1
|
Debbi K, Grellier N, Loganadane G, Boukhobza C, Mahé M, Cherif MA, Rida H, Gligorov J, Belkacemi Y. Interaction between Radiation Therapy and Targeted Therapies in HER2-Positive Breast Cancer: Literature Review, Levels of Evidence for Safety and Recommendations for Optimal Treatment Sequence. Cancers (Basel) 2023; 15:cancers15082278. [PMID: 37190205 DOI: 10.3390/cancers15082278] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Purpose: Over the past twenty years, anti-HER2 targeted therapies have proven to be a revolution in the management of human epidermal growth receptor 2 (HER2)-positive breast cancers. Anti-HER2 therapies administered alone or in combination with chemotherapy have been specifically studied. Unfortunately, the safety of anti-HER2 therapies in combination with radiation remains largely unknown. Thus, we propose a literature review of the risks and safety of combining radiotherapy with anti-HER2 therapies. We will focus on the benefit/risk rationale and try to understand the risk of toxicity in early-stage and advanced breast cancer. Methods: Research was carried out on the following databases: PubMed, EMBASE, ClinicalTrial.gov, Medline, and Web of Science for the terms "radiotherapy", "radiation therapy", "radiosurgery", "local ablative therapy", and "stereotactic", combined with "trastuzumab", "pertuzumab", "trastuzumab emtansine", "TDM-1", "T-Dxd", "trastuzumab deruxtecan", "tucatinib", "lapatinib", "immune checkpoint inhibitors", "atezolizumab", "pembrolizumab", "nivolumab", "E75 vaccine", "interferon", "anti-IL-2", "anti-IL 12", and "ADC". Results: Association of radiation and monoclonal antibodies such as trastuzumab and pertuzumab (with limited data) seems to be safe, with no excess risk of toxicity. Preliminary data with radiation and of antibody-drug conjugate of trastuzumab combined cytotoxic (trastuzumab emtansine, trastuzumab deruxtecan), given the underlying mechanism of action, suggest that one must be particularly cautious with the association. The safety of the combination of a tyrosine kinase inhibitor (lapatinib, tucatinib) and radiation remains under-studied. The available evidence suggests that checkpoint inhibitors can be safely administrated with radiation. Conclusions: HER2-targeting monoclonal antibodies and checkpoint inhibitors can be combined with radiation, apparently with no excess toxicities. Caution is required when associating radiation with TKI and antibody drugs, considering the limited evidence.
Collapse
Affiliation(s)
- Kamel Debbi
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
- Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, i-Biot, UPEC, 94000 Créteil, France
| | - Noémie Grellier
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Gokoulakrichenane Loganadane
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
- Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, i-Biot, UPEC, 94000 Créteil, France
| | - Chahrazed Boukhobza
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Mathilde Mahé
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Mohamed Aziz Cherif
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Hanan Rida
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Joseph Gligorov
- APHP-Medical Oncology Department, Institut Universitaire de Cancérologie, Sorbonne Université, 75013 Paris, France
| | - Yazid Belkacemi
- APHP-Radiation Oncology Department and Henri Mondor Breast Center, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
- Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, i-Biot, UPEC, 94000 Créteil, France
| |
Collapse
|
2
|
Zhu Z, Goel PN, Zheng C, Nagai Y, Lam L, Samanta A, Ji M, Zhang H, Greene MI. HED, a Human-Engineered Domain, Confers a Unique Fc-Binding Activity to Produce a New Class of Humanized Antibody-like Molecules. Int J Mol Sci 2023; 24:ijms24076477. [PMID: 37047449 PMCID: PMC10094569 DOI: 10.3390/ijms24076477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/15/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Our laboratory has identified and developed a unique human-engineered domain (HED) structure that was obtained from the human Alpha-2-macroglobulin receptor-associated protein based on the three-dimensional structure of the Z-domain derived from Staphylococcal protein A. This HED retains µM binding activity to the human IgG1CH2-CH3 elbow region. We determined the crystal structure of HED in association with IgG1’s Fc. This demonstrated that HED preserves the same three-bundle helix structure and Fc-interacting residues as the Z domain. HED was fused to the single chain variable fragment (scFv) of mAb 4D5 to produce an antibody-like protein capable of interacting with the p185Her2/neu ectodomain and the Fc of IgG. When further fused with murine IFN-γ (mIFN-γ) at the carboxy terminus, the novel species exhibited antitumor efficacy in vivo in a mouse model of human breast cancer. The HED is a novel platform for the therapeutic utilization of engineered proteins to alleviate human disease.
Collapse
Affiliation(s)
- Zhiqiang Zhu
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peeyush N. Goel
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA 19104, USA
| | - Cai Zheng
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yasuhiro Nagai
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lian Lam
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arabinda Samanta
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meiqing Ji
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongtao Zhang
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence: or (H.Z.); or (M.I.G.)
| | - Mark I. Greene
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence: or (H.Z.); or (M.I.G.)
| |
Collapse
|
3
|
Chen D, Zhu Q, Li T, Fan X, Lou Y, Zhang Y, Huang K, Sun H. KLF4 loss in hepatocellular carcinoma: Improving prognostic prediction and correlating immune infiltrates. Front Genet 2023; 14:1106952. [PMID: 36936440 PMCID: PMC10017851 DOI: 10.3389/fgene.2023.1106952] [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: 11/24/2022] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction: Although the molecular mechanisms of Krüpple-like factor 4 (KLF4) as a tumor suppressor in HCC tumorigenesis have been thoroughly examined, its clinical application in terms of precise prognostication and its influence on tumor immune microenvironment in patients with HCC require further investigation. Methods: Bioinformatics and immunohistochemistry (IHC) were used to validate KLF4 expressions in a tissue microarray (TMA) containing HCC samples. Using Cox regression models, independent prognostic factors were identified and employed in the development of nomograms. Decision curve analysis (DCA) demonstrated the superiority of the nomograms. GO and KEGG pathway analyses were applied to the functional study of KLF4. The GSVA program explored the link between KLF4 expression and tumor-infiltrating immune cells, and CAMOIP was used to construct KLF4 expression immune scores. Changes in immune-related gene markers were also investigated in relation to KLF4 expression. The association between immune cell infiltration and KLF4 expression was validated by IHC in TMA. Results: HCC was reported to have a notable depletion of KLF4. The absence of KLF4 was associated with advanced clinicopathological characteristics of HCC and predicted a bad prognosis for patients. Nomograms constructed using KLF4 expression, tumor differentiation, and TNM stage provided a more accurate prognostic assessment of HCC patients than TNM stage alone. KLF4 expression was associated with immunological-related functions, infiltration of macrophages, CD8+ T cells, and other immune cells, and elevation of immune checkpoints. Higher levels of CD8+ T cells and macrophage infiltration are associated with increased KLF4 expression in HCC TMA. Conclusion: KLF4 loss in HCC is a prognostic biomarker that influences the tumor immune microenvironment (TIME).
Collapse
Affiliation(s)
- Desheng Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Zhu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiewen Li
- Institution for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuhui Fan
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichao Lou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kejie Huang
- College of Information Science Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Hongcheng Sun
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Huober J, Barrios CH, Niikura N, Jarząb M, Chang YC, Huggins-Puhalla SL, Pedrini J, Zhukova L, Graupner V, Eiger D, Henschel V, Gochitashvili N, Lambertini C, Restuccia E, Zhang H. Atezolizumab With Neoadjuvant Anti-Human Epidermal Growth Factor Receptor 2 Therapy and Chemotherapy in Human Epidermal Growth Factor Receptor 2-Positive Early Breast Cancer: Primary Results of the Randomized Phase III IMpassion050 Trial. J Clin Oncol 2022; 40:2946-2956. [PMID: 35763704 PMCID: PMC9426828 DOI: 10.1200/jco.21.02772] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Combining standard of care (pertuzumab-trastuzumab [PH], chemotherapy) with cancer immunotherapy may potentiate antitumor immunity, cytotoxic activity, and patient outcomes in high-risk, human epidermal growth factor receptor 2 (HER2)–positive early breast cancer. We report the phase III IMpassion050 primary analysis of neoadjuvant atezolizumab, PH, and chemotherapy in these patients.
Collapse
Affiliation(s)
- Jens Huober
- Cantonal Hospital, Breast Center St Gallen, St Gallen, Switzerland.,University Hospital, Ulm, Germany
| | - Carlos H Barrios
- Centro de Pesquisa em Oncologia, Hospital São Lucas, PUCRS, Porto Alegre, Brazil
| | - Naoki Niikura
- Department of Breast Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Michał Jarząb
- Breast Cancer Unit, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | | | | | - José Pedrini
- Hospital Nossa Senhora da Conceição, Porto Alegre, Brazil
| | - Lyudmila Zhukova
- SBIH Moscow Clinical Scientific and Practical Center named after A.S. Loginov of DHM, Moscow, Russia
| | - Vilma Graupner
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Daniel Eiger
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Volkmar Henschel
- Product Development Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Nino Gochitashvili
- Product Development Safety, Roche Products Limited, Welwyn Garden City, United Kingdom
| | - Chiara Lambertini
- Oncology Biomarker Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Eleonora Restuccia
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Hong Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
5
|
Moragon S, Hernando C, Martinez-Martinez MT, Tapia M, Ortega-Morillo B, Lluch A, Bermejo B, Cejalvo JM. Immunological Landscape of HER-2 Positive Breast Cancer. Cancers (Basel) 2022; 14:3167. [PMID: 35804943 PMCID: PMC9265068 DOI: 10.3390/cancers14133167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Understanding the biological aspects of immune response in HER2+ breast cancer is crucial to implementing new treatment strategies in these patients. It is well known that anti-HER2 therapy has improved survival in this population, yet a substantial percentage may relapse, creating a need within the scientific community to uncover resistance mechanisms and determine how to overcome them. This systematic review indicates the immunological mechanisms through which trastuzumab and other agents target cancer cells, also outlining the main trials studying immune checkpoint blockade. Finally, we report on anti-HER2 vaccines and include a figure exemplifying their mechanisms of action.
Collapse
Affiliation(s)
- Santiago Moragon
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Cristina Hernando
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Maria Teresa Martinez-Martinez
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Marta Tapia
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Belen Ortega-Morillo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Ana Lluch
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
- Instituto de Salud Carlos III, CIBERONC (Centro De Investigacion Biomedica En Red De Cancer), 28220 Madrid, Spain
| | - Begoña Bermejo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
- Instituto de Salud Carlos III, CIBERONC (Centro De Investigacion Biomedica En Red De Cancer), 28220 Madrid, Spain
| | - Juan Miguel Cejalvo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
- Instituto de Salud Carlos III, CIBERONC (Centro De Investigacion Biomedica En Red De Cancer), 28220 Madrid, Spain
| |
Collapse
|
6
|
Interferon-γ increases sensitivity to chemotherapy and provides immunotherapy targets in models of metastatic castration-resistant prostate cancer. Sci Rep 2022; 12:6657. [PMID: 35459800 PMCID: PMC9033763 DOI: 10.1038/s41598-022-10724-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 04/12/2022] [Indexed: 01/26/2023] Open
Abstract
Interferon-γ (IFNγ) is a cytokine with limited evidence of benefit in cancer clinical trials to date. However, it could potentially play a role in potentiating anti-tumor immunity in the immunologically "cold" metastatic castration-resistant prostate cancer (mCRPC) by inducing antigen presentation pathways and concurrently providing targets for immune checkpoint blockade therapy. Moreover, it could additionally increase sensitivity to chemotherapy based on its pleiotropic effects on cell phenotype. Here, we show that IFNγ treatment induced expression of major histocompatibility class-I (MHC-I) genes and PD-L1 in prostate cancer cells in vitro. Furthermore, IFNγ treatment led to a decrease in E-cadherin expression with a consequent increase in sensitivity to chemotherapy in vitro. In an in vivo murine tumor model of spontaneous metastatic prostate cancer, IFNγ systemic pretreatment upregulated the expression of HLA-A and decreased E-cadherin expression in the primary tumor, and more importantly in the metastatic site led to increased apoptosis and limited micrometastases in combination with paclitaxel treatment compared to diffuse metastatic disease in control and monotherapy treatment groups. These findings suggest that IFNγ may be useful in combinatorial regimens to induce sensitivity to immunotherapy and chemotherapy in hepatic metastases of mCRPC.
Collapse
|
7
|
Th1 cytokine interferon gamma improves response in HER2 breast cancer by modulating the ubiquitin proteasomal pathway. Mol Ther 2021; 29:1541-1556. [PMID: 33412308 PMCID: PMC8058490 DOI: 10.1016/j.ymthe.2020.12.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/27/2020] [Accepted: 12/22/2020] [Indexed: 01/12/2023] Open
Abstract
HER2 breast cancer (BC) remains a significant problem in patients with locally advanced or metastatic BC. We investigated the relationship between T helper 1 (Th1) immune response and the proteasomal degradation pathway (PDP), in HER2-sensitive and -resistant cells. HER2 overexpression is partially maintained because E3 ubiquitin ligase Cullin5 (CUL5), which degrades HER2, is frequently mutated or underexpressed, while the client-protective co-chaperones cell division cycle 37 (Cdc37) and heat shock protein 90 (Hsp90) are increased translating to diminished survival. The Th1 cytokine interferon (IFN)-γ caused increased CUL5 expression and marked dissociation of both Cdc37 and Hsp90 from HER2, causing significant surface loss of HER2, diminished growth, and induction of tumor senescence. In HER2-resistant mammary carcinoma, either IFN-γ or Th1-polarizing anti-HER2 vaccination, when administered with anti-HER2 antibodies, demonstrated increased intratumor CUL5 expression, decreased surface HER2, and tumor senescence with significant therapeutic activity. IFN-γ synergized with multiple HER2-targeted agents to decrease surface HER2 expression, resulting in decreased tumor growth. These data suggest a novel function of IFN-γ that regulates HER2 through the PDP pathway and provides an opportunity to impact HER2 responses through anti-tumor immunity.
Collapse
|
8
|
Showalter L, Czerniecki BJ, Koski GK. Th1 cytokines in conjunction with pharmacological Akt inhibition potentiate apoptosis of breast cancer cells in vitro and suppress tumor growth in vivo. Oncotarget 2020; 11:2873-2888. [PMID: 32774769 PMCID: PMC7392628 DOI: 10.18632/oncotarget.27556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/19/2020] [Indexed: 11/25/2022] Open
Abstract
Targeted drug approaches have been a major focus for developing new anticancer therapies. Although many such agents approved in the last 20 years have improved outcomes, almost all have underperformed expectations. The full potential of such agents may yet be obtained through novel combinations. Previously, we showed that anti-estrogen drugs combined with a dendritic cell-based anti-HER-2 vaccine known to induce strong Th1-polarized immunity dramatically improved clinical response rates in patients with HER-2pos/ERpos early breast cancer. Here, we show that the small molecule Akt antagonist MK-2206, when combined with the Th1 cytokines IFN-gamma and TNF-alpha, maximize indicators of apoptotic cell death in a panel of phenotypically-diverse human breast cancer lines. These findings were mirrored by other, structurally-unrelated Akt-targeting drugs that work through different mechanisms. Interestingly, we found that MK-2206, as well as the other Akt antagonist drugs, also had a tendency to suppress Th1 cytokine expression in stimulated human and murine lymphocytes, potentially complicating their use in conjunction with active immunotherapy. After verifying that MK-2206 plus IFN-gamma could show similar combined effects against breast cancer lines, even in the absence of TNF-alpha, we tested in a rodent HER-2pos breast cancer model either a HER-2-based DC vaccine, or recombinant IFN-gamma with or without MK-2206 administration. We found that for MK-2206, co-administration of recombinant IFN-gamma outperformed co-administration of DC vaccination for slowing tumor growth kinetics. These findings suggest a combined therapy approach for Akt-targeting drugs that incorporates recombinant Interferon-gamma and is potentially translatable to humans.
Collapse
Affiliation(s)
- Loral Showalter
- Department of Biological Sciences, University Esplanade, Kent State University, Kent, Ohio, USA
| | - Brian J Czerniecki
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Gary K Koski
- Department of Biological Sciences, University Esplanade, Kent State University, Kent, Ohio, USA
| |
Collapse
|
9
|
Costa RLB, Czerniecki BJ. Clinical development of immunotherapies for HER2 + breast cancer: a review of HER2-directed monoclonal antibodies and beyond. NPJ Breast Cancer 2020; 6:10. [PMID: 32195333 PMCID: PMC7067811 DOI: 10.1038/s41523-020-0153-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer accounts for ~25% of breast cancer cases. Monoclonal antibodies (mAbs) against HER2 have led to unparalleled clinical benefit for a subset of patients with HER2+ breast cancer. In this narrative review, we summarize advances in the understanding of immune system interactions, examine clinical developments, and suggest rationales for future investigation of immunotherapies for HER2+ breast cancer. Complex interactions have been found between different branches of the immune system, HER2+ breast cancer, and targeted treatments (approved and under investigation). A new wave of immunotherapies, such as novel HER2-directed mAbs, antibody drug conjugates, vaccines, and adoptive T-cell therapies, are being studied in a broad population of patients with HER2-expressing tumors. The development of immunotherapies for HER2+ breast cancer represents an evolving field that should take into account interactions between different components of the immune system.
Collapse
Affiliation(s)
- Ricardo L B Costa
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Brian J Czerniecki
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| |
Collapse
|
10
|
Wu Y, Liu J, Movahedi F, Gu W, Xu T, Xu ZP. Enhanced Prevention of Breast Tumor Metastasis by Nanoparticle-Delivered Vitamin E in Combination with Interferon-Gamma. Adv Healthc Mater 2020; 9:e1901706. [PMID: 32052565 DOI: 10.1002/adhm.201901706] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/16/2020] [Indexed: 12/15/2022]
Abstract
Preventing cancer metastasis is one of the remaining challenges in cancer therapy. As an efficient natural product, alpha-tocopheryl succinate (α-TOS), the most effective form of vitamin E, holds great anticancer potential. To improve its efficacy and bioavailability, lipid-coated calcium carbonate/phosphate (LCCP) nanoparticles (NPs) with folic acid and PEG modification are synthesized for efficient delivery of α-TOS to 4T1 cancer cells. The optimized LCCP-FA NPs (NP-TOS15) show an α-TOS loading efficiency of around 60%, and enhanced uptake by 4T1 metastatic cancer cells. Consequently, NP-TOS15 significantly enhance the anticancer effect in combination with interferon-gamma (IFN-γ) in terms of apoptosis facilitation and migration inhibition. Importantly, NP-TOS15 upregulate the anticancer immunity via downregulating program death ligand 1 (PD-L1) expression that is initially induced by IFN-γ, and remarkably prevent the lung metastasis, particularly in combination with IFN-γ. Further investigation reveals that this combination therapy also modulates the cytotoxic lymphocyte infiltration into the tumor microenvironment for tumor elimination. Taken together, the NP delivery of α-TOS in combination with IFN-γ provides an applicable strategy for cancer therapy.
Collapse
Affiliation(s)
- Yilun Wu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Fatemeh Movahedi
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Tiefeng Xu
- The First Affiliated Hospital of Hainan Medical University Cancer Institute of Hainan Medical University Haikou Hainan 570102 China
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| |
Collapse
|
11
|
Krasniqi E, Barchiesi G, Pizzuti L, Mazzotta M, Venuti A, Maugeri-Saccà M, Sanguineti G, Massimiani G, Sergi D, Carpano S, Marchetti P, Tomao S, Gamucci T, De Maria R, Tomao F, Natoli C, Tinari N, Ciliberto G, Barba M, Vici P. Immunotherapy in HER2-positive breast cancer: state of the art and future perspectives. J Hematol Oncol 2019; 12:111. [PMID: 31665051 PMCID: PMC6820969 DOI: 10.1186/s13045-019-0798-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/25/2019] [Indexed: 02/08/2023] Open
Abstract
Breast cancer (BC) is a complex disease with primary or acquired incurability characteristics in a significant part of patients. Immunotherapeutical agents represent an emerging option for breast cancer treatment, including the human epidermal growth factor 2 positive (HER2+) subtype. The immune system holds the ability to spontaneously implement a defensive response against HER2+ BC cells through complex mechanisms which can be exploited to modulate this response for obtaining a clinical benefit. Initial immune system modulating strategies consisted mostly in vaccine therapies, which are still being investigated and improved. However, the entrance of trastuzumab into the scenery of HER2+ BC treatment was the real game changing event, which embodied a dominant immune-mediated mechanism. More recently, the advent of the immune checkpoint inhibitors has caused a new paradigm shift for immuno-oncology, with promising initial results also for HER2+ BC. Breast cancer has been traditionally considered poorly immunogenic, being characterized by relatively low tumor mutation burden (TMB). Nevertheless, recent evidence has revealed high tumor infiltrating lymphocytes (TILs) and programmed cell death-ligand 1 (PD-L1) expression in a considerable proportion of HER2+ BC patients. This may translate into a higher potential to elicit anti-cancer response and, therefore, wider possibilities for the use and implementation of immunotherapy in this subset of BC patients. We are herein presenting and critically discussing the most representative evidence concerning immunotherapy in HER2+ BC cancer, both singularly and in combination with therapeutic agents acting throughout HER2-block, immune checkpoint inhibition and anti-cancer vaccines. The reader will be also provided with hints concerning potential future projection of the most promising immutherapeutic agents and approaches for the disease of interest.
Collapse
Affiliation(s)
- E Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - G Barchiesi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - L Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - M Mazzotta
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy
| | - A Venuti
- HPV-UNIT, UOSD Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostic and Technological Innovation (RIDAIT), Translational Research Functional Departmental Area, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - M Maugeri-Saccà
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - G Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - G Massimiani
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - D Sergi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - S Carpano
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - P Marchetti
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy.,Medical Oncology Unit B, Policlinico Umberto I, Rome, Italy
| | - S Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Policlinico Umberto I, 'Sapienza' University of Rome, Rome, Italy
| | - T Gamucci
- Medical Oncology, Sandro Pertini Hospital, Rome, Italy
| | - R De Maria
- Institute of General Pathology, Catholic University of the Sacred Heart, Rome, Italy.,Department of Medical Oncology, Policlinico Universitario "A. Gemelli", Rome, Italy
| | - F Tomao
- Department of Gynecology-Obstetrics and Urology, "Sapienza" University of Rome, Rome, Italy
| | - C Natoli
- Department of Medical, Oral and Biotechnological Sciences and Center of Aging Science & Translational Medicine (CeSI-MeT), G. d'Annunzio University, Chieti, Italy
| | - N Tinari
- Department of Medical, Oral and Biotechnological Sciences and Center of Aging Science & Translational Medicine (CeSI-MeT), G. d'Annunzio University, Chieti, Italy
| | - G Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - M Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy.
| | - P Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| |
Collapse
|
12
|
Kodumudi KN, Ramamoorthi G, Snyder C, Basu A, Jia Y, Awshah S, Beyer AP, Wiener D, Lam L, Zhang H, Greene MI, Costa RLB, Czerniecki BJ. Sequential Anti-PD1 Therapy Following Dendritic Cell Vaccination Improves Survival in a HER2 Mammary Carcinoma Model and Identifies a Critical Role for CD4 T Cells in Mediating the Response. Front Immunol 2019; 10:1939. [PMID: 31475002 PMCID: PMC6702967 DOI: 10.3389/fimmu.2019.01939] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/31/2019] [Indexed: 12/27/2022] Open
Abstract
Patients with metastatic HER2 breast cancer (MBC) often become resistant to HER 2 targeted therapy and have recurrence of disease. The Panacea trial suggested that HER2 MBC patients were more likely to respond to checkpoint therapy if TIL were present or if tumor expressed PD-L1. We assessed whether type I polarized dendritic cells (DC1) could improve checkpoint therapy in a preclinical model of HER2+ breast cancer. TUBO bearing mice were vaccinated with either MHC class I or class II HER2 peptide pulsed DC1 (class I or class II HER2-DC1) concurrently or sequentially with administration of anti-PD-1 or anti-PDL1. Infiltration of tumors by immune cells, induction of anti-HER2 immunity and response to therapy was evaluated. Class I or class II HER2-DC1 vaccinated mice generated anti-HER2 CD8 or CD4+ T cell immune responses and demonstrated delayed tumor growth. Combining both MHC class I and II HER2-pulsed DC1 did not further result in inhibition of tumor growth or enhanced survival compared to individual administration. Interestingly class II HER2-DC1 led to both increased CD4 and CD8 T cells in the tumor microenvironment while class I peptides typically resulted in only increased CD8 T cells. Anti-PD-1 but not anti-PD-L1 administered sequentially with class I or class II HER2-DC1 vaccine could improve the efficacy of HER2-DC1 vaccine as measured by tumor growth, survival, infiltration of tumors by T cells and increase in systemic anti-HER2 immune responses. Depletion of CD4+ T cells abrogated the anti-tumor efficacy of combination therapy with class II HER2-DC1 and anti-PD-1, suggesting that tumor regression was CD4 dependent. Since class II HER2-DC1 was as effective as class I, we combined class II HER2-DC1 vaccine with anti-rat neu antibodies and anti-PD-1 therapy. Combination therapy demonstrated further delay in tumor growth, and enhanced survival compared to control mice. In summary, Class II HER2-DC1 drives both a CD4 and CD8 T cell tumor infiltration that leads to increased survival, and in combination with anti-HER2 therapy and checkpoint blockade can improve survival in preclinical models of HER2 positive breast cancer and warrants exploration in patients with HER2 MBC.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/therapy
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Combined Modality Therapy
- Dendritic Cells/immunology
- Female
- Humans
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/therapy
- Mice, Inbred BALB C
- Mice, Transgenic
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Rats
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Survival Analysis
- Treatment Outcome
- Tumor Burden/drug effects
- Tumor Burden/immunology
Collapse
Affiliation(s)
- Krithika N. Kodumudi
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Ganesan Ramamoorthi
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Colin Snyder
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Amrita Basu
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Yongsheng Jia
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Sabrina Awshah
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Amber P. Beyer
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Doris Wiener
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Lian Lam
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongtao Zhang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark I. Greene
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ricardo L. B. Costa
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Brian J. Czerniecki
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
13
|
Nagai Y, Ji MQ, Zhu F, Xiao Y, Tanaka Y, Kambayashi T, Fujimoto S, Goldberg MM, Zhang H, Li B, Ohtani T, Greene MI. PRMT5 Associates With the FOXP3 Homomer and When Disabled Enhances Targeted p185 erbB2/neu Tumor Immunotherapy. Front Immunol 2019; 10:174. [PMID: 30800128 PMCID: PMC6375878 DOI: 10.3389/fimmu.2019.00174] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are a subpopulation of T cells that are specialized in suppressing immune responses. Here we show that the arginine methyl transferase protein PRMT5 can complex with FOXP3 transcription factors in Tregs. Mice with conditional knock out (cKO) of PRMT5 expression in Tregs develop severe scurfy-like autoimmunity. In these PRMT5 cKO mice, the spleen has reduced numbers of Tregs, but normal numbers of Tregs are found in the peripheral lymph nodes. These peripheral Tregs that lack PRMT5, however, display a limited suppressive function. Mass spectrometric analysis showed that FOXP3 can be di-methylated at positions R27, R51, and R146. A point mutation of Arginine (R) 51 to Lysine (K) led to defective suppressive functions in human CD4 T cells. Pharmacological inhibition of PRMT5 by DS-437 also reduced human Treg functions and inhibited the methylation of FOXP3. In addition, DS-437 significantly enhanced the anti-tumor effects of anti-erbB2/neu monoclonal antibody targeted therapy in Balb/c mice bearing CT26Her2 tumors by inhibiting Treg function and induction of tumor immunity. Controlling PRMT5 activity is a promising strategy for cancer therapy in situations where host immunity against tumors is attenuated in a FOXP3 dependent manner.
Collapse
Affiliation(s)
- Yasuhiro Nagai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mei Q Ji
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Fuxiang Zhu
- Unit of Molecular Immunology, Key Laboratory of Molecular Virology & Immunology, CAS Center for Excellence in Molecular Cell Science, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Xiao
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yukinori Tanaka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | | | - Hongtao Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Bin Li
- The Department of Immunology and Microbiology & Shanghai, Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Takuya Ohtani
- Penn Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark I Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
14
|
Yaghoobi H, Azizi H, Oskooei VK, Taheri M, Ghafouri-Fard S. Assessment of expression of interferon γ (IFN-G) gene and its antisense (IFNG-AS1) in breast cancer. World J Surg Oncol 2018; 16:211. [PMID: 30336781 PMCID: PMC6194673 DOI: 10.1186/s12957-018-1508-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/08/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The role of long non-coding RNAs has been extensively appreciated in the contexts of cancer. Interferon γ-antisense RNA1 (IFNG-AS1) is an lncRNA located near to IFN-γ-encoding (IFNG) gene and regulates expression of IFNG in Th1 cells. METHODS In the present study, we evaluated expression of IFNG and IFNG-AS1 in 108 breast samples including tumoral tissues and their adjacent non-cancerous tissues (ANCTs) using real-time PCR. IFNG-AS1 was significantly upregulated in tumoral tissues compared with ANCTs (expression ratio = 2.23, P = 0.03). RESULTS Although the expression of IFNG was higher in tumoral tissues compared with ANCTs (relative expression = 1.89), it did not reach the level of significance (P = 0.07). IFNG expression was significantly higher in HER2-negative tumoral tissues compared with HER2-positive ones (P = 0.01) and in grade 1 samples compared with grade 2 ones (P = 0.03). No other significant difference was found in expressions of genes between other groups. CONCLUSION Significant strong correlations were detected between expression of IFNG and IFNG-AS1 in both tumoral tissues and ANCTs. The present study provides evidences for participation of IFNG and IFNG-AS1 in the pathogenesis of breast cancer and warrants future studies to elaborate the underlying mechanism.
Collapse
Affiliation(s)
- Hajar Yaghoobi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hakim Azizi
- Department of Medical Parasitology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Vahid Kholghi Oskooei
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Zhang H, Lam L, Nagai Y, Zhu Z, Chen X, Ji MQ, Greene MI. A targeted immunotherapy approach for HER2/neu transformed tumors by coupling an engineered effector domain with interferon-γ. Oncoimmunology 2018; 7:e1300739. [PMID: 29632709 DOI: 10.1080/2162402x.2017.1300739] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 01/05/2023] Open
Abstract
Despite substantial clinical progress with targeted therapies, current antibody-based approaches have limited efficacy at controlling HER2/neu-positive breast cancers, especially in the absence of chemotherapies. Previously, we showed that the combination of IFNγ and anti-HER2/neu antibody synergistically reduces tumor growth in an in vivo implanted mammary tumor model. Here, we report a recombinant approach to produce an anti-HER2/neu scFv and IFNγ fusion protein using an engineered effector domain (EED) scaffold. The new molecule induces in vitro apoptosis in an IFNγ receptor-dependent manner. At a very low dose in the in vivo xenografted tumor models, the new EED-IFNγ fusion protein demonstrates superior activity over the anti-HER2/neu antibody and is even active on tumors that are resistant to anti-HER2/neu antibody therapy. Examination of tumor infiltrated macrophages and lymphocytes reveals that the fusion protein can induce changes in tumor microenvironment to support immune reactivity against tumors. Our studies have defined a targeted immunotherapy approach for the treatment of cancers.
Collapse
Affiliation(s)
- Hongtao Zhang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lian Lam
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasuhiro Nagai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhiqiang Zhu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xi Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mei Q Ji
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark I Greene
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
Chen X, Nagai Y, Zhu Z, Ruan H, Peehl DM, Greene MI, Zhang H. A spliced form of CD44 expresses the unique glycan that is recognized by the prostate cancer specific antibody F77. Oncotarget 2018; 9:3631-3640. [PMID: 29423071 PMCID: PMC5790488 DOI: 10.18632/oncotarget.23341] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/26/2017] [Indexed: 01/08/2023] Open
Abstract
Prostate cancer is the most common cancer occurring in men in the United States. The monoclonal antibody F77 that was originally developed in our laboratory recognizes mainly glycolipids as well as O-linked glycosylation on proteins in prostate cancer cells. We have identified a spliced form of glycoprotein CD44 as one critical protein expressing the F77 antigen. The F77-specific glycosylation occurs on multiple potential glycosylation sites on the CD44 protein encoded by the fourteenth exon. CD44 is a tumor stem cell marker and is known to induce tumor stemness and metastasis. Knockdown of CD44 or FUT1 genes dramatically reduced F77-induced apoptosis in prostate cancer cell lines. We developed an ELISA using both a CD44 antibody and F77 to identify the special form of glycosylated CD44 from prostate cancer cells as well as from serum samples of prostate cancer patients. These results reveal a CD44-dependent mechanism for F77 to induce tumor cell apoptosis, and a new strategy for the detection of glycosylated CD44 proteins secreted by prostate cancer cells.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yasuhiro Nagai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiqiang Zhu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hang Ruan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donna M. Peehl
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark I. Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongtao Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
Nallar SC, Kalvakolanu DV. GRIM-19: A master regulator of cytokine induced tumor suppression, metastasis and energy metabolism. Cytokine Growth Factor Rev 2016; 33:1-18. [PMID: 27659873 DOI: 10.1016/j.cytogfr.2016.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 09/14/2016] [Indexed: 12/31/2022]
Abstract
Cytokines induce cell proliferation or growth suppression depending on the context. It is increasingly becoming clear that success of standard radiotherapy and/or chemotherapeutics to eradicate solid tumors is dependent on IFN signaling. In this review we discuss the molecular mechanisms of tumor growth suppression by a gene product isolated in our laboratory using a genome-wide expression knock-down strategy. Gene associated with retinoid-IFN-induced mortality -19 (GRIM-19) functions as non-canonical tumor suppressor by antagonizing oncoproteins. As a component of mitochondrial respiratory chain, GRIM-19 influences the degree of "Warburg effect" in cancer cells as many advanced and/or aggressive tumors show severely down-regulated GRIM-19 levels. In addition, GRIM-19 appears to regulate innate and acquired immune responses in mouse models. Thus, GRIM-19 is positioned at nodes that favor cell protection and/or prevent aberrant cell growth.
Collapse
Affiliation(s)
- Shreeram C Nallar
- Department of Microbiology and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dhan V Kalvakolanu
- Department of Microbiology and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|