1
|
Karlsen EA, Walpole E, Simpson F. Steroid Premedication and Monoclonal Antibody Therapy: Should We Reconsider? Curr Treat Options Oncol 2024; 25:275-283. [PMID: 38270799 PMCID: PMC10894762 DOI: 10.1007/s11864-023-01170-4] [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] [Accepted: 11/30/2023] [Indexed: 01/26/2024]
Abstract
OPINION STATEMENT Monoclonal antibody (mAb) therapy is now considered a main component of cancer therapy in Australia. Although traditionally thought of as pure signalling inhibitors, a large proponent of these medications function through antibody-dependent cell-mediated cytotoxicity (ADCC). Currently, most protocols and institutional guidelines for ADCC-mediated mAbs promote the use of corticosteroids as premedication: this is implemented to reduce infusion-related reactions (IRRs) and antiemesis prophylaxis and combat concurrently administered chemotherapy-related syndromes. Concerningly, the inhibitory effects of ADCC by corticosteroids are well documented; henceforth, it is possible the current standard of care is misaligned to the literature surrounding ADCC. Subsequently, clinicians' decisions to act in contrast to this literature may be reducing the efficacy of mAbs. The literature suggests that the redundant use of corticosteroids should be cautioned against when used in conjunction with ADCC-mediated mAbs-this is due to the consequent reduction in anti-tumour activity. Owing to the fact IRRs typically occur upon initial infusion, the authors advocate for individual clinicians and institutional protocols to considering augmenting their practice to corticosteroid premedication at the first dose only, unless clinically indicated. Additionally, product information (PI) and consumer medicine information (CMI) documents distributed by Australian and international regulatory agencies should consider disclosing the risk of concurrent steroids with these medications. Moreover, the authors suggest considering alternative medications for the management of side effects.
Collapse
Affiliation(s)
- Emma-Anne Karlsen
- Frazer Institute, The University of Queensland, Brisbane, Australia.
- Department of General Surgery, Mater Hospital Brisbane, Brisbane, Australia.
- School of Medicine, The University of Queensland, Brisbane, Australia.
- Simpson Laboratory - Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Euan Walpole
- School of Medicine, The University of Queensland, Brisbane, Australia
- Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - Fiona Simpson
- Frazer Institute, The University of Queensland, Brisbane, Australia
- Simpson Laboratory - Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
2
|
Zheng Q, Chen D, Wang X, Yang Y, Zhao S, Dong X, Ma C, Zhang X, Duan H, Sun Y, Zheng S. WX-0593 combined with an epithelial growth factor receptor (EGFR) monoclonal antibody in the treatment of xenograft tumors carrying triple EGFR mutations. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:696. [PMID: 35845484 PMCID: PMC9279820 DOI: 10.21037/atm-22-2780] [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: 05/10/2022] [Accepted: 06/20/2022] [Indexed: 11/06/2022]
Abstract
Background To evaluate the safety and therapeutic efficacy of WX-0593, a newly developed potent anaplastic lymphoma kinase (ALK) inhibitor, in combination with an epithelial growth factor receptor (EGFR) monoclonal antibody (QL1203 or Vectibix) for the treatment of xenograft tumors carrying mutant EGFR and osimertinib-resistant mutations (EGFR/T790M/C797S). Methods The inhibition of tumor cell proliferation by WX-0593 and Vectibix alone or combined was evaluated in four EGFR triple-mutant cell lines: PC9 (EGFR Del19/T790M/C797S), NCI-H1975 (EGFR L858R/T790M/C797S), Ba/F3 (EGFR L858R/T790M/C797S and EGFR Del19/T790M/C797S). The in vivo antitumor efficacy of WX-0593 alone or combined with QL1203 or Vectibix was evaluated in xenograft tumor models of BALB/c nude mice developed from H1975 (EGFR-Del19/T790M/C797S) and Ba/F3 (EGFR-L858R/T790M/C797S) cell lines. Mice were randomized into groups and treated with or without WX-0593, QL1203, Vectibix, or their combination. The tumor volume, mouse body weight, and therapeutic side effects were monitored routinely. Blood samples were obtained from all mice at different time points after the last dosage of treatment to evaluate the pharmacokinetic parameters of the drugs. Results WX-0593 and Vectibix showed a strong synergistic inhibitory effect on the proliferation of two EGFR triple-mutant Ba/F3 cell lines (EGFR L858R/T790M/C797S and Del19/T790M/C797S), but little synergistic inhibitory effect on the proliferation of NCI-H1975 (EGFR L858R/T790M/C797S) and PC9 (EGFR Del19/T790M/C797S). In vivo, WX-0593 (25 mg/kg) showed a modest therapeutic effect when combined with QL1203 or Vectibix, but had no effect on tumor growth as a monotherapy at this dosage. WX-0593 (75 mg/kg) exhibited modest antitumor efficacy that was further enhanced in combination with QL1203 or Vectibix in both tumor models (H1975 and Ba/F3). No significant body weight alteration, any other side effect, or deaths were observed during treatment. Pharmacokinetic analysis showed that the serum level of QL1203 or Vectibix was significantly increased and lasted longer when combined with WX-0593. Conclusions WX-0593 exhibited a synergetic effect with an EGFR monoclonal antibody on osimertinib-resistant EGFR-mutant non-small cell lung cancer (NSCLC) both in vitro and in vivo. Their combination showed potent antitumor efficacy and an acceptable safety profile, which may be a promising strategy for the treatment of patients with EGFR triple-mutant NSCLC resistant to osimertinib.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Dong
- Qilu Pharmaceutical Co., Ltd., Jinan, China
| | - Cuicui Ma
- Qilu Pharmaceutical Co., Ltd., Jinan, China
| | - Xin Zhang
- Qilu Pharmaceutical Co., Ltd., Jinan, China
| | | | - Yan Sun
- Qilu Pharmaceutical Co., Ltd., Jinan, China
| | | |
Collapse
|
3
|
Kono M, Yamaki H, Komatsuda H, Kumai T, Hayashi R, Wakisaka R, Sato R, Ohara K, Kishibe K, Takahara M, Katada A, Hayashi T, Harabuchi Y. IL-2 complex recovers steroid-induced inhibition in immunochemotherapy for head and neck cancer. Transl Oncol 2022; 18:101358. [PMID: 35123188 PMCID: PMC8819385 DOI: 10.1016/j.tranon.2022.101358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Steroid diminished the antitumor effect of combination therapy with anti-PD-1 Ab and CDDP in an HNSCC mouse model by reducing the T cell proliferation and suppressing memory T cells. In vitro assessment using antigen-specific T cells demonstrated that steroid induced apoptosis, decreased proliferation, and reduced tumor cytotoxicity. IL-2 or IL-2 Cx restored steroid-induced immunosuppression of T cells by restoring the proliferation and function of T cells in vitro and in vivo.
Background: A combination therapy with immune checkpoint inhibitors (ICIs) and platinum-based chemotherapy has become the first-line treatment for recurrent or metastatic head and neck squamous carcinoma (HNSCC). Although steroids are often used as anti-emetic medications during chemotherapy, their adverse effects on immune-combined chemotherapy are unclear in HNSCC. Methods: The effects of dexamethasone on tumor growth and immune cell population were evaluated in a mouse HNSCC model treated with PD-1 blockade combined with cisplatin. The effect of various doses of dexamethasone on cell proliferation, survival, surface markers, IFN-γ production, and antitumor effects in antigen-specific T cells was examined in vitro. The recovery of T cell dysfunction by IL-2 was assessed in vitro and in vivo. Results: In a mouse HNSCC model, dexamethasone showed limited antitumor effects on immunochemotherapy. Dexamethasone decreased the number of T cells and inhibited T cell differentiation into effector and central memory T cells. In the in vitro assessment, dexamethasone induced cell death, limited proliferation, and reduced the reactivity against HNSCC cell lines of antigen-specific T cells in a dose-dependent manner. The expression of inhibitory receptors on T cells was not affected by steroids. This inhibition was recovered by IL-2 and IL-2/anti-IL-2 complexes (IL-2 Cx) in vitro and in vivo, respectively. Conclusion: Our preclinical data indicate that dexamethasone diminishes the antitumor effects of immunochemotherapy in patients with HNSCC. IL-2 Cx recovered the inhibition of antitumor immunity by steroids and might be a potent immune adjuvant for patients who require steroids during PD-1 blockade and chemotherapy.
Collapse
Affiliation(s)
- Michihisa Kono
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Hidekiyo Yamaki
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Hiroki Komatsuda
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan; Department of Innovative Head and Neck Cancer Research and Treatment, Asahikawa Medical University, Japan
| | - Ryusuke Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Risa Wakisaka
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Ryosuke Sato
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Kenzo Ohara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Kan Kishibe
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Miki Takahara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Akihiro Katada
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | - Tatsuya Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan; Department of Innovative Head and Neck Cancer Research and Treatment, Asahikawa Medical University, Japan
| | - Yasuaki Harabuchi
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| |
Collapse
|
4
|
Pradier A, Papaserafeim M, Li N, Rietveld A, Kaestel C, Gruaz L, Vonarburg C, Spirig R, Puga Yung GL, Seebach JD. Small-Molecule Immunosuppressive Drugs and Therapeutic Immunoglobulins Differentially Inhibit NK Cell Effector Functions in vitro. Front Immunol 2019; 10:556. [PMID: 30972058 PMCID: PMC6445861 DOI: 10.3389/fimmu.2019.00556] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/01/2019] [Indexed: 12/14/2022] Open
Abstract
Small-molecule immunosuppressive drugs (ISD) prevent graft rejection mainly by inhibiting T lymphocytes. Therapeutic immunoglobulins (IVIg) are used for substitution, antibody-mediated rejection (AbMR) and HLA-sensitized recipients by targeting distinct cell types. Since the effect of ISD and IVIg on natural killer (NK) cells remains somewhat controversial in the current literature, the aim of this comparative study was to investigate healthy donor's human NK cell functions after exposure to ISD and IVIg, and to comprehensively review the current literature. NK cells were incubated overnight with IL2/IL12 and different doses and combinations of ISD and IVIg. Proliferation was evaluated by 3[H]-thymidine incorporation; phenotype, degranulation and interferon gamma (IFNγ) production by flow cytometry and ELISA; direct NK cytotoxicity by standard 51[Cr]-release and non-radioactive DELFIA assays using K562 as stimulator and target cells; porcine endothelial cells coated with human anti-pig antibodies were used as targets in antibody-dependent cellular cytotoxicity (ADCC) assays. We found that CD69, CD25, CD54, and NKG2D were downregulated by ISD. Proliferation was inhibited by methylprednisolone (MePRD), mycophenolic acid (MPA), and everolimus (EVE). MePRD and MPA reduced degranulation, MPA only of CD56bright NK cells. MePRD and IVIg inhibited direct cytotoxicity and ADCC. Combinations of ISD demonstrated cumulative inhibitory effects. IFNγ production was inhibited by MePRD and ISD combinations, but not by IVIg. In conclusion, IVIg, ISD and combinations thereof differentially inhibit NK cell functions. The most potent drug with an effect on all NK functions was MePRD. The fact that MePRD and IVIg significantly block NK cytotoxicity, especially ADCC, has major implications for AbMR as well as therapeutic strategies targeting cancer and immune cells with monoclonal antibodies.
Collapse
Affiliation(s)
- Amandine Pradier
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Maria Papaserafeim
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Ning Li
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Anke Rietveld
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Charlotte Kaestel
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Lyssia Gruaz
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| | | | | | - Gisella L Puga Yung
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Jörg D Seebach
- Division of Immunology and Allergy, University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
5
|
Fiedler W, Stoeger H, Perotti A, Gastl G, Weidmann J, Dietrich B, Baumeister H, Danielczyk A, Goletz S, Salzberg M, De Dosso S. Phase I study of TrasGEX, a glyco-optimised anti-HER2 monoclonal antibody, in patients with HER2-positive solid tumours. ESMO Open 2018; 3:e000381. [PMID: 30018811 PMCID: PMC6045773 DOI: 10.1136/esmoopen-2018-000381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 01/21/2023] Open
Abstract
Purpose TrasGEX is a second-generation monoclonal antibody of trastuzumab, glyco-optimised to enhance antibody-dependent cellular cytotoxicity while fully retaining trastuzumab’s antigen-binding properties to human epidermal growth factor receptor 2 (HER2). A phase I dose-escalation study was conducted to establish the optimal TrasGEX dose and regimen for phase II studies and to define the safety, pharmacokinetics (PK) and preliminary antitumour activity of TrasGEX. Patients and methods A total of 37 patients with advanced HER2-positive carcinomas and progressive disease received TrasGEX intravenously every 3 weeks until disease progression in doses of 12–720 mg in a three-plus-three dose escalation design, including an expansion cohort at the highest dose. Results No dose limiting toxicity was observed, and no maximum tolerated dose was reached. Drug-related adverse events were mainly infusion-related reactions occurring during the first infusion in 51% of patients; all but two were mild-to-moderate. Compared with trastuzumab, the PK parameters were dose dependent, with a mean terminal half-life (t1/2) of 263±99 hours for the 720 mg dose. Clinical benefit in 15 out of 30 (50%) evaluable patients included one ongoing complete response, two partial remissions lasting 16 and 77 weeks and disease stabilisation (SD) in 12 patients lasting a median (range) of 17 (7–26) weeks; three of them had SD of 24, 25 and 26 weeks, respectively. Conclusion TrasGEX was safe, well-tolerated and showed antitumour activity in 50% of evaluable patients, all with progressive disease at study entry. Infusions at an interval of 2–3 weeks should achieve clinically relevant trough levels for future studies (NCT01409343).
Collapse
Affiliation(s)
- Walter Fiedler
- Department of Medicine II, Hubertus-Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Herbert Stoeger
- Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Guenther Gastl
- Department of Internal Medicine V (Hematology and Oncology), Innsbruck Medical University (IMU), Innsbruck, Austria
| | - Jens Weidmann
- Department of Medicine II, Hubertus-Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | - Sara De Dosso
- Oncology Institute of Southern Switzerland, Ospedale Regionale Bellinzona e Valli, Bellinzona, Switzerland
| |
Collapse
|
6
|
Sasaki K, Miyashita Y, Asai D, Funamoto D, Sato K, Yamaguchi Y, Mishima Y, Iino T, Takaishi S, Nagano J, Kishimura A, Mori T, Katayama Y. A peptide inhibitor of antibody-dependent cell-mediated cytotoxicity against EGFR/folate receptor-α double positive cells. MEDCHEMCOMM 2018; 9:783-788. [PMID: 30108967 PMCID: PMC6072457 DOI: 10.1039/c8md00010g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/24/2018] [Indexed: 12/20/2022]
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) is caused by natural killer (NK) cells upon recognition of antigen-bound IgG via FcγRIIIa. This mechanism is crucial for cytolysis of pathogen-infected cells and monoclonal antibody (mAb)-mediated elimination of cancer cells. However, there is concern that mAb-based cancer therapy induces ADCC against non-target cells expressing antigens. To date, no strategy has been reported to enhance the selectivity of ADCC to protect non-target cells expressing antigens. Here, we introduce a model inhibitor which specifically blocks ADCC of anti-EGFR mAbs towards EGFR/folate receptor α (FRα) double positive cells. This inhibitor recruits mAbs on the FRα of the cell surface independent of Fab antigen recognition. The resulting ternary and/or quaternary complexes formed on the cell surface suppress signal transduction of FcγRIIIa in NK cells, consequently leading to more specific ADCC.
Collapse
Affiliation(s)
- Koichi Sasaki
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Yoshiki Miyashita
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Daisuke Asai
- Department of Microbiology , St. Marianna University School of Medicine , 2-16-1 Sugao, Miyamae-ku , Kawasaki , 216-8511 , Japan
| | - Daiki Funamoto
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Kazuki Sato
- Department of Environmental Science , Fukuoka Women's University , 1-1-1 Kasumigaoka , Fukuoka , 813-8529 , Japan
| | - Yoko Yamaguchi
- Department of Environmental Science , Fukuoka Women's University , 1-1-1 Kasumigaoka , Fukuoka , 813-8529 , Japan
| | - Yuji Mishima
- Clinical Chemotherapy , Cancer Chemotherapy Center , Japanese Foundation for Cancer Research , 3-8-31, Ariake, Koto-ku , Tokyo , 135-8550 , Japan
| | - Tadafumi Iino
- Centre for Advanced Medicine Innovation , Kyushu University , 3-1-1 Maidashi, Higashi-ku , Fukuoka , 812-8582 , Japan
| | - Shigeo Takaishi
- Centre for Advanced Medicine Innovation , Kyushu University , 3-1-1 Maidashi, Higashi-ku , Fukuoka , 812-8582 , Japan
| | - Jun Nagano
- Faculty of Arts and Science , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka 819-0395 , Fukuoka , Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Takeshi Mori
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| | - Yoshiki Katayama
- Department of Applied Chemistry , Faculty of Engineering , Kyushu University , 744 Motooka, Nishi-ku , Fukuoka , 819-0395 , Japan . ;
| |
Collapse
|
7
|
Regulatory effects of dexamethasone on NK and T cell immunity. Inflammopharmacology 2017; 26:1331-1338. [PMID: 29159714 PMCID: PMC6153920 DOI: 10.1007/s10787-017-0418-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 11/08/2017] [Indexed: 11/29/2022]
Abstract
Glucocorticoids (GCs) act via the intracellular glucocorticoid receptor (GR), which can regulate the expression of target genes. With regard to the immune system, GCs may affect both innate and adaptive immunity. Our study analyzed the immunoregulatory effects of dexamethasone (Dex) treatment on splenic T, Treg, NK and NKT cells by treating C57Bl6 mice with various doses of Dex. We observed that treatment with Dex decreased the number of NK cells in the spleen and suppressed their activity. In particular, the expression of both Ly49G and NKG2D receptors was decreased by Dex. However, Dex did not affect the population of NKT cells. With regard to splenic T cells, our results show a dose-dependent reduction in CD3+, CD4+, CD8+, CD44+ and CD8+CD122+ T cells, but a stimulatory effect on CD4+CD25+ regulatory T cells by Dex treatment. In addition, treatment with Dex suppressed anti-tumor immune response in a mouse EG7 tumor model. We conclude that Dex may suppress both T- and NK-mediated immunity.
Collapse
|