1
|
Conlon NT, Roche S, Mahdi AF, Browne A, Breen L, Gaubatz J, Meiller J, O'Neill F, O'Driscoll L, Cremona M, Hennessy BT, Eli LD, Crown J, Collins DM. Neratinib plus dasatinib is highly synergistic in HER2-positive breast cancer in vitro and in vivo. Transl Oncol 2024; 49:102073. [PMID: 39191139 PMCID: PMC11396364 DOI: 10.1016/j.tranon.2024.102073] [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: 02/19/2024] [Revised: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND HER2-targeted therapies have revolutionised the treatment of HER2-positive breast cancer. However, de novo resistance or the emergence of acquired resistance is a persistent clinical problem. Here we report that neratinib, an irreversible pan-HER inhibitor, in combination with the multi-kinase inhibitor dasatinib, currently used to treat certain leukemias, has strong anti-proliferative effects against models of HER2-positive breast cancer that are innately resistant to trastuzumab or have acquired resistance to neratinib. METHODS Neratinib plus dasatinib was examined in a panel of 20 breast cancer cell lines, including HER2-positive, estrogen-receptor-positive, triple negative, and acquired HER2-targeted therapy resistant models. Drug effects on migration and apoptosis induction was evaluated and signaling alterations were determined by reverse phase protein array (RPPA). In vivo efficacy was examined using orthotopically-implanted HCC1954 cells. RESULTS Synergy was observed in cell lines innately resistant to trastuzumab, models with acquired resistance to neratinib, and in triple negative breast cancer cell lines. Further investigation showed that neratinib plus dasatinib induced apoptosis and inhibited cell migration to a greater degree than either drug alone. RPPA revealed that the combination caused suppression of key survival signaling through EGFR, Akt, and MAPK inhibition. In vivo, neratinib plus dasatinib was well tolerated and had a prolonged anti-tumor effect against HCC1954 xenografts. CONCLUSIONS This study provides a strong pre-clinical rationale for the clinical investigation neratinib and dasatinib in HER2+ breast cancer.
Collapse
Affiliation(s)
- Neil T Conlon
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Sandra Roche
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Amira F Mahdi
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Alacoque Browne
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Laura Breen
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Johanna Gaubatz
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Justine Meiller
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Fiona O'Neill
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Science & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mattia Cremona
- Molecular Medicine - Laboratory of Molecular Oncology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bryan T Hennessy
- Molecular Medicine - Laboratory of Molecular Oncology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Lisa D Eli
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - John Crown
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland; Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Denis M Collins
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| |
Collapse
|
2
|
Panaampon J, Sungwan P, Fujikawa S, Sampattavanich S, Jirawatnotai S, Okada S. Trastuzumab, a monoclonal anti-HER2 antibody modulates cytotoxicity against cholangiocarcinoma via multiple mechanisms. Int Immunopharmacol 2024; 138:112612. [PMID: 38968862 DOI: 10.1016/j.intimp.2024.112612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024]
Abstract
Cholangiocarcinoma (CCA) is an aggressive and fatal cancer. The prognosis is very poor and no optimal chemotherapy has been established. Human epidermal growth factor receptor 2 (HER2, neu, and erbB2) is highly-expressed in breast cancer and is expressed in many other tumors but poorly expressed in CCA. The anti-HER2 antibody, trastuzumab, has been used for the treatment of HER2-positive breast and gastric cancer. In this study, we examined the surface expression of HER2 on seven Thai liver-fluke-associated CCA cell lines by flow cytometry, and found all of these CCA cells were weakly positive for HER2. MTT assay revealed that trastuzumab directly suppressed the growth of CCA. By using FcR-bearing recombinant Jurkat T-cell-expressing firefly luciferase gene under the control of NFAT response elements, we defined the activities of antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP). ADCC was confirmed by using expanded NK cells. ADCP was confirmed by using mouse peritoneal macrophages and human monocyte-derived macrophages as effector cells. Rabbit serum was administered to test the complement-dependent cytotoxicity (CDC) activity of trastuzumab. Finally, we evaluated the efficacy of trastuzumab in in vivo patient-derived cell xenograft and patient-derived xenograft (PDX) models. Our results showed that a distinct population of CCA (liver-fluke-associated CCA) expressed HER2. Trastuzumab demonstrated a potent inhibitory effect on even HER2 weakly positive CCA both in vitro and in vivo via multiple mechanisms. Thus, HER2 is a promising target in anti-CCA therapy, and trastuzumab can be considered a promising antibody immunotherapy agent for the treatment of CCA.
Collapse
Affiliation(s)
- Jutatip Panaampon
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan; Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Prin Sungwan
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Sawako Fujikawa
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Siwanon Jirawatnotai
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan; Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan.
| |
Collapse
|
3
|
Gao Y, Shelling AN, Nolan E, Porter D, Leung E, Wu Z. Liposome-enabled bufalin and doxorubicin combination therapy for trastuzumab-resistant breast cancer with a focus on cancer stem cells. J Liposome Res 2024; 34:489-506. [PMID: 38269490 DOI: 10.1080/08982104.2024.2305866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024]
Abstract
Breast cancer stem cells (BCSCs) play a key role in therapeutic resistance in breast cancer treatments and disease recurrence. This study aimed to develop a combination therapy loaded with pH-sensitive liposomes to kill both BCSCs and the okbulk cancer cells using trastuzumab-sensitive and resistant human epidermal growth factor receptor 2 positive (HER2+) breast cancer cell models. The anti-BCSCs effect and cytotoxicity of all-trans retinoic acid, salinomycin, and bufalin alone or in combination with doxorubicin were compared in HER2+ cell line BT-474 and a validated trastuzumab-resistant cell line, BT-474R. The most potent anti-BCSC agent was selected and loaded into a pH-sensitive liposome system. The effects of the liposomal combination on BCSCs and bulk cancer cells were assessed. Compared with BT-474, the aldehyde dehydrogenase positive BCSC population was elevated in BT-474R (3.9 vs. 23.1%). Bufalin was the most potent agent and suppressed tumorigenesis of BCSCs by ∼50%, and showed strong synergism with doxorubicin in both BT-474 and BT-474R cell lines. The liposomal combination of bufalin and doxorubicin significantly reduced the BCSC population size by 85%, and inhibited both tumorigenesis and self-renewal, although it had little effect on the migration and invasiveness. The cytotoxicity against the bulk cancer cells was also enhanced by the liposomal combination than either formulation alone in both cell lines (p < 0.001). The liposomal bufalin and doxorubicin combination therapy may effectively target both BCSCs and bulk cancer cells for a better outcome in trastuzumab-resistant HER2+ breast cancer.
Collapse
Affiliation(s)
- Yu Gao
- Faculty of Medical and Health Sciences, School of Pharmacy, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Faculty of Medical and Health Sciences, School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Emma Nolan
- Faculty of Medical and Health Sciences, Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - David Porter
- Auckland Regional Cancer and Blood Service, Auckland City Hospital, Auckland, New Zealand
| | - Euphemia Leung
- Faculty of Medical and Health Sciences, Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Zimei Wu
- Faculty of Medical and Health Sciences, School of Pharmacy, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
4
|
Zhang H, Zhang L, He Y, Jiang D, Sun J, Luo Q, Liang H, Wang T, Li F, Tang Y, Yang Z, Liu W, Rao Y, Chen C. PI3K PROTAC overcomes the lapatinib resistance in PIK3CA-mutant HER2 positive breast cancer. Cancer Lett 2024; 598:217112. [PMID: 38986734 DOI: 10.1016/j.canlet.2024.217112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Although anti-HER2 therapy has made significant strides in reducing metastasis and relapse in HER2-positive breast cancer, resistance to agents like trastuzumab, pertuzumab, and lapatinib frequently develops in patients undergoing treatment. Previous studies suggest that the hyperactivation of the PI3K-AKT signaling pathway by PIK3CA/PTEN gene mutations is implicated in HER2 resistance. In this study, we introduce a novel PI3K-p110α Proteolysis TAargeting Chimera (PROTAC) that effectively inhibits the proliferation of breast cancer cells by degrading PI3K-p110α. When tested in two lapatinib-resistant cell lines, JIMT1 and MDA-MB-453, both of which harbor PIK3CA mutations, the PI3K PROTAC notably reduced cell proliferation and induced G1 phase cell cycle arrest. Importantly, even at very low concentrations, PI3K PROTAC restored sensitivity to lapatinib. Furthermore, the efficacy of PI3K PROTAC surpassed that of Alpelisib, a selective PI3K-p110α kinase inhibitor in clinic. The superior performance of PI3K PROTAC was also confirmed in lapatinib-resistant breast cancer xenograft tumors and patient-derived breast cancer organoids (PDOs). In conclusion, this study reveals that the novel PI3K PROTAC we synthesized could serve as an effective agent to overcome lapatinib resistance.
Collapse
Affiliation(s)
- Hongyan Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China; Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Longlong Zhang
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China
| | - Yuna He
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Dewei Jiang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jian Sun
- The Third Affiliated Hospital, Kunming Medical University, Kunming, 650118, China
| | - Qianmei Luo
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Huichun Liang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Tiantian Wang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; School of Life Science, University of Science & Technology of China, Hefei, 230027, Anhui, China
| | - Fubing Li
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China
| | - Yu Tang
- The Third Affiliated Hospital, Kunming Medical University, Kunming, 650118, China
| | - Zimo Yang
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjing Liu
- The Third Affiliated Hospital, Kunming Medical University, Kunming, 650118, China.
| | - Yu Rao
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Ceshi Chen
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China; The Third Affiliated Hospital, Kunming Medical University, Kunming, 650118, China.
| |
Collapse
|
5
|
Pesce Viglietti AI, Bordignon MB, Ostinelli A, Rizzo MM, Cueto G, Sanchez MB, Perazzo F, Amat M, Coló F, Costanzo MV, Nervo A, Nadal J, Crimi G, Mc Lean I, Spengler EA, Mordoh J, Mandó P, Levy EM. In-Depth Analysis of the Peripheral Immune Profile of HER2+ Breast Cancer Patients on Neoadjuvant Treatment with Chemotherapy Plus Trastuzumab Plus Pertuzumab. Int J Mol Sci 2024; 25:9268. [PMID: 39273217 PMCID: PMC11395157 DOI: 10.3390/ijms25179268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Currently, therapy for early-stage human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC) is based on the combination of trastuzumab and pertuzumab plus chemotherapy in a neoadjuvant regimen. The INMUNOHER study aimed to detect immunological markers in peripheral blood and their association with treatment response. Sixty-two HER2+ BC patients were recruited. Pre-treatment samples were obtained before the start of treatment, while post-treatment samples were obtained after completing therapy and before surgery and were analyzed by flow cytometry. The pathologic complete response (pCR) rate achieved was 82.3%. The expression of the NKp30, PD-1, and TIM-3 receptors was reduced in the Natural Killer (NK)-CD56dim subset of patients who did not achieve pCR. Following therapy, many changes were found in leukocytes, including alterations in T cell lymphocyte proportions. Also, the percentage of NK cells decreased, and several phenotypic changes were observed in this population. After treatment, IFN-γ production by NK cells against HER2+-cells with or without trastuzumab was significantly reduced. HER2-targeted therapy plus chemotherapy demonstrated high efficacy in most patients, reducing the statistical power for finding immunological markers. However, NK subset phenotypes correlated better with response groups, and numerous changes in the percentage of leukocytes and T and NK cells, as well as changes in the functionality of NK cells, were observed in most patients after treatment, encouraging further research into these immune populations.
Collapse
Affiliation(s)
- Ayelén Ivana Pesce Viglietti
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (A.I.P.V.); (M.B.B.)
| | - María Belén Bordignon
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (A.I.P.V.); (M.B.B.)
| | - Alexis Ostinelli
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina
| | - Manglio Miguel Rizzo
- Clinical Oncology Unit, Hospital Universitario Austral, Derqui-Pilar, Buenos Aires B1629ODT, Argentina
| | - Gerardo Cueto
- Grupo de Bioestadística Aplicada, Departamento de Ecología, Genética y Evolución, Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA-UBA/CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1428AOE, Argentina
| | - María Belén Sanchez
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (A.I.P.V.); (M.B.B.)
| | - Florencia Perazzo
- Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Ciudad Autónoma de Buenos Aires C1431AOE, Argentina
| | - Mora Amat
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina
| | - Federico Coló
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina
| | | | - Adrián Nervo
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina
| | - Jorge Nadal
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina
| | - Gabriel Crimi
- Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Ciudad Autónoma de Buenos Aires C1431AOE, Argentina
| | - Ignacio Mc Lean
- Clinical Oncology Unit, Hospital Universitario Austral, Derqui-Pilar, Buenos Aires B1629ODT, Argentina
| | - Eunice Amancay Spengler
- Clinical Oncology Unit, Hospital Universitario Austral, Derqui-Pilar, Buenos Aires B1629ODT, Argentina
| | - José Mordoh
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (A.I.P.V.); (M.B.B.)
| | - Pablo Mandó
- Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Ciudad Autónoma de Buenos Aires C1431AOE, Argentina
| | - Estrella Mariel Levy
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (A.I.P.V.); (M.B.B.)
| |
Collapse
|
6
|
Pham TT, Chenoweth A, Patel N, Banu A, Osborn G, Blower PJ, Karagiannis SN, Ma MT. In Vivo PET Imaging of 89Zr-Labeled Natural Killer Cells and the Modulating Effects of a Therapeutic Antibody. J Nucl Med 2024; 65:1035-1042. [PMID: 38844362 PMCID: PMC11218727 DOI: 10.2967/jnumed.124.267876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/13/2024] [Indexed: 07/03/2024] Open
Abstract
Natural killer (NK) cells can kill cancer cells via antibody-dependent cell-mediated cytotoxicity (ADCC): a tumor-associated IgG antibody binds to the Fcγ receptor CD16 on NK cells via the antibody Fc region and activates the cytotoxic functions of the NK cell. Here, we used PET imaging to assess NK cell migration to human epidermal growth factor receptor 2 (HER2)-positive HCC1954 breast tumors, examining the influence of HER2-targeted trastuzumab antibody treatment on NK cell tumor accumulation. Methods: Human NK cells from healthy donors were expanded ex vivo and labeled with [89Zr]Zr-oxine. In vitro experiments compared the phenotypic markers, viability, proliferation, migration, degranulation, and ADCC behaviors of both labeled (89Zr-NK) and unlabeled NK cells. Female mice bearing orthotopic human breast HCC1954 tumors were administered 89Zr-NK cells alongside trastuzumab treatment or a sham treatment and then scanned using PET/CT imaging over 7 d. Flow cytometry and γ-counting were used to analyze the presence of 89Zr-NK cells in liver and spleen tissues. Results: 89Zr cell radiolabeling yields measured 42.2% ± 8.0%. At an average specific activity of 16.7 ± 4.7 kBq/106 cells, 89Zr-NK cells retained phenotypic and functional characteristics including CD56 and CD16 expression, viability, migration, degranulation, and ADCC capabilities. In vivo PET/CT studies indicated predominant accumulation of 89Zr-NK cells in the liver and spleen. Ex vivo analyses of liver and spleen tissues indicated that the administered human 89Zr-NK cells retained their radioactivity in vivo and that 89Zr did not transfer to cells of murine soft tissues, thus validating this 89Zr PET method for NK cell tracking. Notably, 89Zr-NK cells migrated to HER2-positive tumors, both with and without trastuzumab treatment. Trastuzumab treatment was associated with an increased 89Zr-NK cell signal at days 1 and 3 after injection. Conclusion: In vitro, 89Zr-NK cells maintained key cellular and cytotoxic functions. In vivo, 89Zr-NK cells trafficked to HER2-postive tumors, with trastuzumab treatment correlating with enhanced 89Zr-NK infiltration. This study demonstrates the feasibility of using PET to image 89Zr-NK cell infiltration into solid tumors.
Collapse
Affiliation(s)
- Truc T Pham
- Department of Imaging Chemistry and Biology, School of Bioengineering and Imaging Sciences, King's College London, London, United Kingdom;
| | - Alicia Chenoweth
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom; and
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Natasha Patel
- Department of Imaging Chemistry and Biology, School of Bioengineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Arshiya Banu
- Department of Imaging Chemistry and Biology, School of Bioengineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Gabriel Osborn
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom; and
| | - Philip J Blower
- Department of Imaging Chemistry and Biology, School of Bioengineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom; and
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Michelle T Ma
- Department of Imaging Chemistry and Biology, School of Bioengineering and Imaging Sciences, King's College London, London, United Kingdom;
| |
Collapse
|
7
|
Poskus MD, McDonald J, Laird M, Li R, Norcoss K, Zervantonakis IK. Rational design of HER2-targeted combination therapies to reverse drug resistance in fibroblast-protected HER2+ breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.18.594826. [PMID: 38798591 PMCID: PMC11118562 DOI: 10.1101/2024.05.18.594826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Introduction Fibroblasts, an abundant cell type in the breast tumor microenvironment, interact with cancer cells and orchestrate tumor progression and drug resistance. However, the mechanisms by which fibroblast-derived factors impact drug sensitivity remain poorly understood. Here, we develop rational combination therapies that are informed by proteomic profiling to overcome fibroblast-mediated therapeutic resistance in HER2+ breast cancer cells. Methods Drug sensitivity to the HER2 kinase inhibitor lapatinib was characterized under conditions of monoculture and exposure to breast fibroblast-conditioned medium. Protein expression was measured using reverse phase protein arrays. Candidate targets for combination therapy were identified using differential expression and multivariate regression modeling. Follow-up experiments were performed to evaluate the effects of HER2 kinase combination therapies in fibroblast-protected cancer cell lines and fibroblasts. Results Compared to monoculture, fibroblast-conditioned medium increased the expression of plasminogen activator inhibitor-1 (PAI1) and cell cycle regulator polo like kinase 1 (PLK1) in lapatinib-treated breast cancer cells. Combination therapy of lapatinib with inhibitors targeting either PAI1 or PLK1, eliminated fibroblast-protected cancer cells, under both conditions of direct coculture with fibroblasts and protection by fibroblast-conditioned medium. Analysis of publicly available, clinical transcriptomic datasets revealed that HER2-targeted therapy fails to suppress PLK1 expression in stroma-rich HER2+ breast tumors and that high PAI1 gene expression associates with high stroma density. Furthermore, we showed that an epigenetics-directed approach using a bromodomain and extraterminal inhibitor to globally target fibroblast-induced proteomic adaptions in cancer cells, also restored lapatinib sensitivity. Conclusions Our data-driven framework of proteomic profiling in breast cancer cells identified the proteolytic degradation regulator PAI1 and the cell cycle regulator PLK1 as predictors of fibroblast-mediated treatment resistance. Combination therapies targeting HER2 kinase and these fibroblast-induced signaling adaptations eliminates fibroblast-protected HER2+ breast cancer cells.
Collapse
|
8
|
Bruss C, Albert V, Seitz S, Blaimer S, Kellner K, Pohl F, Ortmann O, Brockhoff G, Wege AK. Neoadjuvant radiotherapy in ER +, HER2 +, and triple-negative -specific breast cancer based humanized tumor mice enhances anti-PD-L1 treatment efficacy. Front Immunol 2024; 15:1355130. [PMID: 38742103 PMCID: PMC11089195 DOI: 10.3389/fimmu.2024.1355130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Pre-operative radiation therapy is not currently integrated into the treatment protocols for breast cancer. However, transforming immunological "cold" breast cancers by neoadjuvant irradiation into their "hot" variants is supposed to elicit an endogenous tumor immune defense and, thus, enhance immunotherapy efficiency. We investigated cellular and immunological effects of sub-lethal, neoadjuvant irradiation of ER pos., HER2 pos., and triple-negative breast cancer subtypes in-vitro and in-vivo in humanized tumor mice (HTM). This mouse model is characterized by a human-like immune system and therefore facilitates detailed analysis of the mechanisms and efficiency of neoadjuvant, irradiation-induced "in-situ vaccination", especially in the context of concurrently applied checkpoint therapy. Similar to clinical appearances, we observed a gradually increased immunogenicity from the luminal over the HER2-pos. to the triple negative subtype in HTM indicated by an increasing immune cell infiltration into the tumor tissue. Anti-PD-L1 therapy divided the HER2-pos. and triple negative HTM groups into responder and non-responder, while the luminal HTMs were basically irresponsive. Irradiation alone was effective in the HER2-pos. and luminal subtype-specific HTM and was supportive for overcoming irresponsiveness to single anti-PD-L1 treatment. The treatment success correlated with a significantly increased T cell proportion and PD-1 expression in the spleen. In all subtype-specific HTM combination therapy proved most effective in diminishing tumor growth, enhancing the immune response, and converted non-responder into responder during anti-PD-L1 therapy. In HTM, neoadjuvant irradiation reinforced anti-PD-L1 checkpoint treatment of breast cancer in a subtype -specific manner. According to the "bench to bedside" principle, this study offers a vital foundation for clinical translating the use of neoadjuvant irradiation in the context of checkpoint therapy.
Collapse
Affiliation(s)
- Christina Bruss
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Veruschka Albert
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Stephanie Blaimer
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Kerstin Kellner
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Fabian Pohl
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Radiotherapy, University Medical Center Regensburg, Regensburg, Germany
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Anja K. Wege
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| |
Collapse
|
9
|
Ke CH, Lin CN, Lin CS. Hormone, Targeted, and Combinational Therapies for Breast Cancers: From Humans to Dogs. Int J Mol Sci 2024; 25:732. [PMID: 38255807 PMCID: PMC10815110 DOI: 10.3390/ijms25020732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Breast cancer (BC) is the most frequent cancer in women. In female dogs, canine mammary gland tumor (CMT) is also the leading neoplasm. Comparative oncology indicates similar tumor behaviors between human BCs (HBCs) and CMTs. Therefore, this review summarizes the current research in hormone and targeted therapies and describes the future prospects for HBCs and CMTs. For hormone receptor-expressing BCs, the first medical intervention is hormone therapy. Monoclonal antibodies against Her2 are proposed for the treatment of Her2+ BCs. However, the major obstacle in hormone therapy or monoclonal antibodies is drug resistance. Therefore, increasing alternatives have been developed to overcome these difficulties. We systemically reviewed publications that reported inhibitors targeting certain molecules in BC cells. The various treatment choices for humans decrease mortality in females with BC. However, the development of hormone or targeted therapies in veterinary medicine is still limited. Even though some clinical trials have been proposed, severe side effects and insufficient case numbers might restrict further explorations. This difficulty highlights the urgent need to develop updated hormone/targeted therapy or novel immunotherapies. Therefore, exploring new therapies to provide more precise use in dogs with CMTs will be the focus of future research. Furthermore, due to the similarities shared by humans and dogs, well-planned prospective clinical trials on the use of combinational or novel immunotherapies in dogs with CMTs to obtain solid results for both humans and dogs can be reasonably anticipated in the future.
Collapse
Affiliation(s)
- Chiao-Hsu Ke
- Sustainable Swine Research Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; (C.-H.K.); (C.-N.L.)
- Animal Disease Diagnostic Center, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Chao-Nan Lin
- Sustainable Swine Research Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; (C.-H.K.); (C.-N.L.)
- Animal Disease Diagnostic Center, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Chen-Si Lin
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
10
|
Sanz-Álvarez M, Luque M, Morales-Gallego M, Cristóbal I, Ramírez-Merino N, Rangel Y, Izarzugaza Y, Eroles P, Albanell J, Madoz-Gúrpide J, Rojo F. Generation and Characterization of Trastuzumab/Pertuzumab-Resistant HER2-Positive Breast Cancer Cell Lines. Int J Mol Sci 2023; 25:207. [PMID: 38203378 PMCID: PMC10779249 DOI: 10.3390/ijms25010207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The combination of trastuzumab and pertuzumab as first-line therapy in patients with HER2-positive breast cancer has shown significant clinical benefits compared to trastuzumab alone. However, despite initial therapeutic success, most patients eventually progress, and tumors develop acquired resistance and invariably relapse. Therefore, there is an urgent need to improve our understanding of the mechanisms governing resistance in order to develop targeted therapeutic strategies with improved efficacy. We generated four novel HER2-positive cell lines via prolonged exposure to trastuzumab and pertuzumab and determined their resistance rates. Long-term resistance was confirmed by a significant increase in the colony-forming capacity of the derived cells. We authenticated the molecular identity of the new lines via both immunohistochemistry for the clinical phenotype and molecular profiling of point mutations. HER2 overexpression was confirmed in all resistant cell lines, and acquisition of resistance to trastuzumab and pertuzumab did not translate into differences in ER, PR, and HER2 receptor expression. In contrast, changes in the expression and activity of other HER family members, particularly HER4, were observed. In the same vein, analyses of the receptor and effector kinase status of different cellular pathways revealed that the MAPK pathway may be involved in the acquisition of resistance to trastuzumab and pertuzumab. Finally, proteomic analysis confirmed a significant change in the abundance patterns of more than 600 proteins with implications in key biological processes, such as ribosome formation, mitochondrial activity, and metabolism, which could be relevant mechanisms in the generation of resistance in HER2-positive breast cancer. We concluded that these resistant BCCLs may be a valuable tool to better understand the mechanisms of acquisition of resistance to trastuzumab and pertuzumab-based anti-HER2 therapy.
Collapse
Affiliation(s)
- Marta Sanz-Álvarez
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Melani Luque
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Miriam Morales-Gallego
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Ion Cristóbal
- Translational Oncology Division, OncoHealth Institute, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain;
| | | | - Yamileth Rangel
- Department of Pathology, Infanta Elena University Hospital, 28342 Madrid, Spain;
| | - Yann Izarzugaza
- Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain;
| | - Pilar Eroles
- Institute of Health Research INCLIVA—CIBERONC, 46010 Valencia, Spain;
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain;
- Department of Medical Oncology, Hospital del Mar—CIBERONC, 08003 Barcelona, Spain
- Department of Experimental and Health Sciences, Faculty of Medicine, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Juan Madoz-Gúrpide
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| |
Collapse
|
11
|
Batalha S, Gomes CM, Brito C. Immune microenvironment dynamics of HER2 overexpressing breast cancer under dual anti-HER2 blockade. Front Immunol 2023; 14:1267621. [PMID: 38022643 PMCID: PMC10643871 DOI: 10.3389/fimmu.2023.1267621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The clinical prognosis of the HER2-overexpressing (HER2-OE) subtype of breast cancer (BC) is influenced by the immune infiltrate of the tumor. Specifically, monocytic cells, which are promoters of pro-tumoral immunosuppression, and NK cells, whose basal cytotoxic function may be enhanced with therapeutic antibodies. One of the standards of care for HER2+ BC patients includes the combination of the anti-HER2 antibodies trastuzumab and pertuzumab. This dual combination was a breakthrough against trastuzumab resistance; however, this regimen does not yield complete clinical benefit for a large fraction of patients. Further therapy refinement is still hampered by the lack of knowledge on the immune mechanism of action of this antibody-based dual HER2 blockade. Methods To explore how the dual antibody challenge influences the phenotype and function of immune cells infiltrating the HER2-OE BC microenvironment, we developed in vitro 3D heterotypic cell models of this subtype. The models comprised aggregates of HER2+ BC cell lines and human peripheral blood mononuclear cells. Cells were co-encapsulated in a chemically inert alginate hydrogel and maintained in agitation-based culture system for up to 7 days. Results The 3D models of the HER2-OE immune microenvironment retained original BC molecular features; the preservation of the NK cell compartment was achieved upon optimization of culture time and cytokine supplementation. Challenging the models with the standard-of-care combination of trastuzumab and pertuzumab resulted in enhanced immune cytotoxicity compared with trastuzumab alone. Features of the response to therapy within the immune tumor microenvironment were recapitulated, including induction of an immune effector state with NK cell activation, enhanced cell apoptosis and decline of immunosuppressive PD-L1+ immune cells. Conclusions This work presents a unique human 3D model for the study of immune effects of anti-HER2 biologicals, which can be used to test novel therapy regimens and improve anti-tumor immune function.
Collapse
Affiliation(s)
- Sofia Batalha
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Monteiro Gomes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
12
|
Gaynor N, Blanco A, Madden SF, Moran B, Fletcher JM, Kaukonen D, Ramírez JS, Eustace AJ, McDermott MSJ, Canonici A, Toomey S, Teiserskiene A, Hennessy BT, O'Donovan N, Crown J, Collins DM. Alterations in immune cell phenotype and cytotoxic capacity in HER2+ breast cancer patients receiving HER2-targeted neo-adjuvant therapy. Br J Cancer 2023; 129:1022-1031. [PMID: 37507543 PMCID: PMC10491671 DOI: 10.1038/s41416-023-02375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The phase II neo-adjuvant clinical trial ICORG10-05 (NCT01485926) compared chemotherapy in combination with trastuzumab, lapatinib or both in patients with HER2+ breast cancer. We studied circulating immune cells looking for alterations in phenotype, genotype and cytotoxic capacity (direct and antibody-dependent cell-mediated cytotoxicity (ADCC)) in the context of treatment response. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from pre- (n = 41) and post- (n = 25) neo-adjuvant treatment blood samples. Direct/trastuzumab-ADCC cytotoxicity of patient-derived PBMCs against K562/SKBR3 cell lines was determined ex vivo. Pembrolizumab was interrogated in 21 pre-treatment PBMC ADCC assays. Thirty-nine pre-treatment and 21 post-treatment PBMC samples were immunophenotyped. Fc receptor genotype, tumour infiltrating lymphocyte (TIL) levels and oestrogen receptor (ER) status were quantified. RESULTS Treatment attenuated the cytotoxicity/ADCC of PBMCs. CD3+/CD4+/CD8+ T cells increased following therapy, while CD56+ NK cells/CD14+ monocytes/CD19+ B cells decreased with significant post-treatment immune cell changes confined to patients with residual disease. Pembrolizumab-augmented ex vivo PBMC ADCC activity was associated with residual disease, but not pathological complete response. Pembrolizumab-responsive PBMCs were associated with lower baseline TIL levels and ER+ tumours. CONCLUSIONS PBMCs display altered phenotype and function following completion of neo-adjuvant treatment. Anti-PD-1-responsive PBMCs in ex vivo ADCC assays may be a biomarker of treatment response.
Collapse
Affiliation(s)
- Nicola Gaynor
- Cancer Biotherapeutics Research Group, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Alfonso Blanco
- Flow Cytometry Core Technology, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, School of Population Heath Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Jean M Fletcher
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Damien Kaukonen
- Data Science Centre, School of Population Heath Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Javier Sánchez Ramírez
- Cancer Biotherapeutics Research Group, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Alex J Eustace
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Martina S J McDermott
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Alexandra Canonici
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ausra Teiserskiene
- Cancer Trials Ireland, RCSI House, 121 St. Stephen's Green, Dublin, Ireland
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Cancer Trials Ireland, RCSI House, 121 St. Stephen's Green, Dublin, Ireland
| | - Norma O'Donovan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - John Crown
- Cancer Biotherapeutics Research Group, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Denis M Collins
- Cancer Biotherapeutics Research Group, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland.
| |
Collapse
|
13
|
Cataldo ML, De Placido P, Esposito D, Formisano L, Arpino G, Giuliano M, Bianco R, De Angelis C, Veneziani BM. The effect of the alpha-specific PI3K inhibitor alpelisib combined with anti-HER2 therapy in HER2+/PIK3CA mutant breast cancer. Front Oncol 2023; 13:1108242. [PMID: 37469415 PMCID: PMC10353540 DOI: 10.3389/fonc.2023.1108242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Background HER2 is amplified or overexpressed in around 20% of breast cancers (BC). HER2-targeted therapies have significantly improved the prognosis of patients with HER2+ BC, however, de novo and acquired resistance to anti-HER2 treatment is common. Activating mutations in the PIK3CA gene are reported in ∼30% of HER2+ BC and are associated with resistance to anti-HER2 therapies and a poor prognosis. Here, we investigated the in vitro and in vivo antitumor efficacy of the alpha-specific PI3K inhibitor alpelisib alone or in combination with anti-HER2 therapy using a panel of HER2+ BC cell lines. We also generated models of acquired resistance to alpelisib to investigate the mechanisms underlying resistance to alpha-specific PI3K inhibition. Materials and methods PIK3CA mutant (HCC1954, KPL4 and JMT1) and wild-type (BT474 and SKBR3) HER2+ BC cell lines were used. The HCC1954 and KPL4 cells were chronically exposed to increasing concentrations of alpelisib or to alpelisib + trastuzumab in order to generate derivatives with acquired resistance to alpelisib (AR) and to alpelisib + trastuzumab (ATR). The transcriptomic profiles of HCC1954, KPL4 and their AR and ATR derivatives were determined by RNA sequencing. Cell growth was assessed by MTT assay. Changes in the protein levels of key PI3K pathway components were assessed by Western blotting. Gene expression, cellular and patients' data from the Cancer Dependency Map (DepMap) and KMPlot datasets were interrogated. Results HER2+ BC cell lines harboring activating mutations in PIK3CA were less sensitive to single or dual anti-HER2 blockade compared to PIK3CA wild-type cells. Alpelisib treatment resulted in dose-dependent inhibition of the growth of cells with or without PIK3CA mutations and enhanced the antitumor efficacy of anti-HER2 therapies in vitro. In addition, alpelisib greatly delayed tumor growth of HCC1954 xenografts in vivo. Functional annotation of the significantly differentially expressed genes suggested the common activation of biological processes associated with oxidation reduction, cell proliferation, immune response and RNA synthesis in alpelisib-resistant models compared with native cells. Eight commonly upregulated genes (log2 fold-change >1, False Discovery Rate [FDR] <0.05) in models with acquired resistance to alpelisib or alpelisib + trastuzumab were identified. Among these, AKR1C1 was associated with alpelisib-resistance in vitro and with a poor prognosis in patients with HER2+ BC. Conclusions Our findings support the use of an alpha-selective PI3K inhibitor to overcome the therapeutic limitations associated with single or dual HER2 blockade in PIK3CA-mutant HER2+ breast cancer. Future studies are warranted to confirm the potential role of candidate genes/pathways in resistance to alpelisib.
Collapse
Affiliation(s)
- Maria Letizia Cataldo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Daniela Esposito
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
14
|
Liu H, Ruan S, Larsen ME, Tan C, Liu B, Lyu H. Trastuzumab-resistant breast cancer cells-derived tumor xenograft models exhibit distinct sensitivity to lapatinib treatment in vivo. Biol Proced Online 2023; 25:19. [PMID: 37370010 DOI: 10.1186/s12575-023-00212-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Resistance to HER2-targeted therapies, including the monoclonal antibody trastuzumab and tyrosine kinase inhibitor lapatinib, frequently occurs and currently represents a significant clinical challenge in the management of HER2-positive breast cancer. We previously showed that the trastuzumab-resistant SKBR3-pool2 and BT474-HR20 sublines were refractory to lapatinib in vitro as compared to the parental SKBR3 and BT474 cells, respectively. The in vivo efficacy of lapatinib against trastuzumab-resistant breast cancer remained unclear. RESULTS In tumor xenograft models, both SKBR3-pool2- and BT474-HR20-derived tumors retained their resistance phenotype to trastuzumab; however, those tumors responded differently to the treatment with lapatinib. While lapatinib markedly suppressed growth of SKBR3-pool2-derived tumors, it slightly attenuated BT474-HR20 tumor growth. Immunohistochemistry analyses revealed that lapatinib neither affected the expression of HER3, nor altered the levels of phosphorylated HER3 and FOXO3a in vivo. Interestingly, lapatinib treatment significantly increased the levels of phosphorylated Akt and upregulated the expression of insulin receptor substrate-1 (IRS1) in the tumors-derived from BT474-HR20, but not SKBR3-pool2 cells. CONCLUSIONS Our data indicated that SKBR3-pool2-derived tumors were highly sensitive to lapatinib treatment, whereas BT474-HR20 tumors exhibited resistance to lapatinib. It seemed that the inefficacy of lapatinib against BT474-HR20 tumors in vivo was attributed to lapatinib-induced upregulation of IRS1 and activation of Akt. Thus, the tumor xenograft models-derived from SKBR3-pool2 and BT474-HR20 cells serve as an excellent in vivo system to test the efficacy of other HER2-targeted therapies and novel agents to overcome trastuzumab resistance against HER2-positive breast cancer.
Collapse
Affiliation(s)
- Hao Liu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sanbao Ruan
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Margaret E Larsen
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Congcong Tan
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Bolin Liu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA.
| | - Hui Lyu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA.
| |
Collapse
|
15
|
Liu S, Xie SM, Liu W, Gagea M, Hanker AB, Nguyen N, Singareeka Raghavendra A, Yang-Kolodji G, Chu F, Neelapu SS, Marchese A, Hanash S, Zimmermann J, Arteaga CL, Tripathy D. Targeting CXCR4 abrogates resistance to trastuzumab by blocking cell cycle progression and synergizes with docetaxel in breast cancer treatment. Breast Cancer Res 2023; 25:62. [PMID: 37280713 DOI: 10.1186/s13058-023-01665-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Although trastuzumab and other HER2-targeted therapies have significantly improved survival in patients with HER2 overexpressed or amplified (HER2+) breast cancer, a significant proportion of patients do not respond or eventually develop clinical resistance. Strategies to reverse trastuzumab resistance remain a high clinical priority. We were the first to report the role of CXCR4 in trastuzumab resistance. The present study aims to explore the therapeutic potential of targeting CXCR4 and better understand the associated mechanisms. METHODS Immunofluorescent staining, confocal microscopy analysis, and immunoblotting were used to analyze CXCR4 expression. BrdU incorporation assays and flow cytometry were used to analyze dynamic CXCR4 expression. Three-dimensional co-culture (tumor cells/breast cancer-associated fibroblasts/human peripheral blood mononuclear cells) or antibody-dependent cellular cytotoxicity assay was used to mimic human tumor microenvironment, which is necessary for testing therapeutic effects of CXCR4 inhibitor or trastuzumab. The FDA-approved CXCR4 antagonist AMD3100, trastuzumab, and docetaxel chemotherapy were used to evaluate therapeutic efficacy in vitro and in vivo. Reverse phase protein array and immunoblotting were used to discern the associated molecular mechanisms. RESULTS Using a panel of cell lines and patient breast cancer samples, we confirmed CXCR4 drives trastuzumab resistance in HER2+ breast cancer and further demonstrated the increased CXCR4 expression in trastuzumab-resistant cells is associated with cell cycle progression with a peak in the G2/M phases. Blocking CXCR4 with AMD3100 inhibits cell proliferation by downregulating mediators of G2-M transition, leading to G2/M arrest and abnormal mitosis. Using a panel of trastuzumab-resistant cell lines and an in vivo established trastuzumab-resistant xenograft mouse model, we demonstrated that targeting CXCR4 with AMD3100 suppresses tumor growth in vitro and in vivo, and synergizes with docetaxel. CONCLUSIONS Our findings support CXCR4 as a novel therapeutic target and a predictive biomarker for trastuzumab resistance in HER2+ breast cancer.
Collapse
Affiliation(s)
- Shuying Liu
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shelly M Xie
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wenbin Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ariella B Hanker
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nguyen Nguyen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Gloria Yang-Kolodji
- Department of Medicine, University of South California, Los Angeles, CA, USA
| | - Fuliang Chu
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sattva S Neelapu
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Carlos L Arteaga
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Debasish Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
16
|
Fiascarelli A, Merlino G, Capano S, Talucci S, Bisignano D, Bressan A, Bellarosa D, Carrisi C, Paoli A, Bigioni M, Tunici P, Irrissuto C, Salerno M, Arribas J, de Stanchina E, Scaltriti M, Binaschi M. Antitumor activity of the PI3K δ-sparing inhibitor MEN1611 in PIK3CA mutated, trastuzumab-resistant HER2 + breast cancer. Breast Cancer Res Treat 2023; 199:13-23. [PMID: 36913051 PMCID: PMC10147754 DOI: 10.1007/s10549-023-06895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/14/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE Dysregulation of the PI3K pathway is one of the most common events in breast cancer. Here we investigate the activity of the PI3K inhibitor MEN1611 at both molecular and phenotypic levels by dissecting and comparing its profile and efficacy in HER2 + breast cancer models with other PI3K inhibitors. METHODS Models with different genetic backgrounds were used to investigate the pharmacological profile of MEN1611 against other PI3K inhibitors. In vitro studies evaluated cell viability, PI3K signaling, and cell death upon treatment with MEN1611. In vivo efficacy of the compound was investigated in cell line- and patient-derived xenografts models. RESULTS Consistent with its biochemical selectivity, MEN1611 demonstrated lower cytotoxic activity in a p110δ-driven cellular model when compared to taselisib, and higher cytotoxic activity in the p110β-driven cellular model when compared to alpelisib. Moreover, MEN1611 selectively decreased the p110α protein levels in PIK3CA mutated breast cancer cells in a concentration- and proteasome-dependent manner. In vivo, MEN1611 monotherapy showed significant and durable antitumor activity in several trastuzumab-resistant PIK3CA-mutant HER2 + PDX models. The combination of trastuzumab and MEN1611 significantly improved the efficacy compared to single agent treatment. CONCLUSIONS The profile of MEN1611 and its antitumoral activity suggest an improved profile as compared to pan-inhibitors, which are limited by a less than ideal safety profile, and isoform selective molecules, which may potentially promote development of resistance mechanisms. The compelling antitumor activity in combination with trastuzumab in HER2 + trastuzumab-resistant, PIK3CA mutated breast cancer models is at the basis of the ongoing B-Precise clinical trial (NCT03767335).
Collapse
Affiliation(s)
- Alessio Fiascarelli
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy.
| | - Giuseppe Merlino
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Stefania Capano
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Simone Talucci
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Diego Bisignano
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Alessandro Bressan
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Daniela Bellarosa
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Corrado Carrisi
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Alessandro Paoli
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Mario Bigioni
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Patrizia Tunici
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Clelia Irrissuto
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Massimiliano Salerno
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| | - Joaquin Arribas
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Preclinical and Translational Research Program Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, 28029, Monforte de Lemos, Madrid, Spain.,Department of Biochemistry and Molecular Biology, Universitat Autónoma de Barcelona, Campus de la UAB, 08193, Barcelona, Bellaterra, Spain.,Institució Catalana de Recerca I Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Elisa de Stanchina
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maurizio Scaltriti
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Monica Binaschi
- Menarini Group, Preclinical and Translational Sciences, Menarini Ricerche SpA, Via Tito Speri 10, 00071, Pomezia, Rome, Italy
| |
Collapse
|
17
|
Liu S, Xie SM, Liu W, Gagea M, Hanker AB, Nguyen N, Raghavendra AS, Yang-Kolodji G, Chu F, Neelapu SS, Hanash S, Zimmermann J, Arteaga CL, Tripathy D. Targeting CXCR4 abrogates resistance to trastuzumab by blocking cell cycle progression and synergizes with docetaxel in breast cancer treatment. RESEARCH SQUARE 2023:rs.3.rs-2388864. [PMID: 36824840 PMCID: PMC9949251 DOI: 10.21203/rs.3.rs-2388864/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Background: Although trastuzumab and other HER2-targeted therapies have significantly improved survival in patients with HER2 overexpressed or amplified (HER2+) breast cancer, a significant proportion of patients do not respond or eventually develop clinical resistance. Strategies to reverse trastuzumab resistance remain a high clinical priority. We were the first to report the role of CXCR4 in trastuzumab resistance. The present study aims to explore the therapeutic potential of targeting CXCR4 and better understand the associated mechanisms. Methods: Immunofluorescent staining, confocal microscopy analysis, and immunoblotting were used to analyze CXCR4 expression. BrdU incorporation assays and flow cytometry were used to analyze dynamic CXCR4expression. Three-dimensional co-culture (tumor cells/ breast cancer-associated fibroblasts / human peripheral blood mononuclear cells) or antibody-dependent cellular cytotoxicity assay was used to mimic human tumor microenvironment, which is necessary for testing therapeutic effect of CXCR4 inhibitor or trastuzumab. The FDA-approved CXCR4 antagonist AMD3100, trastuzumab, and docetaxel chemotherapy were used to evaluate therapeutic efficacy in vitro and in vivo. Reverse phase protein array and immunoblotting were used to discern the associated molecular mechanisms. Results: Using multiple cell lines and patient breast cancer samples we confirmed CXCR4 drives trastuzumab resistance in HER2+ breast cancer and further demonstrated that the increased CXCR4 expression in trastuzumab-resistant cells is associated with cell cycle progression with a peak in the G2/M phases. Blocking CXCR4 with AMD3100 inhibits cell proliferation by downregulating mediators of G2-M transition, leading to G2/M arrest and abnormal mitosis. Using multiple trastuzumab-resistant cell lines and an in vivo established trastuzumab-resistant xenograft mouse model, we demonstrated that targeting CXCR4 with AMD3100 suppresses tumor growth in vitro and in vivo, and synergizes with docetaxel. Conclusions: Our findings support CXCR4 as a novel therapeutic target and a predictive biomarker for trastuzumab resistance in HER2+ breast cancer.
Collapse
Affiliation(s)
- Shuying Liu
- The University of Texas MD Anderson Cancer Center
| | | | - Wenbin Liu
- The University of Texas MD Anderson Cancer Center
| | - Mihai Gagea
- The University of Texas MD Anderson Cancer Center
| | | | | | | | | | - Fuliang Chu
- The University of Texas MD Anderson Cancer Center
| | | | - Samir Hanash
- The University of Texas MD Anderson Cancer Center
| | | | | | | |
Collapse
|
18
|
Purazo ML, Ice RJ, Shimpi R, Hoenerhoff M, Pugacheva EN. NEDD9 Overexpression Causes Hyperproliferation of Luminal Cells and Cooperates with HER2 Oncogene in Tumor Initiation: A Novel Prognostic Marker in Breast Cancer. Cancers (Basel) 2023; 15:1119. [PMID: 36831460 PMCID: PMC9954084 DOI: 10.3390/cancers15041119] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/23/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
HER2 overexpression occurs in 10-20% of breast cancer patients. HER2+ tumors are characterized by an increase in Ki67, early relapse, and increased metastasis. Little is known about the factors influencing early stages of HER2- tumorigenesis and diagnostic markers. Previously, it was shown that the deletion of NEDD9 in mouse models of HER2 cancer interferes with tumor growth, but the role of NEDD9 upregulation is currently unexplored. We report that NEDD9 is overexpressed in a significant subset of HER2+ breast cancers and correlates with a limited response to anti-HER2 therapy. To investigate the mechanisms through which NEDD9 influences HER2-dependent tumorigenesis, we generated MMTV-Cre-NEDD9 transgenic mice. The analysis of mammary glands shows extensive ductal epithelium hyperplasia, increased branching, and terminal end bud expansion. The addition of oncogene Erbb2 (neu) leads to the earlier development of early hyperplastic benign lesions (~16 weeks), with a significantly shorter latency than the control mice. Similarly, NEDD9 upregulation in MCF10A-derived acini leads to hyperplasia-like DCIS. This phenotype is associated with activation of ERK1/2 and AURKA kinases, leading to an increased proliferation of luminal cells. These findings indicate that NEDD9 is setting permissive conditions for HER2-induced tumorigenesis, thus identifying this protein as a potential diagnostic marker for early detection.
Collapse
Affiliation(s)
- Marc L. Purazo
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Ryan J. Ice
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Rahul Shimpi
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | - Mark Hoenerhoff
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elena N. Pugacheva
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
- Department of Biochemistry & Molecular Medicine, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
19
|
Swain SM, Shastry M, Hamilton E. Targeting HER2-positive breast cancer: advances and future directions. Nat Rev Drug Discov 2023; 22:101-126. [PMID: 36344672 PMCID: PMC9640784 DOI: 10.1038/s41573-022-00579-0] [Citation(s) in RCA: 277] [Impact Index Per Article: 277.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2022] [Indexed: 11/09/2022]
Abstract
The long-sought discovery of HER2 as an actionable and highly sensitive therapeutic target was a major breakthrough for the treatment of highly aggressive HER2-positive breast cancer, leading to approval of the first HER2-targeted drug - the monoclonal antibody trastuzumab - almost 25 years ago. Since then, progress has been swift and the impressive clinical activity across multiple trials with monoclonal antibodies, tyrosine kinase inhibitors and antibody-drug conjugates that target HER2 has spawned extensive efforts to develop newer platforms and more targeted therapies. This Review discusses the current standards of care for HER2-positive breast cancer, mechanisms of resistance to HER2-targeted therapy and new therapeutic approaches and agents, including strategies to harness the immune system.
Collapse
Affiliation(s)
- Sandra M Swain
- Department of Medicine, Georgetown Lombardi Comprehensive Cancer Center and MedStar Health, Washington, DC, USA.
| | | | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, TN, USA
- Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
20
|
Margetuximab and trastuzumab deruxtecan: New generation of anti-HER2 immunotherapeutic agents for breast cancer. Mol Immunol 2022; 152:45-54. [DOI: 10.1016/j.molimm.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 09/24/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
|
21
|
Aryanpour N, Farnam G, Behtaj R, H Shirazi F. The Complexity of Response to the Proliferation Agonist and Antagonist Agents, in the Breast Cancer Cell Lines with Various Receptors. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e123823. [PMID: 35765511 PMCID: PMC9191223 DOI: 10.5812/ijpr.123823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/24/2021] [Accepted: 09/05/2021] [Indexed: 11/16/2022]
Abstract
Breast cancer is a heterogeneous disease in which many factors and receptors are effective in the disease process and response to treatment. Currently, estrogen, progesterone, and HER2 receptors are among the most important factors in choosing a treatment regimen. Other metabolic factors that may affect the treatment outcome include diabetes and hyperinsulinemia. In order to evaluate the role and complexity of cross-talk between different pathways initiating from various receptors, value the most common drugs in the treatment of breast cancer are investigated on different cell lines in this manuscript at the cell culture level. The result of different doses of Tamoxifen and estradiol on the cells with various levels of the estrogenic, progesterone, and HER2 receptors is examined alone, or in combinations, and the presence or absence of insulin. The effects of these variables on the cells' growth pattern and survival in various breast cancer cells are investigated using cell counting, colony counting, and MTT assays. Our results have further confirmed the complexity of deciding on the outcome of treatment for breast cancer with such a wide variability in the kind of receptors and biochemical agents present in the body of a cancer patient.
Collapse
Affiliation(s)
- Narges Aryanpour
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Golrokh Farnam
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Behtaj
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshad H Shirazi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Furrer D, Dragic D, Chang SL, Fournier F, Droit A, Jacob S, Diorio C. Association between genome-wide epigenetic and genetic alterations in breast cancer tissue and response to HER2-targeted therapies in HER2-positive breast cancer patients: new findings and a systematic review. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:995-1015. [PMID: 36627894 PMCID: PMC9771759 DOI: 10.20517/cdr.2022.63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/25/2022] [Accepted: 10/08/2022] [Indexed: 01/12/2023]
Abstract
Recent evidence suggests that genetic and epigenetic mechanisms might be associated with acquired resistance to cancer therapies. The aim of this study was to assess the association of genome-wide genetic and epigenetic alterations with the response to anti-HER2 agents in HER2-positive breast cancer patients. PubMed was screened for articles published until March 2021 on observational studies investigating the association of genome-wide genetic and epigenetic alterations, measured in breast cancer tissues or blood, with the response to targeted treatment in HER2-positive breast cancer patients. Sixteen studies were included in the review along with ours, in which we compared the genome-wide DNA methylation pattern in breast tumor tissues of patients who acquired resistance to treatment (case group, n = 6) to that of patients who did not develop resistance (control group, n = 6). Among genes identified as differentially methylated between the breast cancer tissue of cases and controls, one of them, PRKACA, was also reported as differentially expressed in two studies included in the review. Although included studies were heterogeneous in terms of methodology and study population, our review suggests that genes of the PI3K pathway may play an important role in developing resistance to anti-HER2 agents in breast cancer patients. Genome-wide genetic and epigenetic alterations measured in breast cancer tissue or blood might be promising markers of resistance to anti-HER2 agents in HER2-positive breast cancer patients. Further studies are needed to confirm these data.
Collapse
Affiliation(s)
- Daniela Furrer
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine sociale et préventive, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Dzevka Dragic
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine sociale et préventive, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada.,Université Paris-Saclay, UVSQ, Inserm, CESP U1018, Exposome and Heredity Team, Gustave Roussy, Villejuif 94807, France
| | - Sue-Ling Chang
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada
| | - Frédéric Fournier
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine moléculaire, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Arnaud Droit
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine moléculaire, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Simon Jacob
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada.,Centre des Maladies du Sein, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada
| | - Caroline Diorio
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine sociale et préventive, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada.,Centre des Maladies du Sein, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada.,Correspondence to: Prof. Caroline Diorio, Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, 1050 chemin Ste-Foy, Québec, QC G1S 4L8, Canada. E-mail:
| |
Collapse
|
23
|
Zhu MY, Gong ZS, Feng HP, Zhang QY, Liu K, Lin B, Zhang MN, Lin HF, Li MS. Vincosamide Has a Function for Inhibiting Malignant Behaviors of Hepatocellular Carcinoma Cells. World J Oncol 2022; 13:272-288. [PMID: 36406198 PMCID: PMC9635790 DOI: 10.14740/wjon1514] [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: 07/03/2022] [Accepted: 08/13/2022] [Indexed: 08/30/2023] Open
Abstract
BACKGROUND Vincosamide (Vinco) was first identified in the methanolic extract of the leaves of Psychotria leiocarpa, and Vinco has important anti-inflammatory effects and activity against cholinesterase, Vinco also has a trait to anti-tumor. However, whether Vinco can inhibit the malignant behaviors of hepatocellular carcinoma (HCC) cells is still unclear. In the present study, we explored the role of Vinco in suppressing the malignant behaviors of HCC cells. METHODS MTT (3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-tetrazolium bromide), trypan blue exclusion assay, the Cell Counting Kit (CCK)-8 and flow cytometric analysis were applied to detect the proliferation and apoptosis of HCC cells; electron microscopy was performed to observe the change of cellular mitochondrial morphology; scratch repair and Transwell assays were used to analyze the migration and invasion of HCC cells; expression and localization of proteins were detected by laser confocal microscopy and Western blotting; the growth of the cancer cells in vivo was assessed in a mouse tumorous model. RESULTS At a dose of 10 - 80 µg/mL, Vinco inhibited the proliferation, migration, invasion and promoted apoptosis of HCC cells in a dose-dependent manner but had low cytotoxicity effect on normal liver cells. Additionally, 80 µg/mL of Vinco could significantly disrupt the morphology of mitochondria, suppress the migration and invasion of HCC cells. The growth of HCC cells in the animal tumorous model was significantly inhibited after treatment with Vinco (10 mg/kg/day) for 3 days. The results of the present study indicated that Vinco (10 - 80 µg/mL) played a role in activating caspase-3, promoting the expression of phosphate and tension homology deleted on chromosome 10 (PTEN), and inhibiting the phosphorylation of AKT (Ser473) and mTOR (Thr2448); Vinco also has a trait for suppressing the expression of CXCR4, Src, MMP9, EpCAM, Ras, Oct4 and cancer stem cell "stemness markers" CD133 and CD44 in HCC cells. CONCLUSIONS Vinco has a role in inhibiting the malignant behaviors of HCC cells; the role molecular mechanism of Vinco may be involved in restraining expression of the growth-, metastasis-related factors, such as Src, Ras, MMP9, EpCAM, CXCR4; activating the activity of caspase-3 and blocking PI3K/AKT signaling pathway. Thus, Vinco should be considered as a new chemotherapy agent for HCC patients.
Collapse
Affiliation(s)
- Ming Yue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
- These authors contributed equally to this work and are co-first authors
| | - Zhi Sun Gong
- Department of Radiotherapy, Second Affiliated Hospital, Hainan Medical College, Haikou, China
- These authors contributed equally to this work and are co-first authors
| | - Hai Peng Feng
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
- These authors contributed equally to this work and are co-first authors
| | - Qiu Yue Zhang
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Kun Liu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Bo Lin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Min Ni Zhang
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Hai Feng Lin
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical College, Haikou, China
| | - Meng Sen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical College, Haikou, China
- Institution of Tumor, Hainan Medical College, Hiakou 570102, Hainan Province, China
| |
Collapse
|
24
|
Kim LM, Kim PY, Gebreyohannes YK, Leung CT. Sustained Oncogenic Signaling in the Cytostatic State Enables Targeting of Nonproliferating Persistent Cancer Cells. Cancer Res 2022; 82:3045-3057. [PMID: 35792658 PMCID: PMC9444958 DOI: 10.1158/0008-5472.can-21-2908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
Many advanced therapeutics possess cytostatic properties that suppress cancer cell growth without directly inducing death. Treatment-induced cytostatic cancer cells can persist and constitute a reservoir from which recurrent growth and resistant clones can develop. Current management approaches primarily comprise maintenance and monitoring because strategies for targeting nonproliferating cancer cells have been elusive. Here, we used targeted therapy paradigms and engineered cytostatic states to explore therapeutic opportunities for depleting treatment-mediated cytostatic cancer cells. Sustained oncogenic AKT signaling was common, while nonessential, in treatment-mediated cytostatic cancer cells harboring PI3K-pathway mutations, which are associated with cancer recurrence. Engineering oncogenic signals in quiescent mammary organotypic models showed that sustained, aberrant activation of AKT sensitized cytostatic epithelial cells to proteasome inhibition. Mechanistically, sustained AKT signaling altered cytostatic state homeostasis and promoted an oxidative and proteotoxic environment, which imposed an increased proteasome dependency for maintaining cell viability. Under cytostatic conditions, inhibition of the proteasome selectively induced apoptosis in the population with aberrant AKT activation compared with normal cells. Therapeutically exploiting this AKT-driven proteasome vulnerability was effective in depleting treatment-mediated cytostatic cancer cells independent of breast cancer subtype, epithelial origin, and cytostatic agent. Moreover, transient targeting during cytostatic treatment conditions was sufficient to reduce recurrent tumor growth in spheroid and mouse models. This work identified an AKT-driven proteasome-vulnerability that enables depletion of persistent cytostatic cancer cells harboring PTEN-PI3K pathway mutations, revealing a viable strategy for targeting nonproliferating persistent cancer cell populations before drug resistance emerges. SIGNIFICANCE This study finds that sustained oncogenic signaling in therapy-induced cytostatic cancer cells confers targetable vulnerabilities to deplete persistent cancer cell populations and reduce cancer recurrence.
Collapse
Affiliation(s)
| | | | | | - Cheuk T. Leung
- Corresponding author: Cheuk T. Leung, Address: 321 Church Street SE, 6-120 Jackson Hall, Minneapolis, MN 55455, USA, , Phone: 612-626-5309
| |
Collapse
|
25
|
Yang C, Fan S, Wang X, Liu W, Yang L, He B, Dai W, Zhang H, Wang X, Zhang Q. Optimize the combination regimen of Trastuzumab and Nab-paclitaxel in HER2-positive tumors via modulating Caveolin-1 expression by lovastatin. Asian J Pharm Sci 2022; 17:697-712. [PMID: 36382307 PMCID: PMC9640369 DOI: 10.1016/j.ajps.2022.06.002] [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: 05/07/2022] [Revised: 06/12/2022] [Accepted: 06/25/2022] [Indexed: 11/02/2022] Open
Abstract
The combination regimen of trastuzumab (Tras) plus Nab-paclitaxel (Nab) is recommended to treat HER2-positive (HER2+) cancers. However, they exert effects in different mechanisms: Tras need to stay on cell membranes, while Nab need to be endocytosed, therefore the concurrent combination regimen may not be the best one in HER2+ tumors treatment. Caveolin-1 (Cav-1) is a key player in mediating their endocytosis and is associated with their efficacy, but few researches noticed the opposite effect of Cav-1 expression on the combination efficacy. Herein, we systematically studied the Cav-1 expression level on the combination efficacy and proposed an optimized and clinically feasible combination regimen for HER2+ Cav-1High tumor treatment. In the regimen, lovastatin (Lova) was introduced to modulate the Cav-1 expression and the results indicated that Lova could downregulate Cav-1 expression, increase Tras retention on cell membrane and enhance the in vitro cytotoxicity of Tras in HER2+ Cav-1High cells but not in HER2+ Cav-1Low cells. Therefore, by exchanging the dosing sequence of Nab and Tras, and by adding Lova at appropriate time points, the precise three-drug-sequential regimen (PTDS, Nab(D1)-Lova(D2)-Lova & Tras(D2+12 h)) was established. Compared with the concurrent regimen, the PTDS regimen exhibited a higher in vitro cytotoxicity and a stronger tumor growth inhibition in HER2+ Cav-1High tumors, which might be a promising combination regimen for these patients in clinics.
Collapse
Affiliation(s)
- Canyu Yang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
| | - Shumin Fan
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xing Wang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
| | - Wei Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100083, China
| | - Long Yang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| |
Collapse
|
26
|
Wang H, Miao J, Wen Y, Xia X, Chen Y, Huang M, Chen S, Zhao Z, Zhang Y, Chen C, Zhu X. Molecular Landscape of ERBB2 Alterations in 14,956 Solid Tumors. Pathol Oncol Res 2022; 28:1610360. [PMID: 35911441 PMCID: PMC9325965 DOI: 10.3389/pore.2022.1610360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/23/2022] [Indexed: 12/11/2022]
Abstract
ERBB2 abnormalities frequently occur and serve as rationale therapeutic targets in cancer. In this study, clinical and next-generation sequencing data from 14,956 patients across more than 20 tumor types were collected. A total of 406 (2.7%) patients were identified with ERBB2 amplifications, and 303 (2.0%) patients with pathogenic somatic ERBB2 mutations. ERBB2 amplifications fell most frequently in breast (15.9%) and stomach (8.3%) cancers. Somatic ERBB2 SNVs/indels occurred most common in bladder/urinary tract (7.3%) and intestine (6.1%) cancers. The top mutated ERBB2 SNVs/indels were p.Y772_A775dup (25.5%) and p.S310F/Y (19.9%). Significantly higher rates of ERBB2 SNV/indels were found in women compared to men (2.8% vs. 1.5%, p < 0.0001). CDK12 was the most common co-amplification gene with ERBB2 in cancers with a high frequency of ERBB2 amplifications. Patients with ERBB2 amplifications or mutations had higher TMB compared with patients with non-ERBB2 alterations. The study provided the landscape of ERBB2 alterations across a variety of solid tumors that may benefit from anti-HER2 agents.
Collapse
Affiliation(s)
- Hao Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ji Miao
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yazhou Wen
- Department of Anesthesiology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xihua Xia
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yanan Chen
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Mengli Huang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Shiqing Chen
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Zhengyi Zhao
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yuzi Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Chunzhu Chen
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Xinhua Zhu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Xinhua Zhu,
| |
Collapse
|
27
|
Gobbo MG, de Mendonça Fernandes GM, Fernandes-Ferreira R, Caires LP, Caldas HC, de Campos Zuccari DAP, Bordin-Junior NA, Gonçalves Vidotti GA, Souza DRS. Evaluation of doxorubicin in three-dimensional culture of breast cancer cells and the response in PI3K/AKT/PTEN signaling pathways: a pilot study. Women Health 2022; 62:467-475. [DOI: 10.1080/03630242.2022.2085842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Marina Guimarães Gobbo
- Biologia Molecular (NPBIM), Faculdade de Medicina de São Jose do Rio Preto, (FAMERP)Núcleo de Pesquisa em Bioquímica e , São Paulo, Brazil
| | | | - Rafael Fernandes-Ferreira
- Biologia Molecular (NPBIM), Faculdade de Medicina de São Jose do Rio Preto, (FAMERP)Núcleo de Pesquisa em Bioquímica e , São Paulo, Brazil
| | - Lennon Pereira Caires
- Laboratory of Immunology and Experimental Transplantation (LITEX), Faculdade de Medicina de São Jose do Rio Preto, (FAMERP), São Paulo, Brazil
| | - Heloisa Cristina Caldas
- Laboratory of Immunology and Experimental Transplantation (LITEX), Faculdade de Medicina de São Jose do Rio Preto, (FAMERP), São Paulo, Brazil
| | | | - Newton Antonio Bordin-Junior
- Departamento de Ginecologia do Hospital de Base da Faculdade de Medicina de São José do Rio Preto Serviço de mastologia do Hospital de Base da Faculdade de Medicina de São José do Rio Preto, São Paulo, Brazil
| | | | - Doroteia Rossi Silva Souza
- Biologia Molecular (NPBIM), Faculdade de Medicina de São Jose do Rio Preto, (FAMERP)Núcleo de Pesquisa em Bioquímica e , São Paulo, Brazil
| |
Collapse
|
28
|
Madera S, Izzo F, Chervo MF, Dupont A, Chiauzzi VA, Bruni S, Petrillo E, Merin SS, De Martino M, Montero D, Levit C, Lebersztein G, Anfuso F, Roldán Deamicis A, Mercogliano MF, Proietti CJ, Schillaci R, Elizalde PV, Cordo Russo RI. Halting ErbB-2 isoforms retrograde transport to the nucleus as a new theragnostic approach for triple-negative breast cancer. Cell Death Dis 2022; 13:447. [PMID: 35534460 PMCID: PMC9084267 DOI: 10.1038/s41419-022-04855-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022]
Abstract
Triple-negative breast cancer (TNBC) is clinically defined by the absence of estrogen and progesterone receptors and the lack of membrane overexpression or gene amplification of receptor tyrosine kinase ErbB-2/HER2. Due to TNBC heterogeneity, clinical biomarkers and targeted therapies for this disease remain elusive. We demonstrated that ErbB-2 is localized in the nucleus (NErbB-2) of TNBC cells and primary tumors, from where it drives growth. We also discovered that TNBC expresses both wild-type ErbB-2 (WTErbB-2) and alternative ErbB-2 isoform c (ErbB-2c). Here, we revealed that the inhibitors of the retrograde transport Retro-2 and its cyclic derivative Retro-2.1 evict both WTErbB-2 and ErbB-2c from the nucleus of BC cells and tumors. Using BC cells from several molecular subtypes, as well as normal breast cells, we demonstrated that Retro-2 specifically blocks proliferation of BC cells expressing NErbB-2. Importantly, Retro-2 eviction of both ErbB-2 isoforms from the nucleus resulted in a striking growth abrogation in multiple TNBC preclinical models, including tumor explants and xenografts. Our mechanistic studies in TNBC cells revealed that Retro-2 induces a differential accumulation of WTErbB-2 at the early endosomes and the plasma membrane, and of ErbB-2c at the Golgi, shedding new light both on Retro-2 action on endogenous protein cargoes undergoing retrograde transport, and on the biology of ErbB-2 splicing variants. In addition, we revealed that the presence of a functional signal peptide and a nuclear export signal (NES), both located at the N-terminus of WTErbB-2, and absent in ErbB-2c, accounts for the differential subcellular distribution of ErbB-2 isoforms upon Retro-2 treatment. Our present discoveries provide evidence for the rational repurposing of Retro-2 as a novel therapeutic agent for TNBC.
Collapse
Affiliation(s)
- Santiago Madera
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Franco Izzo
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - María F Chervo
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Agustina Dupont
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Violeta A Chiauzzi
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Sofia Bruni
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Ezequiel Petrillo
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología, Biología Molecular y Celular and CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), C1428EHA, Buenos Aires, Argentina
| | - Sharon S Merin
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Mara De Martino
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Diego Montero
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Claudio Levit
- Servicio de Ginecología, Sanatorio Sagrado Corazón, Buenos Aires, Argentina
| | | | - Fabiana Anfuso
- Servicio de Ginecología, Sanatorio Sagrado Corazón, Buenos Aires, Argentina
| | - Agustina Roldán Deamicis
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - María F Mercogliano
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Cecilia J Proietti
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Roxana Schillaci
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Patricia V Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| | - Rosalía I Cordo Russo
- Laboratory of Molecular Mechanisms of Carcinogenesis and Molecular Endocrinology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| |
Collapse
|
29
|
O'Brien NA, Huang HKT, McDermott MSJ, Madrid AM, Luo T, Ayala R, Issakhanian S, Gong KW, Lu M, Zhang J, Slamon DJ. Tucatinib has Selective Activity in HER2-Positive Cancers and Significant Combined Activity with Approved and Novel Breast Cancer-Targeted Therapies. Mol Cancer Ther 2022; 21:751-761. [PMID: 35417017 DOI: 10.1158/1535-7163.mct-21-0847] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/19/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022]
Abstract
Pharmacologically targeting the HER2 oncoprotein with therapeutics such as the mAb, trastuzumab, provides clinical benefit for patients with HER2-positive (HER2+) cancers. However, a significant number of patients eventually progress on these therapies. Efforts to overcome therapeutic resistance through combination therapy with small-molecule inhibitors of HER2 have been limited by toxicities associated with off-target activity and/or limited efficacy. In this preclinical study, we explore single-agent and combined activity of tucatinib, a novel HER2-selective small-molecule inhibitor. Tucatinib demonstrated potent, selective activity in a panel of 456 human cancer cell lines, with activity restricted to cell lines (breast and non-breast) with HER2-amplification, including models of acquired resistance to trastuzumab. Within the HER2+ population, tucatinib response tracked strongly with HER2-driven signaling. Single-agent tucatinib induced tumor regressions in xenograft models of HER2+ breast cancer and combination with trastuzumab induced a complete and sustained blockade of HER2/PI3K/AKT signaling. Efficacy of the tucatinib/trastuzumab combination matched that induced by current standard-of-care trastuzumab/pertuzumab/docetaxel combination, with the exception that the chemotherapy-sparing tucatinib/trastuzumab combination did not require a dosing holiday to achieve the same efficacy. In xenograft models of HER2+ breast cancer that also express estrogen receptor (ER; HER2+/ER+), tucatinib showed combined efficacy with inhibitors of CDK4/6 and ER, indicating potential novel therapeutic strategies for difficult-to-treat subtypes of HER2+ breast cancer. These data support expanded clinical investigations of tucatinib as a combination partner for other novel and approved targeted therapies for HER2-driven malignancies.
Collapse
Affiliation(s)
- Neil A O'Brien
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Holly K T Huang
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Martina S J McDermott
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Athena M Madrid
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Tong Luo
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Raul Ayala
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Shawnt Issakhanian
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ke Wei Gong
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ming Lu
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jun Zhang
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Dennis J Slamon
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
30
|
Chang CA, Jen J, Jiang S, Sayad A, Mer AS, Brown KR, Nixon AM, Dhabaria A, Tang KH, Venet D, Sotiriou C, Deng J, Wong KK, Adams S, Meyn P, Heguy A, Skok JA, Tsirigos A, Ueberheide B, Moffat J, Singh A, Haibe-Kains B, Khodadadi-Jamayran A, Neel BG. Ontogeny and Vulnerabilities of Drug-Tolerant Persisters in HER2+ Breast Cancer. Cancer Discov 2022; 12:1022-1045. [PMID: 34911733 PMCID: PMC8983469 DOI: 10.1158/2159-8290.cd-20-1265] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022]
Abstract
Resistance to targeted therapies is an important clinical problem in HER2-positive (HER2+) breast cancer. "Drug-tolerant persisters" (DTP), a subpopulation of cancer cells that survive via reversible, nongenetic mechanisms, are implicated in resistance to tyrosine kinase inhibitors (TKI) in other malignancies, but DTPs following HER2 TKI exposure have not been well characterized. We found that HER2 TKIs evoke DTPs with a luminal-like or a mesenchymal-like transcriptome. Lentiviral barcoding/single-cell RNA sequencing reveals that HER2+ breast cancer cells cycle stochastically through a "pre-DTP" state, characterized by a G0-like expression signature and enriched for diapause and/or senescence genes. Trajectory analysis/cell sorting shows that pre-DTPs preferentially yield DTPs upon HER2 TKI exposure. Cells with similar transcriptomes are present in HER2+ breast tumors and are associated with poor TKI response. Finally, biochemical experiments indicate that luminal-like DTPs survive via estrogen receptor-dependent induction of SGK3, leading to rewiring of the PI3K/AKT/mTORC1 pathway to enable AKT-independent mTORC1 activation. SIGNIFICANCE DTPs are implicated in resistance to anticancer therapies, but their ontogeny and vulnerabilities remain unclear. We find that HER2 TKI-DTPs emerge from stochastically arising primed cells ("pre-DTPs") that engage either of two distinct transcriptional programs upon TKI exposure. Our results provide new insights into DTP ontogeny and potential therapeutic vulnerabilities. This article is highlighted in the In This Issue feature, p. 873.
Collapse
Affiliation(s)
- Chewei Anderson Chang
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jayu Jen
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Shaowen Jiang
- Applied Bioinformatics Laboratories, Office of Science and Research, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Azin Sayad
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Arvind Singh Mer
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Kevin R. Brown
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Avantika Dhabaria
- Proteomics Laboratory, Division of Advanced Research and Technology, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Kwan Ho Tang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - David Venet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet Brussels and Université Libre de Bruxelles (ULB), Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet Brussels and Université Libre de Bruxelles (ULB), Belgium
- Medical Oncology Department, Institut Jules Bordet Brussels and Université Libre de Bruxelles (ULB), Belgium
| | - Jiehue Deng
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Kwok-kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Sylvia Adams
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Peter Meyn
- Genome Technology Center, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Adriana Heguy
- Genome Technology Center, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Jane A. Skok
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Department of Pathology, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Aristotelis Tsirigos
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Applied Bioinformatics Laboratories, Office of Science and Research, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Department of Pathology, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Beatrix Ueberheide
- Proteomics Laboratory, Division of Advanced Research and Technology, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Jason Moffat
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, University of Delaware, Newark, Delaware, USA
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Benjamin Haibe-Kains
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario, Canada
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories, Office of Science and Research, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Benjamin G. Neel
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| |
Collapse
|
31
|
Casadevall D, Hernandez-Prat A, Garc A-Alonso S, Arpi-Llucia O, Menendez S, Qin M, Guardia C, Morancho B, Sanchez-Mart N FJ, Zazo S, Gavilan E, Sabbaghi M, Eroles P, Cejalvo JM, Lluch A, Rojo F, Pandiella A, Rovira A, Albanell J. mTOR inhibition and trastuzumab-emtansine (T-DM1) in HER2-positive breast cancer. Mol Cancer Res 2022; 20:1108-1121. [PMID: 35348729 DOI: 10.1158/1541-7786.mcr-21-0545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 02/06/2022] [Accepted: 03/23/2022] [Indexed: 12/09/2022]
Abstract
In patients with trastuzumab-resistant HER2-positive breast cancer, the combination of everolimus (mTORC1 inhibitor) with trastuzumab failed to show a clinically significant benefit. However, the combination of mTOR inhibition and the antibody-drug conjugate (ADC) trastuzumab-emtansine (T-DM1) remains unexplored. We tested T-DM1 plus everolimus in a broad panel of HER2-positive breast cancer cell lines. The combination was superior to T-DM1 alone in four cell lines (HCC1954, SKBR3, EFM192A, and MDA-MB-36) and in two cultures from primary tumor cells derived from HER2-positive patient-derived xenografts (PDX), but not in BT474 cells. In the trastuzumab-resistant HCC1954 cell line, we characterized the effects of the combination using TAK-228 (mTORC1 and 2 inhibitor) and knockdown of the different mTOR complex components. T-DM1 did not affect mTOR downstream signaling nor induct autophagy. Importantly, mTOR inhibition increased intracellular T-DM1 levels, leading to increased lysosomal accumulation of the compound. The increased efficacy of mTOR inhibition plus T-DM1 was abrogated by lysosome inhibitors (chloroquine and bafilomycin A1). Our experiments suggest that BT474 are less sensitive to T-DM1 due to lack of optimal lysosomal processing and intrinsic resistance to the DM1 moiety. Finally, we performed several in vivo experiments that corroborated the superior activity of T-DM1 and everolimus in HCC1954 and PDX-derived mouse models. In summary, everolimus in combination with T-DM1 showed strong antitumor effects in HER2-positive breast cancer, both in vitro and in vivo. This effect might be related, at least partially, to mTOR-dependent lysosomal processing of T-DM1, a finding that might apply to other ADCs that require lysosomal processing. Implications: Inhibition of mTOR increases the anti-tumor activity of T-DM1, supporting that the combination of mTOR inhibitors and antibody-drug conjugates warrants clinical evaluation in patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
| | | | | | - Oriol Arpi-Llucia
- IMIM (Institut Hospital del Mar d'Investigacions M�diques), Barcelona, Barcelona, Spain
| | | | - Mengjuan Qin
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Cristina Guardia
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Barcelona, Spain
| | | | | | - Sandra Zazo
- IIS-Fundaci�n Jim�nez D�az-CIBERONC, UAM, Madrid, Spain
| | | | | | - Pilar Eroles
- INCLIVA Biomedical Research Institute - University of Valencia, Spain. CIBERONC, Valencia, Valencia, Spain
| | - Juan Miguel Cejalvo
- Biomedical Health Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Ana Lluch
- University of Valencia - Biomedical Research Institute INCLIVA-Hospital Cl�nico de Valencia-CIBERONC, Valencia, Valencia, Spain
| | - Federico Rojo
- IIS-Fundaci�n Jim�nez D�az-CIBERONC, UAM, Madrid, Spain
| | - Atanasio Pandiella
- Centro de Investigaci�n del C�ncer, CIBERONC and IBSAL, Salamanca, Spain
| | | | - Joan Albanell
- Hospital Del Mar Medical Research Instiiute, Barcelona, Spain
| |
Collapse
|
32
|
Zhou Z, Zhang C, Ma Z, Wang H, Tuo B, Cheng X, Liu X, Li T. Pathophysiological role of ion channels and transporters in HER2-positive breast cancer. Cancer Gene Ther 2022; 29:1097-1104. [PMID: 34997219 DOI: 10.1038/s41417-021-00407-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/21/2021] [Accepted: 11/08/2021] [Indexed: 11/09/2022]
Abstract
The incidence of breast cancer (BC) has been increasing each year, and BC is now the most common malignant tumor in women. Among the numerous BC subtypes, HER2-positive BC can be treated with a variety of strategies based on targeting HER2. Although there has been great progress in the treatment of HER2-positive BC, recurrence, metastasis and drug resistance remain considerable challenges. The dysfunction of ion channels and transporters can affect the development and progression of HER2-positive BC, so these entities are expected to be new therapeutic targets. This review summarizes various ion channels and transporters associated with HER2-positive BC and suggests potential targets for the development of new and effective therapies.
Collapse
Affiliation(s)
- Zhengxing Zhou
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Chengmin Zhang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Hu Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Xiaoming Cheng
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China
| | - Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China.
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province, China.
| |
Collapse
|
33
|
Mishra A, Hourigan D, Lindsay AJ. Inhibition of the endosomal recycling pathway downregulates HER2 activation and overcomes resistance to tyrosine kinase inhibitors in HER2-positive breast cancer. Cancer Lett 2022; 529:153-167. [PMID: 35007696 DOI: 10.1016/j.canlet.2022.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
The development of HER2-targeted therapies has led to a dramatic improvement in outcomes for breast cancer patients. However, nearly all patients with metastatic HER2-positive breast cancer will eventually progress on these therapies due to innate or acquired resistance. Recent evidence suggests that the endosomal recycling of HER2 plays an important role in regulating its oncogenic signalling. Here we report that the expression of Rab coupling protein (RCP), a key regulator of endosomal recycling, positively correlates with that of HER2 and HER3 in breast tumours, and high RCP expression is predictive of poor relapse-free and overall survival in patients with HER2-amplified breast cancer. Chemical and genetic inhibition of endosomal recycling leads to a reduction in the total cellular levels of HER2 and HER3 and inhibits the activation of their downstream signalling pathways. We find that HER2 and HER3 that have been internalised from the plasma membrane are diverted to lysosomes for degradation when endosomal recycling is blocked. Primaquine (PQ), a small molecule inhibitor of the endosomal recycling pathway, synergises with HER2-targeting tyrosine kinase inhibitors and overcomes innate and acquired resistance to these TKIs. Moreover, TKI-induced drug tolerant persister cells are vulnerable to endosomal recycling inhibitors. These findings suggest that inhibition of endosomal recycling represents a promising therapeutic strategy for treating drug resistant HER2-positive breast cancer.
Collapse
Affiliation(s)
- Anurag Mishra
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| | - David Hourigan
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| | - Andrew J Lindsay
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland.
| |
Collapse
|
34
|
Targeted Therapy Modulates the Secretome of Cancer-Associated Fibroblasts to Induce Resistance in HER2-Positive Breast Cancer. Int J Mol Sci 2021; 22:ijms222413297. [PMID: 34948097 PMCID: PMC8706990 DOI: 10.3390/ijms222413297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
The combination of trastuzumab plus pertuzumab plus docetaxel as a first-line therapy in patients with HER2-positive metastatic breast cancer has provided significant clinical benefits compared to trastuzumab plus docetaxel alone. However, despite the therapeutic success of existing therapies targeting HER2, tumours invariably relapse. Therefore, there is an urgent need to improve our understanding of the mechanisms governing resistance, so that specific therapeutic strategies can be developed to provide improved efficacy. It is well known that the tumour microenvironment (TME) has a significant impact on cancer behaviour. Cancer-associated fibroblasts (CAFs) are essential components of the tumour stroma that have been linked to acquired therapeutic resistance and poor prognosis in breast cancer. For this reason, it would be of interest to identify novel biomarkers in the tumour stroma that could emerge as therapeutic targets for the modulation of resistant phenotypes. Conditioned medium experiments carried out in our laboratory with CAFs derived from HER2-positive patients showed a significant capacity to promote resistance to trastuzumab plus pertuzumab therapies in two HER2-positive breast cancer cell lines (BCCLs), even in the presence of docetaxel. In order to elucidate the components of the CAF-conditioned medium that may be relevant in the promotion of BCCL resistance, we implemented a multiomics strategy to identify cytokines, transcription factors, kinases and miRNAs in the secretome that have specific targets in cancer cells. The combination of cytokine arrays, label-free LC-MS/MS quantification and miRNA analysis to explore the secretome of CAFs under treatment conditions revealed several up- and downregulated candidates. We discuss the potential role of some of the most interesting candidates in generating resistance in HER2-positive breast cancer.
Collapse
|
35
|
Zhang C, Röder J, Scherer A, Bodden M, Pfeifer Serrahima J, Bhatti A, Waldmann A, Müller N, Oberoi P, Wels WS. Bispecific antibody-mediated redirection of NKG2D-CAR natural killer cells facilitates dual targeting and enhances antitumor activity. J Immunother Cancer 2021; 9:jitc-2021-002980. [PMID: 34599028 PMCID: PMC8488744 DOI: 10.1136/jitc-2021-002980] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2021] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Natural killer group 2D (NKG2D) is an activating receptor of natural killer (NK) cells and other lymphocytes that mediates lysis of malignant cells through recognition of stress-induced ligands such as MICA and MICB. Such ligands are broadly expressed by cancer cells of various origins and serve as targets for adoptive immunotherapy with effector cells endogenously expressing NKG2D or carrying an NKG2D-based chimeric antigen receptor (CAR). However, shedding or downregulation of NKG2D ligands (NKG2DL) can prevent NKG2D activation, resulting in escape of cancer cells from NKG2D-dependent immune surveillance. METHODS To enable tumor-specific targeting of NKG2D-expressing effector cells independent of membrane-anchored NKG2DLs, we generated a homodimeric recombinant antibody which harbors an N-terminal single-chain fragment variable (scFv) antibody domain for binding to NKG2D, linked via a human IgG4 Fc region to a second C-terminal scFv antibody domain for recognition of the tumor-associated antigen ErbB2 (HER2). The ability of this molecule, termed NKAB-ErbB2, to redirect NKG2D-expressing effector cells to ErbB2-positive tumor cells of different origins was investigated using peripheral blood mononuclear cells, ex vivo expanded NK cells, and NK and T cells engineered with an NKG2D-based chimeric receptor. RESULTS On its own, bispecific NKAB-ErbB2 increased lysis of ErbB2-positive breast carcinoma cells by peripheral blood-derived NK cells endogenously expressing NKG2D more effectively than an ErbB2-specific IgG1 mini-antibody able to induce antibody-dependent cell-mediated cytotoxicity via activation of CD16. Furthermore, NKAB-ErbB2 synergized with NK-92 cells or primary T cells engineered to express an NKG2D-CD3ζ chimeric antigen receptor (NKAR), leading to targeted cell killing and greatly enhanced antitumor activity, which remained unaffected by soluble MICA known as an inhibitor of NKG2D-mediated natural cytotoxicity. In an immunocompetent mouse glioblastoma model mimicking low or absent NKG2DL expression, the combination of NKAR-NK-92 cells and NKAB-ErbB2 effectively suppressed outgrowth of ErbB2-positive tumors, resulting in treatment-induced endogenous antitumor immunity and cures in the majority of animals. CONCLUSIONS Our results demonstrate that combining an NKAB antibody with effector cells expressing an activating NKAR receptor represents a powerful and versatile approach to simultaneously enhance tumor antigen-specific as well as NKG2D-CAR and natural NKG2D-mediated cytotoxicity, which may be particularly useful to target tumors with heterogeneous target antigen expression.
Collapse
Affiliation(s)
- Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Jasmin Röder
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Anne Scherer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Malena Bodden
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | | | - Anita Bhatti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Anja Waldmann
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Nina Müller
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Pranav Oberoi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany .,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| |
Collapse
|
36
|
Woo J, Kim JB, Cho T, Yoo EH, Moon BI, Kwon H, Lim W. Selenium inhibits growth of trastuzumab-resistant human breast cancer cells via downregulation of Akt and beclin-1. PLoS One 2021; 16:e0257298. [PMID: 34525121 PMCID: PMC8443054 DOI: 10.1371/journal.pone.0257298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 08/27/2021] [Indexed: 01/01/2023] Open
Abstract
The response rate to treatment with trastuzumab (Tz), a recombinant humanized anti-HER2 monoclonal antibody, is only 12–34% despite demonstrated effectiveness on improving the survival of patients with HER2-positive breast cancers. Selenium has an antitumor effect against cancer cells and can play a cytoprotective role on normal cells. This study investigated the effect of selenium on HER2-positive breast cancer cells and the mechanism in relation to the response of the cells to Tz. HER2-positive breast cancer cell lines, SK-BR-3 as trastuzumab-sensitive cells, and JIMT-1 as Tz-resistant cells were treated with Tz and sodium selenite (selenite). Cell survival rates and expression of Her2, Akt, and autophagy-related proteins, including LC3B and beclin 1, in both cell lines 72 h after treatment were evaluated. Significant cell death was induced at different concentrations of selenite in both cell lines. A combined effect of selenite and Tz at 72 h was similar to or significantly greater than each drug alone. The expression of phosphorylated Akt (p-Akt) was decreased in JIMT-1 after combination treatment compared to that after only Tz treatment, while p-Akt expression was increased in SK-BR-3. The expression of beclin1 increased particularly in JIMT-1 after only Tz treatment and was downregulated by combination treatment. These results showed that combination of Tz and selenite had an antitumor effect in Tz-resistant breast cancer cells through downregulation of phosphorylated Akt and beclin1-related autophagy. Selenite might be a potent drug to treat Tz-resistant breast cancer by several mechanisms.
Collapse
Affiliation(s)
- Joohyun Woo
- Department of Surgery, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Jong Bin Kim
- Ewha Institute of Convergence Medicine, Seoul, South Korea
| | - Taeeun Cho
- Ewha Institute of Convergence Medicine, Seoul, South Korea
| | - Eun Hye Yoo
- Ewha Institute of Convergence Medicine, Seoul, South Korea
| | - Byung-In Moon
- Department of Surgery, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Hyungju Kwon
- Department of Surgery, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Woosung Lim
- Department of Surgery, Ewha Womans University College of Medicine, Seoul, South Korea
- * E-mail:
| |
Collapse
|
37
|
Huynh TK, Huang CH, Chen JY, Yao JH, Yang YS, Wei YL, Chen HF, Chen CH, Tu CY, Hsu YM, Liu LC, Huang WC. MiR-221 confers lapatinib resistance by negatively regulating p27 kip1 in HER2-positive breast cancer. Cancer Sci 2021; 112:4234-4245. [PMID: 34382727 PMCID: PMC8486195 DOI: 10.1111/cas.15107] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 12/02/2022] Open
Abstract
Development of acquired resistance to lapatinib, a dual epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) tyrosine kinase inhibitor, severely limits the duration of clinical response in advanced HER2‐driven breast cancer patients. Although the compensatory activation of the PI3K/Akt survival signal has been proposed to cause acquired lapatinib resistance, comprehensive molecular mechanisms remain required to develop more efficient strategies to circumvent this therapeutic difficulty. In this study, we found that suppression of HER2 by lapatinib still led to Akt inactivation and elevation of FOX3a protein levels, but failed to induce the expression of their downstream pro‐apoptotic effector p27kip1, a cyclin‐dependent kinase inhibitor. Elevation of miR‐221 was found to contribute to the development of acquired lapatinib resistance by targeting p27kip1 expression. Furthermore, upregulation of miR‐221 was mediated by the lapatinib‐induced Src family tyrosine kinase and subsequent NF‐κB activation. The reversal of miR‐221 upregulation and p27kip1 downregulation by a Src inhibitor, dasatinib, can overcome lapatinib resistance. Our study not only identified miRNA‐221 as a pivotal factor conferring the acquired resistance of HER2‐positive breast cancer cells to lapatinib through negatively regulating p27kip1 expression, but also suggested Src inhibition as a potential strategy to overcome lapatinib resistance.
Collapse
Affiliation(s)
- Thanh Kieu Huynh
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Chih-Hao Huang
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan.,Division of Breast Surgery, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Jhen-Yu Chen
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan
| | - Jin-Han Yao
- School of Medicine, China Medical University, Taichung, 404, Taiwan
| | - Yi-Shiang Yang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Ya-Ling Wei
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Hsiao-Fan Chen
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Chia-Hung Chen
- School of Medicine, China Medical University, Taichung, 404, Taiwan.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Chih-Yen Tu
- School of Medicine, China Medical University, Taichung, 404, Taiwan.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan.,Department of Animal Science and Technology, Agriculture College, Tunghai University, Taichung, 40704, Taiwan
| | - Liang-Chih Liu
- Division of Breast Surgery, China Medical University Hospital, Taichung, 40402, Taiwan.,School of Medicine, China Medical University, Taichung, 404, Taiwan
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan.,The Ph.D. program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, 404, Taiwan.,Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| |
Collapse
|
38
|
Gameiro A, Urbano AC, Ferreira F. Emerging Biomarkers and Targeted Therapies in Feline Mammary Carcinoma. Vet Sci 2021; 8:164. [PMID: 34437486 PMCID: PMC8402877 DOI: 10.3390/vetsci8080164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/19/2022] Open
Abstract
Feline mammary carcinoma (FMC) is a common aggressive malignancy with a low survival rate that lacks viable therapeutic options beyond mastectomy. Recently, increasing efforts have been made to understand the molecular mechanisms underlying FMC development, using the knowledge gained from studies on human breast cancer to discover new diagnostic and prognostic biomarkers, thus reinforcing the utility of the cat as a cancer model. In this article, we review the current knowledge on FMC pathogenesis, biomarkers, and prognosis factors and offer new insights into novel therapeutic options for HER2-positive and triple-negative FMC subtypes.
Collapse
Affiliation(s)
| | | | - Fernando Ferreira
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; (A.G.); (A.C.U.)
| |
Collapse
|
39
|
UCP-2 inhibitor enhanced the efficacy of trastuzumab against HER2 positive breast cancer cells. Cancer Chemother Pharmacol 2021; 88:633-642. [PMID: 34146128 PMCID: PMC8367901 DOI: 10.1007/s00280-021-04303-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 05/30/2021] [Indexed: 01/09/2023]
Abstract
Purpose This study aimed to investigate the possibility of UCP-2 inhibitor in reducing acquired resistance of trastuzumab to improve the outcome of patients receiving trastuzumab therapy by exploring the relationship between UCP-2 expression and HER2 signaling pathway and examining whether UCP-2 expression was modulated by trastuzumab treatment. Methods 32 women diagnosed with primary HER2-positive breast cancer were recruited in this study. Needle biopsy was obtained from patients before they received at least four cycles neoadjuvant therapy containing trastuzumab in combination with chemotherapy. Surgical tumor biopsy was obtained during surgical procedure after the neoadjuvant therapy. Levels of HER2 phosphorylation and UCP-2 expression were detected by immunohistochemistry (IHC) and compared between tumor needle biopsy tissue and surgical tumor samples of these patients, as well as in BT474 breast cancer cells before and after trastuzumab treatment. HER2-selective phosphorylation/kinase activity inhibitor ONT-380 was used to identify the correlation between HER2 phosphorylation level and UCP-2 expression. UCP-2 inhibitor Genipin was then used to evaluate the apoptosis index in BT474 cells treated with trastuzumab. Results UCP-2 expression was significantly elevated in surgical tumor samples from breast cancer patients receiving trastuzumab in a neoadjuvant setting. We further confirmed our findings in HER2-positive BT474 cell line and found that trastuzumab treatment induced phosphorylation of HER2 and the overexpression of UCP-2, and the latter can be reversed by HER2 selective kinase inhibitor ONT-380. Moreover, UCP-2 inhibitor Genipin significantly enhanced the proliferation suppression effects of trastuzumab and markedly promoted apoptosis. Conclusion Taken together, our study identified UCP-2 as a novel therapeutic target for HER2 positive breast cancer and UCP-2 inhibitor may have great potential to enhance the response rate and efficacy of trastuzumab therapy.
Collapse
|
40
|
Wehrenberg-Klee E, Sinevici N, Nesti S, Kalomeris T, Austin E, Larimer B, Mahmood U. HER3 PET Imaging Identifies Dynamic Changes in HER3 in Response to HER2 Inhibition with Lapatinib. Mol Imaging Biol 2021; 23:930-940. [PMID: 34101105 DOI: 10.1007/s11307-021-01619-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/30/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Standard therapy for HER2+ breast cancers includes HER2 inhibition. While HER2 inhibitors have significantly improved therapeutic outcomes, many patients remain resistant to therapy. An important intrinsic resistance mechanism to HER2 inhibition in some breast cancers is dynamic upregulation of HER3. Increase in HER3 expression that occurs in response to HER2 inhibition allows for continued growth signaling through HER2/HER3 heterodimers, promoting tumor escape. We hypothesized that a non-invasive method to image changes in HER3 expression would be valuable to identify those breast cancers that dynamically upregulate HER3 in response to HER2 inhibition. We further hypothesized that this imaging method could identify those tumors that would benefit by additional HER3 knockdown. PROCEDURES In a panel of HER2+ breast cancer cell lines treated with the HER2 inhibitor lapatinib, we evaluate changes in HER3 expression and viability. Mouse HER2+ breast cancer models treated with lapatinib were imaged with a peptide-based HER3-specific PET imaging agent [68Ga]HER3P1 to assess for dynamic changes in tumoral HER3 expression and uptake confirmed by biodistribution. Subsequently, HER2+ cell lines were treated with the HER2 inhibitor lapatinib as well HER3-specific siRNA to assess for changes in viability and correlate with HER3 expression upregulation. For all statistical comparisons, P<0.05 was considered statistically significant. RESULTS Lapatinib treatment of a panel of HER2+ breast cancer cell lines increased HER3 expression in the lapatinib-resistant cell line MDA-MB 453 but not the lapatinib-resistant cell-line HCC-1569. Evaluation of [68Ga]HER3P1 uptake in mice implanted with the HER2+ breast cancer cell lines MDA-MB453 or HCC-1569 prior to and after treatment with lapatinib demonstrated a significant increase in MDA-MB453 tumors only, consistent with in vitro findings. The additional knockdown of HER3 increased therapeutic efficacy of lapatinib only in MDA-MB453 cells, but not in HCC-1569 cells. CONCLUSION HER3 PET imaging can be used to visualize dynamic changes in HER3 expression that occur in HER2+ breast cancers with HER2 inhibitor treatment and identify those likely to benefit by the addition of combination HER3 and HER2 inhibition.
Collapse
Affiliation(s)
- Eric Wehrenberg-Klee
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Suite 5.407, Boston, MA, 02129, USA
| | - Nicoleta Sinevici
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Suite 5.407, Boston, MA, 02129, USA
| | - Sarah Nesti
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Suite 5.407, Boston, MA, 02129, USA
| | - Taylor Kalomeris
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Suite 5.407, Boston, MA, 02129, USA
| | - Emily Austin
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Suite 5.407, Boston, MA, 02129, USA
| | - Benjamin Larimer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Suite 5.407, Boston, MA, 02129, USA
| | - Umar Mahmood
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Suite 5.407, Boston, MA, 02129, USA.
| |
Collapse
|
41
|
Sanz-Álvarez M, Martín-Aparicio E, Luque M, Zazo S, Martínez-Useros J, Eroles P, Rovira A, Albanell J, Madoz-Gúrpide J, Rojo F. The Novel Oral mTORC1/2 Inhibitor TAK-228 Reverses Trastuzumab Resistance in HER2-Positive Breast Cancer Models. Cancers (Basel) 2021; 13:cancers13112778. [PMID: 34204960 PMCID: PMC8199905 DOI: 10.3390/cancers13112778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Hyperactivation of the PI3K/AKT/mTOR cell signalling pathway is an important and well-described mechanism of trastuzumab resistance in HER2-positive breast cancer. In cell-line models of acquired trastuzumab resistance generated in our laboratory, we demonstrate this type of activation, which is independent of HER2-mediated regulation. We investigate whether the use of specific mTOR inhibitors, a PI3K/AKT/mTOR pathway effector, could lead to decreased activity of the pathway, influencing trastuzumab resistance. We demonstrate that TAK-228, a mTORC1 and mTORC2 inhibitor, can reverse resistance and increasing response to trastuzumab in models of primary and acquired resistance. Abstract The use of anti-HER2 therapies has significantly improved clinical outcome in patients with HER2-positive breast cancer, yet a substantial proportion of patients acquire resistance after a period of treatment. The PI3K/AKT/mTOR pathway is a good target for drug development, due to its involvement in HER2-mediated signalling and in the emergence of resistance to anti-HER2 therapies, such as trastuzumab. This study evaluates the activity of three different PI3K/AKT/mTOR inhibitors, i.e., BEZ235, everolimus and TAK-228 in vitro, in a panel of HER2-positive breast cancer cell lines with primary and acquired resistance to trastuzumab. We assess the antiproliferative effect and PI3K/AKT/mTOR inhibitory capability of BEZ235, everolimus and TAK-228 alone, and in combination with trastuzumab. Dual blockade with trastuzumab and TAK-228 was superior in reversing the acquired resistance in all the cell lines. Subsequently, we analyse the effects of TAK-228 in combination with trastuzumab on the cell cycle and found a significant increase in G0/G1 arrest in most cell lines. Likewise, the combination of both drugs induced a significant increase in apoptosis. Collectively, these experiments support the combination of trastuzumab with PI3K/AKT/mTOR inhibitors as a potential strategy for inhibiting the proliferation of HER2-positive breast cancer cell lines that show resistance to trastuzumab.
Collapse
Affiliation(s)
- Marta Sanz-Álvarez
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Ester Martín-Aparicio
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Melani Luque
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Sandra Zazo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain;
| | - Pilar Eroles
- Institute of Health Research INCLIVA-CIBERONC, 46010 Valencia, Spain;
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain; (A.R.); (J.A.)
- Medical Oncology Department, Hospital del Mar-CIBERONC, 08003 Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain; (A.R.); (J.A.)
- Medical Oncology Department, Hospital del Mar-CIBERONC, 08003 Barcelona, Spain
- Department of Experimental and Health Sciences, Faculty of Medicine, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Juan Madoz-Gúrpide
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
- Correspondence: (J.M.-G.); (F.R.); Tel.: +34-915-504-800 (J.M.-G.); +34-915-504-800 (F.R.)
| | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
- Correspondence: (J.M.-G.); (F.R.); Tel.: +34-915-504-800 (J.M.-G.); +34-915-504-800 (F.R.)
| |
Collapse
|
42
|
Fararjeh AFS, Al Khader A, Kaddumi E, Obeidat M, Al-Fawares O. Differential Expression and Prognostic Significance of STARD3 Gene in Breast Carcinoma. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:34-41. [PMID: 34268252 PMCID: PMC8256830 DOI: 10.22088/ijmcm.bums.10.1.34] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/21/2021] [Indexed: 12/28/2022]
Abstract
StAR related lipid transfer domain containing 3 (STARD3) gene has been reported to be co-amplified with human epidermal growth factor receptor 2 (HER2) in breast carcinoma. STARD3 is necessary for cholesterol transfer and metabolism in tumor cells. The possible role played by STARD3 as a diagnostic and prognostic biomarker was investigated in breast cancer (BC). Data mining was performed using several bioinformatics websites to investigate the correlation of STARD3 with BC and its molecular subtypes, and conventional PCR was used to detect the STARD3 mRNA levels in a panel of BC cell lines. STARD3 was overexpressed in BC more than the other types of cancer. The results also showed that STARD3 expression was significantly associated with HER2+ BC tumors and BC cell lines, and low STARD3 mRNA and protein expression levels were observed in estrogen receptor-positive (ER+) and triple-negative BC (TNBC) patients. Moreover, high STARD3 expression levels predicted worse overall survival (OS), relapse-free survival (RFS) and disease metastasis-free survival (DMFS) in BC, and HER2+ BC. Notably, low expression of STARD3 was associated with poor OS in ER+ BC. Our findings suggest that STARD3 may have strong diagnostic and prognostic value for HER2+ breast carcinoma.
Collapse
Affiliation(s)
| | - Ali Al Khader
- Department of Pathology and Forensic Medicine, Faculty of Medicine, Al-Balqa Applied University, Al-salt, Jordan.,Department of Pathology, Al-Hussein Salt Hospital, Al-salt, Jordan
| | - Ezidin Kaddumi
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-salt, Jordan
| | - Maher Obeidat
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-salt, Jordan
| | - O'la Al-Fawares
- Department of Medical Laboratory Analysis, Faculty of Science, Al-Balqa Applied University, Al-salt, Jordan
| |
Collapse
|
43
|
Mezynski MJ, Farrelly AM, Cremona M, Carr A, Morgan C, Workman J, Armstrong P, McAuley J, Madden S, Fay J, Sheehan KM, Kay EW, Holohan C, Elamin Y, Rafee S, Morris PG, Breathnach O, Grogan L, Hennessy BT, Toomey S. Targeting the PI3K and MAPK pathways to improve response to HER2-targeted therapies in HER2-positive gastric cancer. J Transl Med 2021; 19:184. [PMID: 33933113 PMCID: PMC8088633 DOI: 10.1186/s12967-021-02842-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/18/2021] [Indexed: 12/24/2022] Open
Abstract
Background Aberrant PI3K signalling is implicated in trastuzumab resistance in HER2-positive gastric cancer (GC). The role of PI3K or MEK inhibitors in sensitising HER2-positive GCs to trastuzumab or in overcoming trastuzumab resistance is unclear. Methods Using mass spectrometry-based genotyping we analysed 105 hotspot, non-synonymous somatic mutations in PIK3CA and ERBB-family (EGFR, ERBB2, ERBB3 and ERBB4) genes in gastric tumour samples from 69 patients. A panel of gastric cell lines (N87, OE19, ESO26, SNU16, KATOIII) were profiled for anti-proliferative response to the PI3K inhibitor copanlisib and the MEK1/2 inhibitor refametinib alone and in combination with anti-HER2 therapies. Results Patients with HER2-positive GC had significantly poorer overall survival compared to HER2-negative patients (15.9 months vs. 35.7 months). Mutations in PIK3CA were only identified in HER2-negative tumours, while ERBB-family mutations were identified in HER2-positive and HER2-negative tumours. Copanlisib had anti-proliferative effects in 4/5 cell lines, with IC50s ranging from 23.4 (N87) to 93.8 nM (SNU16). All HER2-positive cell lines except SNU16 were sensitive to lapatinib (IC50s 0.04 µM–1.5 µM). OE19 cells were resistant to trastuzumab. The combination of lapatinib and copanlisib was synergistic in ESO-26 and OE-19 cells (ED50: 0.83 ± 0.19 and 0.88 ± 0.13, respectively) and additive in NCI-N87 cells (ED50:1.01 ± 0.55). The combination of copanlisib and trastuzumab significantly improved growth inhibition compared to either therapy alone in NCI-N87, ESO26 and OE19 cells (p < 0.05). Conclusions PI3K or MEK inhibition alone or in combination with anti-HER2 therapy may represent an improved treatment strategy for some patients with HER2-positive GC, and warrants further investigation in a clinical trial setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02842-1.
Collapse
Affiliation(s)
- M Janusz Mezynski
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Angela M Farrelly
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Mattia Cremona
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Aoife Carr
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Clare Morgan
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Julie Workman
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Paul Armstrong
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Jennifer McAuley
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Joanna Fay
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Katherine M Sheehan
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elaine W Kay
- Department of Histopathology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ciara Holohan
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Yasir Elamin
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Shereen Rafee
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Patrick G Morris
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Oscar Breathnach
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Liam Grogan
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland.,Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Smurfit Building, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
44
|
Orfanou IM, Argyros O, Papapetropoulos A, Tseleni-Balafouta S, Vougas K, Tamvakopoulos C. Discovery and Pharmacological Evaluation of STEAP4 as a Novel Target for HER2 Overexpressing Breast Cancer. Front Oncol 2021; 11:608201. [PMID: 33842315 PMCID: PMC8034292 DOI: 10.3389/fonc.2021.608201] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 03/08/2021] [Indexed: 01/11/2023] Open
Abstract
Breast cancer (BC) is a highly heterogeneous disease encompassing multiple subtypes with different molecular and histopathological features, disease prognosis, and therapeutic responses. Among these, the Triple Negative BC form (TNBC) is an aggressive subtype with poor prognosis and therapeutic outcome. With respect to HER2 overexpressing BC, although advanced targeted therapies have improved the survival of patients, disease relapse and metastasis remains a challenge for therapeutic efficacy. In this study the aim was to identify key membrane-associated proteins which are overexpressed in these aggressive BC subtypes and can serve as potential biomarkers or drug targets. We leveraged on the development of a membrane enrichment protocol in combination with the global profiling GeLC-MS/MS technique, and compared the proteomic profiles of a HER2 overexpressing (HCC-1954) and a TNBC (MDA-MB-231) cell line with that of a benign control breast cell line (MCF-10A). An average of 2300 proteins were identified from each cell line, of which approximately 600 were membrane-associated proteins. Our global proteomic methodology in tandem with invigoration by Western blot and Immunofluorescence analysis, readily detected several previously-established BC receptors like HER2 and EPHA2, but importantly STEAP4 and CD97 emerged as novel potential candidate markers. This is the first time that the mitochondrial iron reductase STEAP4 protein up-regulation is linked to BC (HER2+ subtype), while for CD97, its role in BC has been previously described, but never before by a global proteomic technology in TNBC. STEAP4 was selected for further detailed evaluation by the employment of Immunohistochemical analysis of BC xenografts and clinical tissue microarray studies. Results showed that STEAP4 expression was evident only in malignant breast tissues whereas all the benign breast cases had no detectable levels. A functional role of STEAP4 intervention was established in HER2 overexpressing BC by pharmacological studies, where blockage of the STEAP4 pathway with an iron chelator (Deferiprone) in combination with the HER2 inhibitor Lapatinib led to a significant reduction in cell growth in vitro. Furthermore, siRNA mediated knockdown of STEAP4 also suppressed cell proliferation and enhanced the inhibition of Lapatinib in HER2 overexpressing BC, confirming its potential oncogenic role in BC. In conclusion, STEAP4 may represent a novel BC related biomarker and a potential pharmacological target for the treatment of HER2 overexpressing BC.
Collapse
Affiliation(s)
- Ioanna-Maria Orfanou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Orestis Argyros
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Tseleni-Balafouta
- Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Vougas
- Proteomics Laboratory, Division of Biotechnology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Constantin Tamvakopoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
45
|
Kumagai S, Koyama S, Nishikawa H. Antitumour immunity regulated by aberrant ERBB family signalling. Nat Rev Cancer 2021; 21:181-197. [PMID: 33462501 DOI: 10.1038/s41568-020-00322-0] [Citation(s) in RCA: 147] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 01/30/2023]
Abstract
Aberrant signalling of ERBB family members plays an important role in tumorigenesis and in the escape from antitumour immunity in multiple malignancies. Molecular-targeted agents against these signalling pathways exhibit robust clinical efficacy, but patients inevitably experience acquired resistance to these molecular-targeted therapies. Although cancer immunotherapies, including immune checkpoint inhibitors (ICIs), have shown durable antitumour response in a subset of the treated patients in multiple cancer types, clinical efficacy is limited in cancers harbouring activating gene alterations of ERBB family members. In particular, ICI treatment of patients with non-small cell lung cancers with epidermal growth factor receptor (EGFR) alterations and breast cancers with HER2 alterations failed to show clinical benefits, suggesting that EGFR and HER2 signalling may have an essential role in inhibiting antitumour immune responses. Here, we discuss the mechanisms by which the signalling of ERBB family members affects not only autonomous cancer hallmarks, such as uncontrolled cell proliferation, but also antitumour immune responses in the tumour microenvironment and the potential application of immune-genome precision medicine into immunotherapy and molecular-targeted therapy focusing on the signalling of ERBB family members.
Collapse
Affiliation(s)
- Shogo Kumagai
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo, Japan
- Division of Cancer Immunology, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba, Japan
| | - Shohei Koyama
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo, Japan
- Division of Cancer Immunology, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo, Japan.
- Division of Cancer Immunology, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba, Japan.
| |
Collapse
|
46
|
Conlon NT, Kooijman JJ, van Gerwen SJC, Mulder WR, Zaman GJR, Diala I, Eli LD, Lalani AS, Crown J, Collins DM. Comparative analysis of drug response and gene profiling of HER2-targeted tyrosine kinase inhibitors. Br J Cancer 2021; 124:1249-1259. [PMID: 33473169 PMCID: PMC8007737 DOI: 10.1038/s41416-020-01257-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Human epidermal growth factor 2 (HER2/ERBB2) is frequently amplified/mutated in cancer. The tyrosine kinase inhibitors (TKIs) lapatinib, neratinib, and tucatinib are FDA-approved for the treatment of HER2-positive breast cancer. Direct comparisons of the preclinical efficacy of the TKIs have been limited to small-scale studies. Novel biomarkers are required to define beneficial patient populations. METHODS In this study, the anti-proliferative effects of the three TKIs were directly compared using a 115 cancer cell line panel. Novel TKI response/resistance markers were identified through cross-analysis of drug response profiles with mutation, gene copy number and expression data. RESULTS All three TKIs were effective against HER2-amplified breast cancer models; neratinib showing the most potent activity, followed by tucatinib then lapatinib. Neratinib displayed the greatest activity in HER2-mutant and EGFR-mutant cells. High expression of HER2, VTCN1, CDK12, and RAC1 correlated with response to all three TKIs. DNA damage repair genes were associated with TKI resistance. BRCA2 mutations were correlated with neratinib and tucatinib response, and high expression of ATM, BRCA2, and BRCA1 were associated with neratinib resistance. CONCLUSIONS Neratinib was the most effective HER2-targeted TKI against HER2-amplified, -mutant, and EGFR-mutant cell lines. This analysis revealed novel resistance mechanisms that may be exploited using combinatorial strategies.
Collapse
Affiliation(s)
- Neil T Conlon
- National Institute of Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Jeffrey J Kooijman
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Suzanne J C van Gerwen
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Winfried R Mulder
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Guido J R Zaman
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Irmina Diala
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - Lisa D Eli
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - Alshad S Lalani
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - John Crown
- National Institute of Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Denis M Collins
- National Institute of Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| |
Collapse
|
47
|
Primary Trastuzumab Resistance After (Neo)adjuvant Trastuzumab-containing Treatment for Patients With HER2-positive Breast Cancer in Real-world Practice. Clin Breast Cancer 2021; 21:191-198. [PMID: 33549471 DOI: 10.1016/j.clbc.2020.09.003] [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: 02/21/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND It is difficult to define patients with primary trastuzumab resistance (PTR) after (neo)adjuvant trastuzumab with minimal data and understanding of the actual prevalence, prognosis, and potential treatment strategies in the real-world setting. PATIENTS AND METHODS The medical records of 1096 patients with human epidermal growth factor receptor 2-positive early-stage breast cancer who had received (neo)adjuvant trastuzumab-containing treatment from 2010 to 2016 in the Cancer Hospital and Chinese Academy of Medical Sciences were reviewed. PTR was defined as recurrence during adjuvant trastuzumab or within 12 months from their last (neo)adjuvant trastuzumab dosage. The cutoff date for data collection was September 1, 2018, with a median follow-up time of 46 months. RESULTS A total of 126 recurrences were observed, and 75 (6.8%; 75/1096) of them were categorized as PTR; the remaining were non-PTR. The prognosis of patients in the PTR group was much inferior to those in the non-PTR group (27 months vs. not reached; P < .01). As expected, patients with PTR did possess a much lower response rate to first-line trastuzumab-containing therapy (27.3% vs. 61.9%; P = .02). Subgroup analyses indicated that patients in the PTR group seemed to get little survival benefit from the reuse of trastuzumab compared with those without trastuzumab (26 months vs. 28 months; P = .80). However, in the non-PTR group, re-treatment with trastuzumab in the metastatic setting prolonged the survival of patients compared to those without trastuzumab (not reached vs. 34 months; P = .04). CONCLUSION This study verified the rationality of present definition of PTR after (neo)adjuvant trastuzumab. Patients with PTR did have a poor prognosis. Further research and clinical trials are required to establish the best treatment patterns for these patients.
Collapse
|
48
|
Collins DM, Madden SF, Gaynor N, AlSultan D, Le Gal M, Eustace AJ, Gately KA, Hughes C, Davies AM, Mahgoub T, Ballot J, Toomey S, O'Connor DP, Gallagher WM, Holmes FA, Espina V, Liotta L, Hennessy BT, O'Byrne KJ, Hasmann M, Bossenmaier B, O'Donovan N, Crown J. Effects of HER Family-targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer. Clin Cancer Res 2020; 27:807-818. [PMID: 33122343 DOI: 10.1158/1078-0432.ccr-20-2007] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/18/2020] [Accepted: 10/22/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Antibody-dependent cell-mediated cytotoxicity (ADCC) is one mechanism of action of the monoclonal antibody (mAb) therapies trastuzumab and pertuzumab. Tyrosine kinase inhibitors (TKIs), like lapatinib, may have added therapeutic value in combination with mAbs through enhanced ADCC activity. Using clinical data, we examined the impact of lapatinib on HER2/EGFR expression levels and natural killer (NK) cell gene signatures. We investigated the ability of three TKIs (lapatinib, afatinib, and neratinib) to alter HER2/immune-related protein levels in preclinical models of HER2-positive (HER2+) and HER2-low breast cancer, and the subsequent effects on trastuzumab/pertuzumab-mediated ADCC. EXPERIMENTAL DESIGN Preclinical studies (proliferation assays, Western blotting, high content analysis, and flow cytometry) employed HER2+ (SKBR3 and HCC1954) and HER2-low (MCF-7, T47D, CAMA-1, and CAL-51) breast cancer cell lines. NCT00524303 provided reverse phase protein array-determined protein levels of HER2/pHER2/EGFR/pEGFR. RNA-based NK cell gene signatures (CIBERSORT/MCP-counter) post-neoadjuvant anti-HER2 therapy were assessed (NCT00769470/NCT01485926). ADCC assays utilized flow cytometry-based protocols. RESULTS Lapatinib significantly increased membrane HER2 levels, while afatinib and neratinib significantly decreased levels in all preclinical models. Single-agent lapatinib increased HER2 or EGFR levels in 10 of 11 (91%) tumor samples. NK cell signatures increased posttherapy (P = 0.03) and associated with trastuzumab response (P = 0.01). TKI treatment altered mAb-induced NK cell-mediated ADCC in vitro, but it did not consistently correlate with HER2 expression in HER2+ or HER2-low models. The ADCC response to trastuzumab and pertuzumab combined did not exceed either mAb alone. CONCLUSIONS TKIs differentially alter tumor cell phenotype which can impact NK cell-mediated response to coadministered antibody therapies. mAb-induced ADCC response is relevant when rationalizing combinations for clinical investigation.
Collapse
Affiliation(s)
- Denis M Collins
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland.
| | - Stephen F Madden
- RCSI Division of Population Health Sciences, Royal College of Surgeons in Ireland, Beaux Lane House, Dublin, Ireland
| | - Nicola Gaynor
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Dalal AlSultan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland.,RCSI Division of Population Health Sciences, Royal College of Surgeons in Ireland, Beaux Lane House, Dublin, Ireland
| | - Marion Le Gal
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Alex J Eustace
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Kathy A Gately
- Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Clare Hughes
- Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Anthony M Davies
- Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Thamir Mahgoub
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Jo Ballot
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Sinead Toomey
- RCSI Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Education & Research Centre, Beaumont Hospital, Beaumont, Dublin, Ireland
| | - Darran P O'Connor
- Royal College of Surgeons in Ireland, School of Pharmacy & Biomolecular Science, Dublin, Ireland
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Frankie A Holmes
- Texas Oncology-Memorial Hermann Memorial City, US Oncology Research, Houston, -Texas
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Lance Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Bryan T Hennessy
- RCSI Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Education & Research Centre, Beaumont Hospital, Beaumont, Dublin, Ireland.,Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Kenneth J O'Byrne
- Princess Alexandra Hospital, Translational Research Institute and Queensland University of Technology, Brisbane, Queensland, Australia
| | - Max Hasmann
- Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Penzberg, Germany
| | | | - Norma O'Donovan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - John Crown
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland.,Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
49
|
Drucker A, Yoo BH, Khan IA, Choi D, Montermini L, Liu X, Jovanovic S, Younis T, Rosen KV. Trastuzumab-induced upregulation of a protein set in extracellular vesicles emitted by ErbB2-positive breast cancer cells correlates with their trastuzumab sensitivity. Breast Cancer Res 2020; 22:105. [PMID: 33023655 PMCID: PMC7541295 DOI: 10.1186/s13058-020-01342-2] [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] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/16/2020] [Indexed: 12/27/2022] Open
Abstract
Background ErbB2/HER2 oncoprotein often drives breast cancers (BCs) which are treated with the anti-ErbB2 antibody trastuzumab. The efficacy of trastuzumab-based metastatic BC therapies is routinely assessed by imaging studies. Trastuzumab typically becomes ineffective in the case of this disease and is then replaced by other drugs. Biomarkers of BC trastuzumab response could allow imaging studies and the switch to other drugs to occur earlier than is now possible. Moreover, bone-only BC metastases can be hard to measure, and biomarkers of their trastuzumab response could facilitate further treatment decisions. Such biomarkers are presently unavailable. In this study, we searched for proteins whose levels in BC cell-emitted extracellular vesicles (EVs) potentially correlate with BC trastuzumab sensitivity. Methods We isolated EVs from cultured trastuzumab-sensitive and trastuzumab-resistant human BC cells before and after trastuzumab treatment and characterized these EVs by nanoparticle tracking analysis and electron microscopy. We found previously that ErbB2 drives BC by downregulating a pro-apoptotic protein PERP. We now tested whether trastuzumab-induced PERP upregulation in EVs emitted by cultured human BC cells correlates with their trastuzumab sensitivity. We also used mass spectrometry to search for additional proteins whose levels in such EVs reflect BC cell trastuzumab sensitivity. Once we identified proteins whose EV levels correlate with this sensitivity in culture, we explored the feasibility of testing whether their levels in the blood EVs of trastuzumab-treated metastatic BC patients correlate with patients’ response to trastuzumab-based treatments. Results We found that neither trastuzumab nor acquisition of trastuzumab resistance by BC cells affects the size or morphology of EVs emitted by cultured BC cells. We established that EV levels of proteins PERP, GNAS2, GNA13, ITB1, and RAB10 correlate with BC cell trastuzumab response. Moreover, these proteins were upregulated during trastuzumab-based therapies in the blood EVs of a pilot cohort of metastatic BC patients that benefited from these therapies but not in those derived from patients that failed such treatments. Conclusions Upregulation of a protein set in EVs derived from cultured breast tumor cells correlates with tumor cell trastuzumab sensitivity. It is feasible to further evaluate these proteins as biomarkers of metastatic BC trastuzumab response.
Collapse
Affiliation(s)
- Arik Drucker
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Byong Hoon Yoo
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Iman Aftab Khan
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Dongsic Choi
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - Laura Montermini
- Research Institute of the McGill University Health Centre, Glen Site, McGill University, Montreal, QC, Canada
| | - Xiaoyang Liu
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Sanja Jovanovic
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Tallal Younis
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Kirill V Rosen
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada. .,Atlantic Research Centre, Rm C-304, CRC, 5849 University Avenue, PO Box 15000, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
50
|
Zervantonakis IK, Poskus MD, Scott AL, Selfors LM, Lin JR, Dillon DA, Pathania S, Sorger PK, Mills GB, Brugge JS. Fibroblast-tumor cell signaling limits HER2 kinase therapy response via activation of MTOR and antiapoptotic pathways. Proc Natl Acad Sci U S A 2020; 117:16500-16508. [PMID: 32601199 PMCID: PMC7368275 DOI: 10.1073/pnas.2000648117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite the implementation of multiple HER2-targeted therapies, patients with advanced HER2+ breast cancer ultimately develop drug resistance. Stromal fibroblasts represent an abundant cell type in the tumor microenvironment and have been linked to poor outcomes and drug resistance. Here, we show that fibroblasts counteract the cytotoxic effects of HER2 kinase-targeted therapy in a subset of HER2+ breast cancer cell lines and allow cancer cells to proliferate in the presence of the HER2 kinase inhibitor lapatinib. Fibroblasts from primary breast tumors, normal breast tissue, and lung tissue have similar protective effects on tumor cells via paracrine factors. This fibroblast-mediated reduction in drug sensitivity involves increased expression of antiapoptotic proteins and sustained activation of the PI3K/AKT/MTOR pathway, despite inhibition of the HER2 and the RAS-ERK pathways in tumor cells. HER2 therapy sensitivity is restored in the fibroblast cocultures by combination treatment with inhibitors of MTOR or the antiapoptotic proteins BCL-XL and MCL-1. Expression of activated AKT in tumor cells recapitulates the effects of fibroblasts resulting in sustained MTOR signaling and poor lapatinib response. Lapatinib sensitivity was not altered by fibroblasts in tumor cells that exhibited sustained MTOR signaling due to a strong gain-of-function PI3KCA mutation. These findings indicate that in addition to tumor cell-intrinsic mechanisms that cause constitutive PI3K/AKT/MTOR pathway activation, secreted factors from fibroblasts can maintain this pathway in the context of HER2 inhibition. Our integrated proteomic-phenotypic approach presents a strategy for the discovery of protective mechanisms in fibroblast-rich tumors and the design of rational combination therapies to restore drug sensitivity.
Collapse
Affiliation(s)
- Ioannis K Zervantonakis
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213;
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Matthew D Poskus
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213
| | - Alexis L Scott
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
| | - Shailja Pathania
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA 02125
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Gordon B Mills
- Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR 97239
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115;
| |
Collapse
|