1
|
Stenmark Tullberg A, Sjöström M, Tran L, Niméus E, Killander F, Kovács A, Lundstedt D, Holmberg E, Karlsson P. Combining histological grade, TILs, and the PD-1/PD-L1 pathway to identify immunogenic tumors and de-escalate radiotherapy in early breast cancer: a secondary analysis of a randomized clinical trial. J Immunother Cancer 2023; 11:e006618. [PMID: 37208129 PMCID: PMC10201214 DOI: 10.1136/jitc-2022-006618] [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: 03/17/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND The implementation of immunological biomarkers for radiotherapy (RT) individualization in breast cancer requires consideration of tumor-intrinsic factors. This study aimed to investigate whether the integration of histological grade, tumor-infiltrating lymphocytes (TILs), programmed cell death protein-1 (PD-1), and programmed death ligand-1 (PD-L1) can identify tumors with aggressive characteristics that can be downgraded regarding the need for RT. METHODS The SweBCG91RT trial included 1178 patients with stage I-IIA breast cancer, randomized to breast-conserving surgery with or without adjuvant RT, and followed for a median time of 15.2 years. Immunohistochemical analyses of TILs, PD-1, and PD-L1 were performed. An activated immune response was defined as stromal TILs ≥10% and PD-1 and/or PD-L1 expression in ≥1% of lymphocytes. Tumors were categorized as high-risk or low-risk using assessments of histological grade and proliferation as measured by gene expression. The risk of ipsilateral breast tumor recurrence (IBTR) and benefit of RT were then analyzed with 10 years follow-up based on the integration of immune activation and tumor-intrinsic risk group. RESULTS Among high-risk tumors, an activated immune infiltrate was associated with a reduced risk of IBTR (HR 0.34, 95% CI 0.16 to 0.73, p=0.006). The incidence of IBTR in this group was 12.1% (5.6-25.0) without RT and 4.4% (1.1-16.3) with RT. In contrast, the incidence of IBTR in the high-risk group without an activated immune infiltrate was 29.6% (21.4-40.2) without RT and 12.8% (6.6-23.9) with RT. Among low-risk tumors, no evidence of a favorable prognostic effect of an activated immune infiltrate was seen (HR 2.0, 95% CI 0.87 to 4.6, p=0.100). CONCLUSIONS Integrating histological grade and immunological biomarkers can identify tumors with aggressive characteristics but a low risk of IBTR despite a lack of RT boost and systemic therapy. Among high-risk tumors, the risk reduction of IBTR conferred by an activated immune infiltrate is comparable to treatment with RT. These findings may apply to cohorts dominated by estrogen receptor-positive tumors.
Collapse
Affiliation(s)
- Axel Stenmark Tullberg
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Martin Sjöström
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
| | - Lena Tran
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
| | - Emma Niméus
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Fredrika Killander
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Dan Lundstedt
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Erik Holmberg
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Per Karlsson
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| |
Collapse
|
2
|
Seyedmirzaei H, Keshavarz-Fathi M, Razi S, Gity M, Rezaei N. Recent progress in immunotherapy of breast cancer targeting the human epidermal growth factor receptor 2 (HER2). J Oncol Pharm Pract 2021; 27:1235-1244. [PMID: 33530866 DOI: 10.1177/1078155221991636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Breast cancer is responsible for most of the cancer-induced deaths in women around the world. The current review will discuss different approaches of targeting HER2, an epidermal growth factor overexpressed in 30% of breast cancer cases. DATA SOURCES We conducted a search on Pubmed and Scopus databases to find studies relevant to HER2+ breast cancers and targeting HER2 as means of immunotherapy. Out of 1043 articles, 105 studies were included in this review. DATA SUMMARY As well as the introduction of HER2 and breast cancer subtypes, we discussed various aspects of HER2-targeting immunotherapy including monoclonal antibodies, Antibody-drug conjugates (ADCs), Chimeric Antigen Receptor (CAR) T-cells and vaccines. CONCLUSIONS Despite several ways of controlling breast cancer, the need to investigate new drugs and approaches seems to be much significant as this cancer still has a heavy burden on people's health and survival.
Collapse
Affiliation(s)
- Homa Seyedmirzaei
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Breast Cancer Association (BrCA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Gity
- Breast Cancer Association (BrCA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Breast Disease Research Center (BDRC), Advanced Imaging Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Braman N, Prasanna P, Whitney J, Singh S, Beig N, Etesami M, Bates DDB, Gallagher K, Bloch BN, Vulchi M, Turk P, Bera K, Abraham J, Sikov WM, Somlo G, Harris LN, Gilmore H, Plecha D, Varadan V, Madabhushi A. Association of Peritumoral Radiomics With Tumor Biology and Pathologic Response to Preoperative Targeted Therapy for HER2 (ERBB2)-Positive Breast Cancer. JAMA Netw Open 2019; 2:e192561. [PMID: 31002322 PMCID: PMC6481453 DOI: 10.1001/jamanetworkopen.2019.2561] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPORTANCE There has been significant recent interest in understanding the utility of quantitative imaging to delineate breast cancer intrinsic biological factors and therapeutic response. No clinically accepted biomarkers are as yet available for estimation of response to human epidermal growth factor receptor 2 (currently known as ERBB2, but referred to as HER2 in this study)-targeted therapy in breast cancer. OBJECTIVE To determine whether imaging signatures on clinical breast magnetic resonance imaging (MRI) could noninvasively characterize HER2-positive tumor biological factors and estimate response to HER2-targeted neoadjuvant therapy. DESIGN, SETTING, AND PARTICIPANTS In a retrospective diagnostic study encompassing 209 patients with breast cancer, textural imaging features extracted within the tumor and annular peritumoral tissue regions on MRI were examined as a means to identify increasingly granular breast cancer subgroups relevant to therapeutic approach and response. First, among a cohort of 117 patients who received an MRI prior to neoadjuvant chemotherapy (NAC) at a single institution from April 27, 2012, through September 4, 2015, imaging features that distinguished HER2+ tumors from other receptor subtypes were identified. Next, among a cohort of 42 patients with HER2+ breast cancers with available MRI and RNaseq data accumulated from a multicenter, preoperative clinical trial (BrUOG 211B), a signature of the response-associated HER2-enriched (HER2-E) molecular subtype within HER2+ tumors (n = 42) was identified. The association of this signature with pathologic complete response was explored in 2 patient cohorts from different institutions, where all patients received HER2-targeted NAC (n = 28, n = 50). Finally, the association between significant peritumoral features and lymphocyte distribution was explored in patients within the BrUOG 211B trial who had corresponding biopsy hematoxylin-eosin-stained slide images. Data analysis was conducted from January 15, 2017, to February 14, 2019. MAIN OUTCOMES AND MEASURES Evaluation of imaging signatures by the area under the receiver operating characteristic curve (AUC) in identifying HER2+ molecular subtypes and distinguishing pathologic complete response (ypT0/is) to NAC with HER2-targeting. RESULTS In the 209 patients included (mean [SD] age, 51.1 [11.7] years), features from the peritumoral regions better discriminated HER2-E tumors (maximum AUC, 0.85; 95% CI, 0.79-0.90; 9-12 mm from the tumor) compared with intratumoral features (AUC, 0.76; 95% CI, 0.69-0.84). A classifier combining peritumoral and intratumoral features identified the HER2-E subtype (AUC, 0.89; 95% CI, 0.84-0.93) and was significantly associated with response to HER2-targeted therapy in both validation cohorts (AUC, 0.80; 95% CI, 0.61-0.98 and AUC, 0.69; 95% CI, 0.53-0.84). Features from the 0- to 3-mm peritumoral region were significantly associated with the density of tumor-infiltrating lymphocytes (R2 = 0.57; 95% CI, 0.39-0.75; P = .002). CONCLUSIONS AND RELEVANCE A combination of peritumoral and intratumoral characteristics appears to identify intrinsic molecular subtypes of HER2+ breast cancers from imaging, offering insights into immune response within the peritumoral environment and suggesting potential benefit for treatment guidance.
Collapse
Affiliation(s)
- Nathaniel Braman
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Prateek Prasanna
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Jon Whitney
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Salendra Singh
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Niha Beig
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Maryam Etesami
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - David D. B. Bates
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine Gallagher
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - B. Nicolas Bloch
- Department of Radiology, Boston Medical Center, Boston, Massachusetts
- Department of Radiology, Boston University School of Medicine, Boston, Massachusetts
| | - Manasa Vulchi
- Department of Hematology and Medical Oncology, The Cleveland Clinic, Cleveland, Ohio
| | - Paulette Turk
- Department of Diagnostic Radiology, The Cleveland Clinic, Cleveland, Ohio
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Jame Abraham
- Department of Hematology and Medical Oncology, The Cleveland Clinic, Cleveland, Ohio
| | - William M. Sikov
- Program in Women’s Oncology, Women and Infants Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - George Somlo
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Lyndsay N. Harris
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Hannah Gilmore
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Donna Plecha
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Vinay Varadan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, Ohio
| |
Collapse
|
4
|
Soltantoyeh T, Bahadori T, Hosseini-Ghatar R, Khoshnoodi J, Roohi A, Mobini M, Golsaz-Shirazi F, Jeddi-Tehrani M, Amiri MM, Shokri F. Differential Effects of Inhibitory and Stimulatory Anti-HER2 Monoclonal Antibodies on AKT/ERK Signaling Pathways. Asian Pac J Cancer Prev 2018; 19:2255-2262. [PMID: 30139234 PMCID: PMC6171393 DOI: 10.22034/apjcp.2018.19.8.2255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Objective: Homo- and heterodimerization of the receptor tyrosine kinase HER2 hyperactivate several downstream signaling pathways, leading to uncontrolled growth and proliferation of tumor cells. Anti-HER2 monoclonal antibodies (mAbs) may induce different effects on HER2 dimerization and signaling. Methods: The effect of two inhibitory (2A8, 1T0) and one stimulatory (1H9) anti-HER2 mAbs either alone or in combination with trastuzumab was investigated on AKT and ERK signaling pathways and HER2 degradation in a human breast cancer cell line (BT-474) by Western blotting. Result: While 1H9 mAb had no significant effect on AKT and ERK signaling pathways, 1T0 and 2A8 mAbs inhibited phosphorylation of both pathways. Combination of 1T0 mAb with trastuzumab resulted in significant synergistic inhibition of both pathways and HER2 degradation, much more potently than the combination of trastuzumab and pertuzumab. Conclusion: Our data indicate that anti-HER2 mAbs may induce different signaling pathways depending on their effect on tumor cell growth and proliferation. The significant inhibition of ERK and AKT phosphorylation by 1T0 alone or particularly in combination with trastuzumab suggests its potential therapeutic application for targeted immunotherapy of HER2 overexpressing malignancies.
Collapse
Affiliation(s)
- Tahereh Soltantoyeh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran. and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Wang X, Li X, Yoshiyuki K, Watanabe Y, Sogo Y, Ohno T, Tsuji NM, Ito A. Comprehensive Mechanism Analysis of Mesoporous-Silica-Nanoparticle-Induced Cancer Immunotherapy. Adv Healthc Mater 2016; 5:1169-76. [PMID: 26987867 DOI: 10.1002/adhm.201501013] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/26/2016] [Indexed: 01/01/2023]
Abstract
A plain mesoporous silica nanoparticle without any immunomodulatory molecules significantly enhances anticancer immunity in vivo. Comprehensive mechanism of mesoporous-silica-nanoparticle-induced cancer immunotherapy is analyzed in this paper. The mesoporous silica nanoparticle promotes both Th1 and Th2 immune responses, as it accelerates lymphocytes proliferation, stimulates IFN-γ, IL-2, IL-4, and IL-10 cytokine secretion by lymphocytes ex vivo, and increases IgG, IgG1, IgG2a, IgM, and IgA antibody titers in mice serum compared with those of alum and adjuvant-free groups. Moreover, the mesoporous silica nanoparticle enhances effector memory CD4(+) and CD8(+) T cell populations in three most important immune organs (bone marrow, lymph node, and spleen) of mice compared with those of alum and adjuvant-free groups three months after adjuvant injection. The present study paves the way for the application of mesoporous silica nanoparticle as immunoadjuvant for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiupeng Wang
- Health Research Institute; Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Xia Li
- Health Research Institute; Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Kazuko Yoshiyuki
- Health Research Institute; Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yohei Watanabe
- Biomedical Research Institute; Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yu Sogo
- Health Research Institute; Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Tadao Ohno
- School of Life Dentistry at Tokyo; The Nippon Dental University; Fujimi Chiyoda-ku Tokyo 102-0071 Japan
| | - Noriko M. Tsuji
- Biomedical Research Institute; Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Atsuo Ito
- Health Research Institute; Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| |
Collapse
|
6
|
Becht E, Giraldo NA, Dieu-Nosjean MC, Sautès-Fridman C, Fridman WH. Cancer immune contexture and immunotherapy. Curr Opin Immunol 2016; 39:7-13. [DOI: 10.1016/j.coi.2015.11.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/27/2015] [Accepted: 11/30/2015] [Indexed: 01/01/2023]
|
7
|
Wang X, Li X, Ito A, Watanabe Y, Sogo Y, Tsuji NM, Ohno T. Stimulation of In Vivo Antitumor Immunity with Hollow Mesoporous Silica Nanospheres. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201506179] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Xiupeng Wang
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Xia Li
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Atsuo Ito
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yohei Watanabe
- Biomedical Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yu Sogo
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Noriko M. Tsuji
- Biomedical Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Tadao Ohno
- School of Life Dentistry at Tokyo; The Nippon Dental University; Fujimi Chiyoda-ku Tokyo 102-0071 Japan
| |
Collapse
|
8
|
Wang X, Li X, Ito A, Watanabe Y, Sogo Y, Tsuji NM, Ohno T. Stimulation of In Vivo Antitumor Immunity with Hollow Mesoporous Silica Nanospheres. Angew Chem Int Ed Engl 2015; 55:1899-903. [DOI: 10.1002/anie.201506179] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/25/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Xiupeng Wang
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Xia Li
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Atsuo Ito
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yohei Watanabe
- Biomedical Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yu Sogo
- Health Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Noriko M. Tsuji
- Biomedical Research Institute, Department of Life Science and Biotechnology; National Institute of Advanced Industrial Science and Technology (AIST); Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Tadao Ohno
- School of Life Dentistry at Tokyo; The Nippon Dental University; Fujimi Chiyoda-ku Tokyo 102-0071 Japan
| |
Collapse
|
9
|
Ciliberto D, Staropoli N, Caglioti F, Gualtieri S, Fiorillo L, Chiellino S, De Angelis AM, Mendicino F, Botta C, Caraglia M, Tassone P, Tagliaferri P. A systematic review and meta-analysis of randomized trials on the role of targeted therapy in the management of advanced gastric cancer: Evidence does not translate? Cancer Biol Ther 2015; 16:1148-59. [PMID: 26061272 PMCID: PMC4623405 DOI: 10.1080/15384047.2015.1056415] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/12/2015] [Accepted: 05/24/2015] [Indexed: 12/15/2022] Open
Abstract
It is still uncertain if targeted therapy-based regimens in advanced gastric cancer actually produce survival benefit. To shed light on this important question, we performed a systematic review and meta-analyses on each relevant targeted-pathway. By searching literature databases and proceedings of major cancer meetings in the time-frame 2005-2014, 22 randomized clinical trials exploring targeted therapy for a total of 7022 advanced gastric cancer patients were selected and included in the final analysis. Benefit was demonstrated for antiangiogenic agents in terms of overall survival (HR 0.759; 95%CI 0.655-0.880; p < 0.001). Conversely no benefit was found for EGFR pathway (HR 1.077; 95%CI 0.847-1.370; p = 0.543). Meta-analysis of HER-2 pathway confirmed improvement in terms of survival outcome, already known for this class of drugs (HR 0.823; 95%CI 0.722-0.939; p = 0.004). Pooled analysis demonstrated a significant survival benefit (OS: HR 0.823; PFS: HR 0.762) with acceptable tolerability profile for targeted-based therapies as compared to conventional treatments. This finding conflicts with the outcome of most individual studies, probably due to poor trial design or patients selection. In conclusion, our findings demonstrate a significant survival benefit for targeted therapy in its whole, which can be ascribed to anti-angiogenic and anti-HER2 agents.
Collapse
Key Words
- ADME, absorption, distribution, metabolism, and excretion
- Ab, monoclonal antibody
- BSC, best supportive care
- CHT, chemotherapy
- EGFR, epidermal growth factor receptor
- GC, gastric cancer
- HER2, human epidermal growth factor receptor 2
- HER3, human epidermal growth factor receptor 3
- MET, mesenchymal epithelial transition factor
- NGS, next generation sequencing
- NSCLC, non-small cell lung cancer
- OR, odds-ratio
- OS, overall survival
- PARP, poly ADP ribose polymerase
- PFS, progression free survival
- PI3K, phosphatidylinositide 3-kinases
- PRISMA, preferred reporting items for systematic reviews and meta-analyses
- RAF, rapidly accelerated fibrosarcoma
- RAS, rat sarcoma viral oncogene homolog
- RCTs, randomized clinical trials
- RR, response rate
- TKI, tyrosine kinase inhibitor
- VEGF, vascular endothelial growth factor
- VEGFR: VEGF receptor
- aGC, advanced gastric cancer
- angiogenesis
- gastric cancer
- mTOR, mammalian target of rapamycin
- mTORC, mTOR complex
- meta-analysis
- randomized clinical trials
- systemic chemotherapy
- targeted pathways
- targeted therapy
Collapse
Affiliation(s)
- Domenico Ciliberto
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Nicoletta Staropoli
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Francesca Caglioti
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Simona Gualtieri
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Lucia Fiorillo
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Silvia Chiellino
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Antonina Maria De Angelis
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Francesco Mendicino
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Cirino Botta
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| | - Michele Caraglia
- Department of Biochemistry; Biophysics and General Pathology; Second University of Naples; Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine; Center for Biotechnology; College of Science and Technology; Temple University; Philadelphia, PA USA
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine; Center for Biotechnology; College of Science and Technology; Temple University; Philadelphia, PA USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine; Magna Græcia University; Campus Salvatore Venuta; Catanzaro, Italy
| |
Collapse
|
10
|
Zavala VA, Kalergis AM. New clinical advances in immunotherapy for the treatment of solid tumours. Immunology 2015; 145:182-201. [PMID: 25826229 PMCID: PMC4427384 DOI: 10.1111/imm.12459] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/08/2015] [Accepted: 02/24/2015] [Indexed: 12/16/2022] Open
Abstract
Advances in understanding the mechanisms of cancer cells for evading the immune system surveillance, including how the immune system modulates the phenotype of tumours, have allowed the development of new therapies that benefit from this complex cellular network to specifically target and destroy cancer cells. Immunotherapy researchers have mainly focused on the discovery of tumour antigens that could confer specificity to immune cells to detect and destroy cancer cells, as well as on the mechanisms leading to an improved activation of effector immune cells. The Food and Drug Administration approval in 2010 of ipilumumab for melanoma treatment and of pembrolizumab in 2014, monoclonal antibodies against T-lymphocyte-associated antigen 4 and programmed cell death 1, respectively, are encouraging examples of how research in this area can successfully translate into clinical use with promising results. Currently, several ongoing clinical trials are in progress testing new anti-cancer therapies based on the enhancement of immune cell activity against tumour antigens. Here we discuss the general concepts related to immunotherapy and the recent application to the treatment of cancer with positive results that support their consideration of clinical application to patients.
Collapse
Affiliation(s)
- Valentina A Zavala
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de ChileSantiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
- Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de ChileSantiago, Chile
- INSERM U1064Nantes, France
| |
Collapse
|
11
|
Foekens JA, Martens JWM, Sleijfer S. Are immune signatures a worthwhile tool for decision making in early-stage human epidermal growth factor receptor 2-positive breast cancer? J Clin Oncol 2015; 33:673-5. [PMID: 25605833 DOI: 10.1200/jco.2014.59.5058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- John A Foekens
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John W M Martens
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|