target dose, dose in organs at risk (OARs), and treatment time between the two groups. Results The target dose was similar between the continuous semiarc and tangent-arc groups. The V5 of the right lung was significantly different between the two groups (Dif 5.52, 95% confidence interval 1.92-9.13, t=3.10, P=0.004), with the patients in the continuous semi-arc and tangent-arc groups having lung V5 values of (9.16 ± 1.62)%, and (3.64 ± 0.73)%, respectively. The maximum dose to the spinal cord was (1835.88 ± 222.17) cGy in the continuous semi-arc group and (599.42 ± 153.91) cGy in the tangent-arc group, yielding a significant difference between the two groups (Dif 1236.46, 95% confidence interval 689.32-1783.6, t=4.57, P<0.001). The treatment times was (311.70 ± 60.45) s for patients in the continuous semi-arc group and (254.66 ± 40.73) s for patients in the tangent-arc group, and there was a significant difference in the mean number of treatment times between the two groups (Dif 57.04, 95% confidence interval 24.05-90.03, t=3.5, P=0.001). Conclusion Both the continuous semi-arc and tangent-arc plans met the clinical prescription dose requirements. The OARs received less radiation with the tangent-arc plan than the continuous semi-arc plan, especially for the lung (measured as V5) and the spinal cord (measured as the maximum dose). Tangent-arc plan took significantly less time than the continuous semi-arc, which can greatly improve treatment efficiency. Therefore, tangent-arc plans are superior continuous semi-arc plans for all cases.
Collapse
Affiliation(s)
- Yucheng Li
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenming Zhan
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yongshi Jia
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hanchu Xiong
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Baihua Lin
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiang Li
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huaxin Liu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lingyun Qiu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yinghao Zhang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jieni Ding
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chao Fu
- Department of Tumor Radiochemotherapy, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijun Chen
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Shimozaki K, Shinozaki E, Yamamoto N, Imamura Y, Osumi H, Nakayama I, Wakatsuki T, Ooki A, Takahari D, Ogura M, Chin K, Watanabe M, Yamaguchi K. KRAS mutation as a predictor of insufficient trastuzumab efficacy and poor prognosis in HER2-positive advanced gastric cancer. J Cancer Res Clin Oncol 2023; 149:1273-1283. [PMID: 35438321 DOI: 10.1007/s00432-022-03966-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/21/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Although RAS and PIK3CA mutations have been associated with resistance to anti-EGFR antibody in colorectal cancer or trastuzumab in breast cancer, their implications for trastuzumab resistance in HER2-positive advanced gastric cancer (AGC) remains unclear. We aimed to assess the relationship between trastuzumab efficacy and mutation status in the HER family signaling pathway. METHODS This study retrospectively evaluated patients with HER2-positive AGC who received first-line trastuzumab-containing chemotherapy between March 2011 and November 2015. Multiplex genotyping, including KRAS, NRAS, PIK3CA, and BRAF, was then performed using the Luminex Assay, after which KRAS amplification was measured using quantitative real-time reverse transcription-polymerase chain reaction. Thereafter, the association between genetic alterations and clinical outcomes were evaluated. RESULTS KRAS mutation (MT) was detected in 6 of 77 patients (7.8%), whereas KRAS amplification was found in 15 of 67 patients (22%). No mutations in NRAS, PIK3CA, or BRAF were identified. The KRAS MT group showed significantly worse response rates (16.7% vs. 66.2%, P = 0.016), progression-free survival [median, 4.8 vs. 11.6 months; hazard ratio (HR), 3.95; 95% CI, 1.60-9.76; P = 0.0029], and overall survival (11.5 vs. 23.6 months; HR, 3.80; 95% CI, 1.56-9.28; P = 0.033) compared to the KRAS wild-type group. KRAS amplification had no effect on clinical outcomes. CONCLUSION KRAS mutation was an independent prognostic factor for poor survival and might predict insufficient trastuzumab efficacy, whereas KRAS amplification showed no prognostic significance during trastuzumab treatment. Further investigations are warranted to confirm the predictive value of KRAS status in HER2-positive AGC.
Collapse
Affiliation(s)
- Keitaro Shimozaki
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan.,Department of Gastroenterology and Hepatology, Division of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Eiji Shinozaki
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan.
| | - Noriko Yamamoto
- Department of Pathology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yu Imamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroki Osumi
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| | - Izuma Nakayama
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| | - Takeru Wakatsuki
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| | - Akira Ooki
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| | - Daisuke Takahari
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| | - Mariko Ogura
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| | - Keisho Chin
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensei Yamaguchi
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake 3-8-3, Koto-ku, Tokyo, Japan
| |
Collapse
|
3
|
Sankaranarayanan RA, Peil J, Vogg ATJ, Bolm C, Terhorst S, Classen A, Bauwens M, Maurer J, Mottaghy F, Morgenroth A. Auger Emitter Conjugated PARP Inhibitor for Therapy in Triple Negative Breast Cancers: A Comparative In-Vitro Study. Cancers (Basel) 2022; 14:cancers14010230. [PMID: 35008392 PMCID: PMC8750932 DOI: 10.3390/cancers14010230] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/20/2021] [Accepted: 12/30/2021] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer, with a high recurrence rate. Since treatment of BRCAmut TNBC patients with PARP inhibitor (PARPi), targeting the nuclear protein PARP1, shows varied responses, its therapeutic efficacy is currently evaluated in combination with chemotherapy. Auger emitters (AEs) are radionuclides that can cause DNA damage when delivered close to the DNA. Due to the nuclear location of PARP1, radiolabelling of PARPi with AEs provide an efficient nuclear delivery mechanism. This study shows the radiosynthesis of an AE radiolabelled PARPi ([125I]-PARPi-01) and its therapeutic effect as monotherapy or in combination with chemotherapeutics in a panel of TNBC cell lines. We found that [125I]-PARPi-01 efficiently induces DNA damage with therapeutic effect irrespective of BRCA mutation. All responsive cell lines have homologous recombination deficiency. Short pretreatment with doxorubicin significantly reduces clonogenic survival of both responsive and resistant cell lines. Abstract PARP1 inhibitors (PARPi) are currently approved for BRCAmut metastatic breast cancer, but they have shown limited response in triple negative breast cancer (TNBC) patients. Combination of an Auger emitter with PARPis enables PARP inhibition and DNA strand break induction simultaneously. This will enhance cytotoxicity and additionally allow a theranostic approach. This study presents the radiosynthesis of the Auger emitter [125I] coupled olaparib derivative: [125I]-PARPi-01, and its therapeutic evaluation in a panel of TNBC cell lines. Specificity was tested by a blocking assay. DNA strand break induction was analysed by γH2AX immunofluorescence staining. Cell cycle analysis and apoptosis assays were studied using flow cytometry in TNBC cell lines (BRCAwt/mut). Anchorage independent growth potential was evaluated using soft agar assay. [125I]-PARPi-01 showed PARP1-specificity and higher cytotoxicity than olaparib in TNBC cell lines irrespective of BRCA their status. Cell lines harbouring DNA repair deficiency showed response to [125I]-PARPi-01 monotherapy. Combined treatment with Dox-NP further enhanced therapeutic efficiency in metastatic resistant BRCAwt cell lines. The clonogenic survival was significantly reduced after treatment with [125I]-PARPi-01 in all TNBC lines investigated. Therapeutic efficacy was further enhanced after combined treatment with chemotherapeutics. [125I]-PARPi-01 is a promising radiotherapeutic agent for low radiation dosages, and mono/combined therapies of TNBC.
Collapse
Affiliation(s)
- Ramya Ambur Sankaranarayanan
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (R.A.S.); (J.P.); (A.T.J.V.); (M.B.); (F.M.)
| | - Jennifer Peil
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (R.A.S.); (J.P.); (A.T.J.V.); (M.B.); (F.M.)
| | - Andreas T. J. Vogg
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (R.A.S.); (J.P.); (A.T.J.V.); (M.B.); (F.M.)
| | - Carsten Bolm
- Institute of Organic Chemistry, RWTH Aachen University, 52056 Aachen, Germany; (C.B.); (S.T.); (A.C.)
| | - Steven Terhorst
- Institute of Organic Chemistry, RWTH Aachen University, 52056 Aachen, Germany; (C.B.); (S.T.); (A.C.)
| | - Arno Classen
- Institute of Organic Chemistry, RWTH Aachen University, 52056 Aachen, Germany; (C.B.); (S.T.); (A.C.)
| | - Matthias Bauwens
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (R.A.S.); (J.P.); (A.T.J.V.); (M.B.); (F.M.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6229HX Maastricht, The Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229HX Maastricht, The Netherlands
| | - Jochen Maurer
- Department of Molecular Gynecology, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany;
| | - Felix Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (R.A.S.); (J.P.); (A.T.J.V.); (M.B.); (F.M.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6229HX Maastricht, The Netherlands
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (R.A.S.); (J.P.); (A.T.J.V.); (M.B.); (F.M.)
- Correspondence:
| |
Collapse
|
4
|
Yan Y, Cheng X, Li L, Zhang R, Zhu Y, Wu Z, Ding K. A Novel Small Molecular Antibody, HER2-Nanobody, Inhibits Tumor Proliferation in HER2-Positive Breast Cancer Cells In Vitro and In Vivo. Front Oncol 2021; 11:669393. [PMID: 34055637 PMCID: PMC8149955 DOI: 10.3389/fonc.2021.669393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/26/2021] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is the most common malignant cancer in women worldwide, especially in developing countries. Herceptin is a monoclonal antibody with an antitumor effect in HER2-positive breast cancer. However, the large molecular weight of Herceptin limited its employment. In this study, we constructed and screened HER2-nanobody and verified its tumor-suppressive effect in HER2-positive breast cancer cells. HER2-nanobody was established, filtrated, purified, and was demonstrated to inhibit cell total number, viability, colony formation and mitosis, and promote cell apoptosis in HER2-positive breast cancer cells in vitro. Treated with HER2-nanobody, tumor growth was significantly inhibited by both intratumor injection and tail intravenous injection in vivo. The phosphorylation of ERK and AKT was restrained by HER2-nanobody in HER2-positive breast cancer cells. RAS-RAF-MAPK and PI3K-AKT-mTOR are two important pathways involved in HER2. It was credible for HER2-nanobody to play the tumor suppressive role by inhibiting the phosphorylation of ERK and AKT. Therefore, HER2-nanobody could be employed as a small molecular antibody to suppress HER2-positive breast cancer.
Collapse
Affiliation(s)
- Yan Yan
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, China.,Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, China
| | - Xiao Cheng
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Lin Li
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Rumeng Zhang
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Yong Zhu
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Zhengsheng Wu
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, China.,Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Keshuo Ding
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, China.,Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Trastuzumab immunogenicity development in patients' sera and in laboratory animals. BMC Immunol 2021; 22:15. [PMID: 33607941 PMCID: PMC7893875 DOI: 10.1186/s12865-021-00405-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Background Immunogenicity is a major challenge in drug development and patient care. Clinicians and regulators are familiar with immunogenicity concerns of monoclonal antibody (mAb) therapeutics, growth factors and enzyme replacements. Although most small therapeutic molecules are unlikely to trigger undesirable immunogenic responses against themselves upon their administration, the biological therapeutic agents are likely to induce such kind of immunogenicity. This imparts a problem that has to be considered upon judging their risk–benefit ratio. In this article, we tested the immunogenicity developed in patients’ sera due to the use of trastuzumab and that developed in laboratory animals injected with this recombinant humanized IgG1 monoclonal antibody. Methods We studied trastuzumab immunogenicity by: I in vitro detection of anti-trastuzumab antibody (Ab) levels in patient’s serum samples withdrawn at different points during trastuzumab treatment course; I.1 using an Affinity Capture Elution (ACE) assay, the assay is both sensitive and highly tolerant to free drug; I.2 using MTT cytotoxicity method against MCF-7 cell line as confirmatory method used in sample showed high level of anti-trastuzumab Ab and to determine neutralizing activity of the anti-trastuzumab Ab. II in vivo immunogenicity testing of trastuzumab in lab animals. Results In vitro analysis of patients’ sera for antibodies developed against trastuzumab revealed that this monoclonal antibody has low immunogenicity since most samples showed low levels of anti-trastuzumab antibodies that decreased progressively along the treatment course. Only 1% of samples showed high levels of anti-trastuzumab antibodies which might affect treatment course. In vivo immunogenicity testing in mice showed also low immunogenicity of trastuzumab that could support the in vitro clinical assessment applied in our study. Conclusions The study gives an evidence for the low trastuzumab immunogenicity when assessed in Egyptian patients under treatment with this biological therapeutic agent. This supports its prescription and continuous use across the approved indications as biological therapeutic agent.
Collapse
|
6
|
Targeted inhibition of cooperative mutation- and therapy-induced AKT activation in AML effectively enhances response to chemotherapy. Leukemia 2020; 35:2030-2042. [PMID: 33299144 DOI: 10.1038/s41375-020-01094-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/30/2020] [Accepted: 11/09/2020] [Indexed: 11/09/2022]
Abstract
Most AML patients exhibit mutational activation of the PI3K/AKT signaling pathway, which promotes downstream effects including growth, survival, DNA repair, and resistance to chemotherapy. Herein we demonstrate that the inv(16)/KITD816Y AML mouse model exhibits constitutive activation of PI3K/AKT signaling, which was enhanced by chemotherapy-induced DNA damage through DNA-PK-dependent AKT phosphorylation. Strikingly, inhibitors of either PI3K or DNA-PK markedly reduced chemotherapy-induced AKT phosphorylation and signaling leading to increased DNA damage and apoptosis of inv(16)/KITD816Y AML cells in response to chemotherapy. Consistently, combinations of chemotherapy and PI3K or DNA-PK inhibitors synergistically inhibited growth and survival of clonogenic AML cells without substantially inhibiting normal clonogenic bone marrow cells. Moreover, treatment of inv(16)/KITD816Y AML mice with combinations of chemotherapy and PI3K or DNA-PK inhibitors significantly prolonged survival compared to untreated/single-treated mice. Mechanistically, our findings implicate that constitutive activation of PI3K/AKT signaling driven by mutant KIT, and potentially other mutational activators such as FLT3 and RAS, cooperates with chemotherapy-induced DNA-PK-dependent activation of AKT to promote survival, DNA repair, and chemotherapy resistance in AML. Hence, our study provides a rationale to select AML patients exhibiting constitutive PI3K/AKT activation for simultaneous treatment with chemotherapy and inhibitors of DNA-PK and PI3K to improve chemotherapy response and clinical outcome.
Collapse
|
7
|
Wen X, Ou YC, Bogatcheva G, Thomas G, Mahadevan-Jansen A, Singh B, Lin EC, Bardhan R. Probing metabolic alterations in breast cancer in response to molecular inhibitors with Raman spectroscopy and validated with mass spectrometry. Chem Sci 2020; 11:9863-9874. [PMID: 34094246 PMCID: PMC8162119 DOI: 10.1039/d0sc02221g] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/19/2020] [Indexed: 01/07/2023] Open
Abstract
Rapid and accurate response to targeted therapies is critical to differentiate tumors that are resistant to treatment early in the regimen. In this work, we demonstrate a rapid, noninvasive, and label-free approach to evaluate treatment response to molecular inhibitors in breast cancer (BC) cells with Raman spectroscopy (RS). Metabolic reprogramming in BC was probed with RS and multivariate analysis was applied to classify the cells into responsive or nonresponsive groups as a function of drug dosage, drug type, and cell type. Metabolites identified with RS were then validated with mass spectrometry (MS). We treated triple-negative BC cells with Trametinib, an inhibitor of the extracellular-signal-regulated kinase (ERK) pathway. Changes measured with both RS and MS corresponding to membrane phospholipids, amino acids, lipids and fatty acids indicated that these BC cells were responsive to treatment. Comparatively, minimal metabolic changes were observed post-treatment with Alpelisib, an inhibitor of the mammalian target of rapamycin (mTOR) pathway, indicating treatment resistance. These findings were corroborated with cell viability assay and immunoblotting. We also showed estrogen receptor-positive MCF-7 cells were nonresponsive to Trametinib with minimal metabolic and viability changes. Our findings support that oncometabolites identified with RS will ultimately enable rapid drug screening in patients ensuring patients receive the most effective treatment at the earliest time point.
Collapse
Affiliation(s)
- Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University Nashville TN 37235 USA
| | - Yu-Chuan Ou
- Department of Chemical and Biomolecular Engineering, Vanderbilt University Nashville TN 37235 USA
| | - Galina Bogatcheva
- Department of Medicine, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - Giju Thomas
- Vanderbilt Biophotonics Center, Vanderbilt University Nashville TN 37232 USA
| | | | - Bhuminder Singh
- Department of Medicine, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - Eugene C Lin
- Department of Chemistry and Biochemistry, National Chung Cheng University Chiayi 62106 Taiwan
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University Ames IA 50012 USA
- Nanovaccine Institute, Iowa State University Ames IA 50012 USA
| |
Collapse
|
8
|
Dirican E. BRAF, KRAS and PIK3CA Mutation and Sensitivity to Trastuzumab in Breast Cancer Cell Line Model. Asian Pac J Cancer Prev 2020; 21:1. [PMID: 31983154 PMCID: PMC7294037 DOI: 10.31557/apjcp.2020.21.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Indexed: 11/25/2022] Open
Affiliation(s)
- Ebubekir Dirican
- Istanbul Aydın University, Vocational School of Health Services, Istanbul, Turkey.,Marmara University, School of Medicine Department of Medical Biology, Istanbul, Turkey
| |
Collapse
|
9
|
Kostrhunova H, Zajac J, Novohradsky V, Kasparkova J, Malina J, Aldrich-Wright JR, Petruzzella E, Sirota R, Gibson D, Brabec V. A Subset of New Platinum Antitumor Agents Kills Cells by a Multimodal Mechanism of Action Also Involving Changes in the Organization of the Microtubule Cytoskeleton. J Med Chem 2019; 62:5176-5190. [PMID: 31030506 DOI: 10.1021/acs.jmedchem.9b00489] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The substitution inert platinum agent [Pt(1 S,2 S-diaminocyclohexane)(5,6-dimethyl-1,10-phenanthroline)]2+ (56MeSS, 5) is a potent cytotoxic metallodrug. In contrast to conventional cisplatin or oxaliplatin, the mechanism of action (MoA) of 5 is fundamentally different. However, details of the mechanism by which the 5,6-dimethyl-1,10-phenanthroline ligand contributes to the cytotoxicity of 5 and its derivatives have not been sufficiently clarified so far. Here, we show that 5 and its Pt(IV) derivatives exhibit an intriguing potency in the triple-negative breast cancer cells MDA-MB-231. Moreover, we show that the Pt(IV) derivatives of 5 act by multimodal MoA resulting in the global biological effects, that is, they damage nuclear DNA, reduce the mitochondrial membrane potential, induce the epigenetic processes, and last but not least, the data provide evidence that changes in the organization of cytoskeleton networks are functionally important for 5 and its derivatives, in contrast to clinically used platinum cytostatics, to kill cancer cells.
Collapse
Affiliation(s)
- Hana Kostrhunova
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Juraj Zajac
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Jaroslav Malina
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Janice R Aldrich-Wright
- School of Science and Health , Western Sydney University , Penrith South DC 1797 , NSW , Australia
| | - Emanuele Petruzzella
- Institute for Drug Research, School of Pharmacy , The Hebrew University , Jerusalem 91120 , Israel
| | - Roman Sirota
- Institute for Drug Research, School of Pharmacy , The Hebrew University , Jerusalem 91120 , Israel
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy , The Hebrew University , Jerusalem 91120 , Israel
| | - Viktor Brabec
- Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| |
Collapse
|
10
|
Liang X, Xin X, Qi D, Fu C, Ding M. Silencing the PIK3CA Gene Enhances the Sensitivity of Childhood Leukemia Cells to Chemotherapy Drugs by Suppressing the Phosphorylation of Akt. Yonsei Med J 2019; 60:182-190. [PMID: 30666840 PMCID: PMC6342719 DOI: 10.3349/ymj.2019.60.2.182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 12/16/2022] Open
Abstract
PURPOSE This study aimed to investigate the effects of PIK3CA on the sensitivity of acute B lymphocytic leukemia cells (Nalm-6 cells) to chemotherapy drugs. MATERIALS AND METHODS Children's normal B lymphocytes and Nalm-6 cells were cultured. Nalm-6 cells were transfected with PIK3CA siRNA (siPIK3CA group) or its negative control (PIK3CA-Control group). Normal Nalm-6 cells were named Mock group. Nalm-6 cells transfected by PIK3CA siRNA were treated with Akt inhibitor (siPIK3CA+Akti-1/2 group). mRNA and protein expression was detected by qRT-PCR and Western blot. Proliferation and sensitivity to chemotherapeutic drugs was detected by MTT assay. Cell cycle and apoptosis was explored by low cytometry. Transwell assay was performed to test invasion. RESULTS PIK3CA mRNA (p=0.008) and protein (p=0.006) expression was higher in Nalm-6 cells than that in normal B lymphocytes. Compared with the Mock group and PIK3CA-Control group, Nalm-6 cells of the siPIK3CA group had lower OD495 values (all p<0.05) and invasion cell numbers (p=0.03 and p=0.025), as well as a higher proportion of G0/G1 phase cells (p=0.020 and p=0.022), percentage of apoptosis (p=0.016 and p=0.022), and inhibition rate (all p<0.05). pAkt expression in the siPIK3CA group (p=0.026 and p=0.031) and siPIK3CA+Akti-1/2 group (p=0.019 and p=0.023) was lower than that in the Mock group. CONCLUSION PIK3CA silencing inhibited Nalm-6 cell proliferation and invasion, and promoted their apoptosis and sensitivity to chemotherapeutic drugs, potentially through regulation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xiuling Liang
- Department of Pediatric Internal Medicine, Affiliated Hospital of Taishan Medical University, Tai'an, China
| | - Xianfang Xin
- Department of Pediatric Internal Medicine, Affiliated Hospital of Taishan Medical University, Tai'an, China
| | - Dongmei Qi
- Department of Pediatric Internal Medicine, Affiliated Hospital of Taishan Medical University, Tai'an, China
| | - Chengyan Fu
- Department of Pediatric Internal Medicine, Affiliated Hospital of Taishan Medical University, Tai'an, China
| | - Mingde Ding
- Department of Gynecology, Affiliated Hospital of Taishan Medical University, Tai'an, China.
| |
Collapse
|
11
|
Ding K, Wu Z, Li X, Sheng Y, Wang X, Tan S. LMO4 mediates trastuzumab resistance in HER2 positive breast cancer cells. Am J Cancer Res 2018; 8:594-609. [PMID: 29736306 PMCID: PMC5934551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 03/04/2018] [Indexed: 06/08/2023] Open
Abstract
Breast cancer is the leading cause of cancer-related mortality in women worldwide. Trastuzumab (Herceptin) is an effective antibody drug for HER2 positive breast cancer; de novo or acquired trastuzumab resistance retarded the use of trastuzumab for at least 70% of HER2 positive breast cancers. In this study, we reported LMO4 (a member of LIM-only proteins) promoted trastuzumab resistance in human breast cancer cells. Over-expression of LMO4 was observed in acquired trastuzumab resistance breast cancer cells SKBR3 HR and BT474 HR. Depletion of LMO4 partly abolished the trastuzumab resistance of SKBR3 HR and BT474 HR cells. Forced expression of LMO4 significantly increased trastuzumab resistance of HER2 positive breast cancer cells both in vitro and in vivo. BCL-2 was regulated by LMO4 and mediated the promoting role of LMO4 in trastuzumab resistance of HER2 positive breast cancer cells. High level of LMO4 was associated with worse clinicopathological parameters (including tumor size and histological grade) and lower survival rate in HER2 positive breast cancer patients. LMO4 therefore could be used as a target to develop diagnostic and therapeutic methods for human HER2 positive breast cancer.
Collapse
Affiliation(s)
- Keshuo Ding
- Department of Pathology, Anhui Medical UniversityHefei, Anhui, P. R. China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical UniversityHefei, Anhui, P. R. China
| | - Xiaocan Li
- Department of Pathology, The Second Hospital of Anhui Medical UniversityHefei, Anhui, P. R. China
| | - Youjing Sheng
- Department of Pathology, Anhui Medical UniversityHefei, Anhui, P. R. China
| | - Xiaonan Wang
- Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical UniversityHefei, Anhui, P. R. China
| | - Sheng Tan
- Laboratory of Molecular Tumor Pathology, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of ChinaHefei, Anhui, P. R. China
| |
Collapse
|